Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
PLoS One ; 18(4): e0284020, 2023.
Article in English | MEDLINE | ID: covidwho-2279364

ABSTRACT

BACKGROUND: Although there have been many studies on antibody responses to SARS-CoV-2 in breast milk, very few have looked at the fate of these in the infant, and whether they are delivered to immunologically relevant sites in infants. METHODS: Mother/infant pairs (mothers who breast milk fed and who were SARS-CoV-2 vaccinated before or after delivery) were recruited for this cross-sectional study. Mother blood, mother breast milk, infant blood, infant nasal specimen, and infant stool was tested for IgA and IgG antibodies against SARS-CoV-2 spike trimer. RESULTS: Thirty-one mother/infant pairs were recruited. Breast milk fed infants acquired systemic anti-spike IgG antibodies only if their mothers were vaccinated antepartum (100% Antepartum; 0% Postpartum; P<0.0001). Breast milk fed infants acquired mucosal anti-spike IgG antibodies (in the nose) only if their mothers were vaccinated antepartum (89% Antepartum; 0% Postpartum; P<0.0001). None of the infants in either group had anti-spike IgA in the blood. Surprisingly, 33% of the infants whose mothers were vaccinated antepartum had high titer anti-spike IgA in the nose (33% Antepartum; 0% Postpartum; P = 0.03). Half-life of maternally transferred plasma IgG antibodies in the Antepartum infant cohort was ~70 days. CONCLUSION: Vaccination antepartum followed by breast milk feeding appears to be the best way to provide systemic and local anti-SARS-CoV-2 antibodies for infants. The presence of high titer SARS-CoV-2-specific IgA in the nose of infants points to the potential importance of breast milk feeding early in life for maternal transfer of mucosal IgA antibodies. Expectant mothers should consider becoming vaccinated antepartum and consider breast milk feeding for optimal transfer of systemic and mucosal antibodies to their infants.


Subject(s)
COVID-19 , Milk, Human , Infant , Female , Humans , Cross-Sectional Studies , COVID-19/prevention & control , SARS-CoV-2 , Breast Feeding , Antibodies, Viral , Immunoglobulin A , Immunoglobulin G
3.
Am J Pathol ; 2023 Mar 22.
Article in English | MEDLINE | ID: covidwho-2274189

ABSTRACT

Ophthalmic manifestations and tissue tropism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been reported in association with coronavirus disease 2019 (COVID-19), but the pathology and cellular localization of SARS-CoV-2 are not well characterized. The objective of this study was to evaluate macroscopic and microscopic changes and investigate cellular localization of SARS-CoV-2 across ocular tissues at autopsy. Ocular tissues were obtained from 25 patients with COVID-19 at autopsy. SARS-CoV-2 nucleocapsid gene RNA was previously quantified by droplet digital PCR from one eye. Herein, contralateral eyes from 21 patients were fixed in formalin and subject to histopathologic examination. Sections of the droplet digital PCR-positive eyes from four other patients were evaluated by in situ hybridization to determine the cellular localization of SARS-CoV-2 spike gene RNA. Histopathologic abnormalities, including cytoid bodies, vascular changes, and retinal edema, with minimal or no inflammation in ocular tissues were observed in all 21 cases evaluated. In situ hybridization localized SARS-CoV-2 RNA to neuronal cells of the retinal inner and outer layers, ganglion cells, corneal epithelia, scleral fibroblasts, and oligodendrocytes of the optic nerve. In conclusion, a range of common histopathologic alterations were identified within ocular tissue, and SARS-CoV-2 RNA was localized to multiple cell types. Further studies will be required to determine whether the alterations observed were caused by SARS-CoV-2 infection, the host immune response, and/or preexisting comorbidities.

4.
American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons ; 23(1):101-107, 2023.
Article in English | EuropePMC | ID: covidwho-2169601

ABSTRACT

Although the risk of SARS-CoV-2 transmission through lung transplantation from acutely infected donors is high, the risks of virus transmission and long-term lung allograft outcomes are not as well described when using pulmonary organs from COVID-19–recovered donors. We describe successful lung transplantation for a COVID-19–related lung injury using lungs from a COVID-19–recovered donor who was retrospectively found to have detectable genomic SARS-CoV-2 RNA in the lung tissue by multiple highly sensitive assays. However, SARS-CoV-2 subgenomic RNA (sgRNA), a marker of viral replication, was not detectable in the donor respiratory tissues. One year after lung transplantation, the recipient has a good functional status, walking 1 mile several times per week without the need for supplemental oxygen and without any evidence of donor-derived SARS-CoV-2 transmission. Our findings highlight the limitations of current clinical laboratory diagnostic assays in detecting the persistence of SARS-CoV-2 RNA in the lung tissue. The persistence of SARS-CoV-2 RNA in the donor tissue did not appear to represent active viral replication via sgRNA testing and, most importantly, did not negatively impact the allograft outcome in the first year after lung transplantation. sgRNA is easily performed and may be a useful assay for assessing viral infectivity in organs from donors with a recent infection.

5.
Nature ; 612(7941): 758-763, 2022 12.
Article in English | MEDLINE | ID: covidwho-2160240

ABSTRACT

Coronavirus disease 2019 (COVID-19) is known to cause multi-organ dysfunction1-3 during acute infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with some patients experiencing prolonged symptoms, termed post-acute sequelae of SARS-CoV-2 (refs. 4,5). However, the burden of infection outside the respiratory tract and time to viral clearance are not well characterized, particularly in the brain3,6-14. Here we carried out complete autopsies on 44 patients who died with COVID-19, with extensive sampling of the central nervous system in 11 of these patients, to map and quantify the distribution, replication and cell-type specificity of SARS-CoV-2 across the human body, including the brain, from acute infection to more than seven months following symptom onset. We show that SARS-CoV-2 is widely distributed, predominantly among patients who died with severe COVID-19, and that virus replication is present in multiple respiratory and non-respiratory tissues, including the brain, early in infection. Further, we detected persistent SARS-CoV-2 RNA in multiple anatomic sites, including throughout the brain, as late as 230 days following symptom onset in one case. Despite extensive distribution of SARS-CoV-2 RNA throughout the body, we observed little evidence of inflammation or direct viral cytopathology outside the respiratory tract. Our data indicate that in some patients SARS-CoV-2 can cause systemic infection and persist in the body for months.


Subject(s)
Autopsy , Brain , COVID-19 , Organ Specificity , SARS-CoV-2 , Humans , Brain/virology , COVID-19/virology , RNA, Viral/analysis , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Virus Replication , Time Factors , Respiratory System/pathology , Respiratory System/virology
6.
J Cardiothorac Vasc Anesth ; 36(6): 1648-1655, 2022 06.
Article in English | MEDLINE | ID: covidwho-1991701

ABSTRACT

OBJECTIVE: To explore if atrial arrhythmias are associated with in-hospital mortality in veno-venous extracorporeal membrane oxygenation (VV-ECMO) patients. DESIGN: Retrospective observational cohort study. SETTING: Quaternary care academic medical center. PARTICIPANTS: Patients with respiratory failure requiring VV-ECMO for >24 hours between January 1, 2016, and January 1, 2019. INTERVENTIONS: None, observational study. MEASUREMENTS AND MAIN RESULTS: Two hundred nineteen VV-ECMO patients were included. Patients were stratified by absence or presence of clinically significant atrial arrhythmias during the VV-ECMO run. Atrial arrhythmias were defined as either atrial fibrillation or atrial flutter that occurred during VV-ECMO and required pharmacologic or electrical intervention. The primary outcome was in-hospital mortality. Secondary outcomes included a composite of thrombotic events, which included ischemic stroke and on-pump arterial thrombosis. Other objectives of this analysis included characterization of atrial arrhythmia incidence, risk factors, and management. A total of 67 patients (30.5%) experienced new-onset atrial arrhythmias post-ECMO cannulation. Age, male sex, and norepinephrine use were independently associated with atrial arrhythmia development. In-hospital mortality was significantly higher in the atrial arrhythmia group (38.8% v 19.1%; p = 0.003). In the multivariate logistic regression analysis, atrial arrhythmias during VV-ECMO were independently associated with increased odds of in-hospital mortality (odds ratio, 2.21; 95% confidence interval, 1.08-4.55; p = 0.03), after controlling for Respiratory Extracorporeal Membrane Oxygenation Survival Prediction score, acute renal failure, total norepinephrine dose, and total cannulation time. CONCLUSIONS: New-onset atrial arrhythmias are a frequent complication during VV-ECMO and are independently associated with excessive in-hospital mortality. Thus, their presence may serve as an important prognostic tool in this patient population.


Subject(s)
Extracorporeal Membrane Oxygenation , Thrombosis , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/therapy , Extracorporeal Membrane Oxygenation/adverse effects , Hospital Mortality , Humans , Male , Norepinephrine , Retrospective Studies , Thrombosis/etiology
7.
ATS Sch ; 3(1): 99-111, 2022 Mar.
Article in English | MEDLINE | ID: covidwho-1929097

ABSTRACT

Background: Recent advances in device technology and image analysis software used to assess the sublingual microcirculation have expanded clinicians' understanding of hemodynamics beyond assessments of blood pressure and end-organ function to provide unique insight into blood flow at the tissue level. Similarly, significant advances in virtual education and telemedicine have transpired recently, especially during the coronavirus disease (COVID-19) pandemic. However, the training of clinicians to acquire microcirculation images continues to rely on in-person instruction, which can be limited by available local expertise and resources, as well as geographic access to instructors. Objective: Our project aimed to test the feasibility of deploying an online curriculum in combination with tele-guidance versus an in-person guided approach to instruct novices to understand basic principle of microcirculatory function and to acquire sublingual microcirculatory images. Methods: After participating in brief didactics, 14 participants were divided into two groups to acquire microcirculatory images on a healthy volunteer. Each participant either 1) obtained images after an in-person demonstration or 2) obtained images with tele-guidance by using FaceTime technology. We recorded individual microcirculation quality scores, necessary time to acquire each image, percentage of correct theoretical questions on assessments, participant satisfaction with the curriculum, and participants' degree of confidence with image acquisition. Results: Participants' image quality scores (14.7 vs. 23.6, P = 0.3) and time to acquire images (191.2 vs. 199.4 s) did not significantly differ. In addition, participants' scores on theoretical knowledge assessments improved over the course of training (19.0% vs. 54.8%, P < 0.05). Conclusion: This feasibility study provides a novel framework for how to successfully deploy asynchronous education and telemedicine to direct novices to acquire sublingual microcirculatory images. Using technological advances to teach microcirculation may enhance wide-scale adoption of a promising clinical monitoring tool for critically ill patients.

8.
EBioMedicine ; 77: 103940, 2022 Mar.
Article in English | MEDLINE | ID: covidwho-1881926

ABSTRACT

BACKGROUND: Limited knowledge exists in post-partum women regarding durability of SARS-CoV-2 vaccine-induced antibody responses and their neutralising ability against SARS-CoV-2 variants of concern (VOC). METHODS: We elucidated longitudinal mRNA vaccination-induced antibody profiles of 13 post-partum and 13 non-post-partum women (control). FINDINGS: The antibody neutralisation titres against SARS-CoV-2 WA-1 strain were comparable between post-partum and non-post-partum women and these levels were sustained up to four months post-second vaccination in both groups. However, neutralisation titers declined against several VOCs, including Beta and Delta. Higher antibody binding was observed against SARS-CoV-2 receptor-binding domain (RBD) mutants with key VOC amino acids when tested with post-second vaccination plasma from post-partum women compared with controls. Importantly, post-vaccination plasma antibody affinity against VOCs RBDs was significantly higher in post-partum women compared with controls. INTERPRETATION: This study demonstrates that there is a differential vaccination-induced immune responses in post-partum women compared with non-post-partum women, which could help inform future vaccination strategies for these groups. FUNDING: The antibody characterisation work described in this manuscript was supported by FDA's Medical Countermeasures Initiative (MCMi) grant #OCET 2021-1565 to S.K and intramural FDA-CBER COVID-19 supplemental funds.


Subject(s)
COVID-19 Vaccines , COVID-19 , Antibodies, Viral , Antibody Affinity , COVID-19/prevention & control , Female , Humans , Immunoglobulin G , Postpartum Period , SARS-CoV-2/genetics , Vaccines, Synthetic , mRNA Vaccines
10.
EBioMedicine ; 77, 2022.
Article in English | EuropePMC | ID: covidwho-1743737

ABSTRACT

Summary Background Limited knowledge exists in post-partum women regarding durability of SARS-CoV-2 vaccine-induced antibody responses and their neutralising ability against SARS-CoV-2 variants of concern (VOC). Methods We elucidated longitudinal mRNA vaccination-induced antibody profiles of 13 post-partum and 13 non-post-partum women (control). Findings The antibody neutralisation titres against SARS-CoV-2 WA-1 strain were comparable between post-partum and non-post-partum women and these levels were sustained up to four months post-second vaccination in both groups. However, neutralisation titers declined against several VOCs, including Beta and Delta. Higher antibody binding was observed against SARS-CoV-2 receptor-binding domain (RBD) mutants with key VOC amino acids when tested with post-second vaccination plasma from post-partum women compared with controls. Importantly, post-vaccination plasma antibody affinity against VOCs RBDs was significantly higher in post-partum women compared with controls. Interpretation This study demonstrates that there is a differential vaccination-induced immune responses in post-partum women compared with non-post-partum women, which could help inform future vaccination strategies for these groups.

11.
Critical Care Medicine ; 50:76-76, 2022.
Article in English | Academic Search Complete | ID: covidwho-1597021

ABSTRACT

In cases with evidence of bronchopneumonia PCR with broad range 16S ribosomal DNA primers was performed followed by sequencing. Secondary infections have been well studied in other viral pneumonias such as influenza, but post-mortem prevalence in fatal COVID-19 cases has not been established. B Introduction: b Coronavirus disease 2019 (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV)-2, has caused a global pandemic. [Extracted from the article] Copyright of Critical Care Medicine is the property of Lippincott Williams & Wilkins and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full . (Copyright applies to all s.)

12.
Front Immunol ; 12: 779026, 2021.
Article in English | MEDLINE | ID: covidwho-1581330

ABSTRACT

A 26-year-old otherwise healthy man died of fulminant myocarditis. Nasopharyngeal specimens collected premortem tested negative for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Histopathological evaluation of the heart showed myocardial necrosis surrounded by cytotoxic T-cells and tissue-repair macrophages. Myocardial T-cell receptor (TCR) sequencing revealed hyper-dominant clones with highly similar sequences to TCRs that are specific for SARS-CoV-2 epitopes. SARS-CoV-2 RNA was detected in the gut, supporting a diagnosis of multisystem inflammatory syndrome in adults (MIS-A). Molecular targets of MIS-associated inflammation are not known. Our data indicate that SARS-CoV-2 antigens selected high-frequency T-cell clones that mediated fatal myocarditis.


Subject(s)
COVID-19/complications , Myocarditis/pathology , Myocarditis/virology , Systemic Inflammatory Response Syndrome/pathology , T-Lymphocytes/immunology , Adult , COVID-19/immunology , COVID-19/pathology , Humans , Male , Myocarditis/immunology , RNA, Viral/analysis , SARS-CoV-2 , Systemic Inflammatory Response Syndrome/immunology
13.
J Intensive Care Med ; 36(10): 1201-1208, 2021 Oct.
Article in English | MEDLINE | ID: covidwho-1305547

ABSTRACT

BACKGROUND: Corticosteroids are part of the treatment guidelines for COVID-19 and have been shown to improve mortality. However, the impact corticosteroids have on the development of secondary infection in COVID-19 is unknown. We sought to define the rate of secondary infection in critically ill patients with COVID-19 and determine the effect of corticosteroid use on mortality in critically ill patients with COVID-19. STUDY DESIGN AND METHODS: One hundred and thirty-five critically ill patients with COVID-19 admitted to the Intensive Care Unit (ICU) at the University of Maryland Medical Center were included in this single-center retrospective analysis. Demographics, symptoms, culture data, use of COVID-19 directed therapies, and outcomes were abstracted from the medical record. The primary outcomes were secondary infection and mortality. Proportional hazards models were used to determine the time to secondary infection and the time to death. RESULTS: The proportion of patients with secondary infection was 63%. The likelihood of developing secondary infection was not significantly impacted by the administration of corticosteroids (HR 1.45, CI 0.75-2.82, P = 0.28). This remained consistent in sub-analysis looking at bloodstream, respiratory, and urine infections. Secondary infection had no significant impact on the likelihood of 28-day mortality (HR 0.66, CI 0.33-1.35, P = 0.256). Corticosteroid administration significantly reduced the likelihood of 28-day mortality (HR 0.27, CI 0.10-0.72, P = 0.01). CONCLUSION: Corticosteroids are an important and lifesaving pharmacotherapeutic option in critically ill patients with COVID-19, which have no impact on the likelihood of developing secondary infections.


Subject(s)
COVID-19 , Coinfection , Adrenal Cortex Hormones , Critical Illness , Humans , Intensive Care Units , Retrospective Studies , SARS-CoV-2
14.
Nat Med ; 27(5): 892-903, 2021 05.
Article in English | MEDLINE | ID: covidwho-1152866

ABSTRACT

Despite signs of infection-including taste loss, dry mouth and mucosal lesions such as ulcerations, enanthema and macules-the involvement of the oral cavity in coronavirus disease 2019 (COVID-19) is poorly understood. To address this, we generated and analyzed two single-cell RNA sequencing datasets of the human minor salivary glands and gingiva (9 samples, 13,824 cells), identifying 50 cell clusters. Using integrated cell normalization and annotation, we classified 34 unique cell subpopulations between glands and gingiva. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral entry factors such as ACE2 and TMPRSS members were broadly enriched in epithelial cells of the glands and oral mucosae. Using orthogonal RNA and protein expression assessments, we confirmed SARS-CoV-2 infection in the glands and mucosae. Saliva from SARS-CoV-2-infected individuals harbored epithelial cells exhibiting ACE2 and TMPRSS expression and sustained SARS-CoV-2 infection. Acellular and cellular salivary fractions from asymptomatic individuals were found to transmit SARS-CoV-2 ex vivo. Matched nasopharyngeal and saliva samples displayed distinct viral shedding dynamics, and salivary viral burden correlated with COVID-19 symptoms, including taste loss. Upon recovery, this asymptomatic cohort exhibited sustained salivary IgG antibodies against SARS-CoV-2. Collectively, these data show that the oral cavity is an important site for SARS-CoV-2 infection and implicate saliva as a potential route of SARS-CoV-2 transmission.


Subject(s)
COVID-19/virology , Mouth/virology , SARS-CoV-2/isolation & purification , Saliva/virology , Angiotensin-Converting Enzyme 2/analysis , Asymptomatic Infections , COVID-19/etiology , Humans , Serine Endopeptidases/analysis , Taste Disorders/etiology , Taste Disorders/virology , Virus Replication
15.
A A Pract ; 14(7): e01236, 2020 May.
Article in English | MEDLINE | ID: covidwho-601387

ABSTRACT

Critically ill patients with coronavirus disease 2019 (COVID-19) have been observed to be hypercoagulable, but the mechanisms for this remain poorly described. Factor VIII is a procoagulant factor that increases during inflammation and is cleaved by activated protein C. To our knowledge, there is only 1 prior study of factor VIII and functional protein C activity in critically ill patients with COVID-19. Here, we present a case series of 10 critically ill patients with COVID-19 who had severe elevations in factor VIII activity and low normal functional protein C activity, which may have contributed to hypercoagulability.


Subject(s)
Coronavirus Infections/blood , Factor VIII/metabolism , Pneumonia, Viral/blood , Protein C/metabolism , Respiratory Distress Syndrome/blood , Thrombophilia/blood , Acute Kidney Injury/epidemiology , Acute Kidney Injury/therapy , Adult , Aged , Aged, 80 and over , Antithrombins/metabolism , Betacoronavirus , C-Reactive Protein/metabolism , COVID-19 , Comorbidity , Coronavirus Infections/epidemiology , Coronavirus Infections/therapy , Critical Illness , Diabetes Mellitus/epidemiology , Dyslipidemias/epidemiology , Extracorporeal Membrane Oxygenation , Female , Ferritins/metabolism , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/metabolism , Humans , Hypertension/epidemiology , International Normalized Ratio , Male , Middle Aged , Obesity/epidemiology , Pandemics , Partial Thromboplastin Time , Pneumonia, Viral/epidemiology , Pneumonia, Viral/therapy , Prothrombin Time , Renal Dialysis , Renal Insufficiency, Chronic/epidemiology , Respiration, Artificial , Respiratory Distress Syndrome/epidemiology , Respiratory Distress Syndrome/therapy , SARS-CoV-2
SELECTION OF CITATIONS
SEARCH DETAIL