Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
- IMPACC group; Al Ozonoff; Joanna Schaenman; Naresh Doni Jayavelu; Carly E. Milliren; Carolyn S. Calfee; Charles B. Cairns; Monica Kraft; Lindsey R. Baden; Albert C. Shaw; Florian Krammer; Harm Van Bakel; Denise Esserman; Shanshan Liu; Ana Fernandez Sesma; Viviana Simon; David A. Hafler; Ruth R. Montgomery; Steven H. Kleinstein; Ofer Levy; Christian Bime; Elias K. Haddad; David J. Erle; Bali Pulendran; Kari C. Nadeau; Mark M. Davis; Catherine L. Hough; William B. Messer; Nelson I. Agudelo Higuita; Jordan P. Metcalf; Mark A. Atkinson; Scott C. Brakenridge; David B. Corry; Farrah Kheradmand; Lauren I. R. Ehrlich; Esther Melamed; Grace A. McComsey; Rafick Sekaly; Joann Diray-Arce; Bjoern Peters; Alison D. Augustine; Elaine F. Reed; Kerry McEnaney; Brenda Barton; Claudia Lentucci; Mehmet Saluvan; Ana C. Chang; Annmarie Hoch; Marisa Albert; Tanzia Shaheen; Alvin Kho; Sanya Thomas; Jing Chen; Maimouna D. Murphy; Mitchell Cooney; Scott Presnell; Leying Guan; Jeremy Gygi; Shrikant Pawar; Anderson Brito; Zain Khalil; James A. Overton; Randi Vita; Kerstin Westendorf; Cole Maguire; Slim Fourati; Ramin Salehi-Rad; Aleksandra Leligdowicz; Michael Matthay; Jonathan Singer; Kirsten N. Kangelaris; Carolyn M. Hendrickson; Matthew F. Krummel; Charles R. Langelier; Prescott G. Woodruff; Debra L. Powell; James N. Kim; Brent Simmons; I.Michael Goonewardene; Cecilia M. Smith; Mark Martens; Jarrod Mosier; Hiroki Kimura; Amy Sherman; Stephen Walsh; Nicolas Issa; Charles Dela Cruz; Shelli Farhadian; Akiko Iwasaki; Albert I. Ko; Evan J. Anderson; Aneesh Mehta; Jonathan E. Sevransky; Sharon Chinthrajah; Neera Ahuja; Angela Rogers; Maja Artandi; Sarah A.R. Siegel; Zhengchun Lu; Douglas A. Drevets; Brent R. Brown; Matthew L. Anderson; Faheem W. Guirgis; Rama V. Thyagarajan; Justin Rousseau; Dennis Wylie; Johanna Busch; Saurin Gandhi; Todd A. Triplett; George Yendewa; Olivia Giddings; Tatyana Vaysman; Bernard Khor; Adeeb Rahman; Daniel Stadlbauer; Jayeeta Dutta; Hui Xie; Seunghee Kim-Schulze; Ana Silvia Gonzalez-Reiche; Adriana van de Guchte; Holden T. Maecker; Keith Farrugia; Zenab Khan; Joanna Schaenman; Elaine F. Reed; Ramin Salehi-Rad; David Elashoff; Jenny Brook; Estefania Ramires-Sanchez; Megan Llamas; Adreanne Rivera; Claudia Perdomo; Dawn C. Ward; Clara E. Magyar; Jennifer Fulcher; Yumiko Abe-Jones; Saurabh Asthana; Alexander Beagle; Sharvari Bhide; Sidney A. Carrillo; Suzanna Chak; Rajani Ghale; Ana Gonzales; Alejandra Jauregui; Norman Jones; Tasha Lea; Deanna Lee; Raphael Lota; Jeff Milush; Viet Nguyen; Logan Pierce; Priya Prasad; Arjun Rao; Bushra Samad; Cole Shaw; Austin Sigman; Pratik Sinha; Alyssa Ward; Andrew - Willmore; Jenny Zhan; Sadeed Rashid; Nicklaus Rodriguez; Kevin Tang; Luz Torres Altamirano; Legna Betancourt; Cindy Curiel; Nicole Sutter; Maria Tercero Paz; Gayelan Tietje-Ulrich; Carolyn Leroux; Jennifer Connors; Mariana Bernui; Michele Kutzler; Carolyn Edwards; Edward Lee; Edward Lin; Brett Croen; Nicholas Semenza; Brandon Rogowski; Nataliya Melnyk; Kyra Woloszczuk; Gina Cusimano; Matthew Bell; Sara Furukawa; Renee McLin; Pamela Marrero; Julie Sheidy; George P. Tegos; Crystal Nagle; Nathan Mege; Kristen Ulring; Vicki Seyfert-Margolis; Michelle Conway; Dave Francisco; Allyson Molzahn; Heidi Erickson; Connie Cathleen Wilson; Ron Schunk; Trina Hughes; Bianca Sierra; Kinga K. Smolen; Michael Desjardins; Simon van Haren; Xhoi Mitre; Jessica Cauley; Xiofang Li; Alexandra Tong; Bethany Evans; Christina Montesano; Jose Humberto Licona; Jonathan Krauss; Jun Bai Park Chang; Natalie Izaguirre; Omkar Chaudhary; Andreas Coppi; John Fournier; Subhasis Mohanty; M. Catherine Muenker; Allison Nelson; Khadir Raddassi; Michael Rainone; William Ruff; Syim Salahuddin; Wade L. Schulz; Pavithra Vijayakumar; Haowei Wang; Elsio Wunder Jr.; H. Patrick Young; Yujiao Zhao; Miti Saksena; Deena Altman; Erna Kojic; Komal Srivastava; Lily Q. Eaker; Maria Carolina Bermudez; Katherine F. Beach; Levy A. Sominsky; Arman Azad; Juan Manuel Carreno; Gagandeep Singh; Ariel Raskin; Johnstone Tcheou; Dominika Bielak; Hisaaki Kawabata; Lubbertus CF Mulder; Giulio Kleiner; Laurel Bristow; Laila Hussaini; Kieffer Hellmeister; Hady Samaha; Andrew Cheng; Christine Spainhour; Erin M. Scherer; Brandi Johnson; Amer Bechnak; Caroline R. Ciric; Lauren Hewitt; Bernadine Panganiban; Chistopher Huerta; Jacob Usher; Erin Carter; Nina Mcnair; Susan Pereira Ribeiro; Alexandra S. Lee; Evan Do; Andrea Fernandes; Monali Manohar; Thomas Hagan; Catherine Blish; Hena Naz Din; Jonasel Roque; Samuel S. Yang; Amanda E. Brunton; Peter E. Sullivan; Matthew Strnad; Zoe L. Lyski; Felicity J. Coulter; John L. Booth; Lauren A. Sinko; Lyle Moldawer; Brittany Borrensen; Brittney Roth-Manning; Li-Zhen Song; Ebony Nelson; Megan Lewis-Smith; Jacob Smith; Pablo Guaman Tipan; Nadia Siles; Sam Bazzi; Janelle Geltman; Kerin Hurley; Giovanni Gabriele; Scott Sieg; Matthew C. Altman; Patrice M. Becker; Nadine Rouphael.
Preprint in English | medRxiv | ID: ppmedrxiv-22273396

ABSTRACT

BackgroundBetter understanding of the association between characteristics of patients hospitalized with coronavirus disease 2019 (COVID-19) and outcome is needed to further improve upon patient management. MethodsImmunophenotyping Assessment in a COVID-19 Cohort (IMPACC) is a prospective, observational study of 1,164 patients from 20 hospitals across the United States. Disease severity was assessed using a 7-point ordinal scale based on degree of respiratory illness. Patients were prospectively surveyed for 1 year after discharge for post-acute sequalae of COVID-19 (PASC) through quarterly surveys. Demographics, comorbidities, radiographic findings, clinical laboratory values, SARS-CoV-2 PCR and serology were captured over a 28-day period. Multivariable logistic regression was performed. FindingsThe median age was 59 years (interquartile range [IQR] 20); 711 (61%) were men; overall mortality was 14%, and 228 (20%) required invasive mechanical ventilation. Unsupervised clustering of ordinal score over time revealed distinct disease course trajectories. Risk factors associated with prolonged hospitalization or death by day 28 included age [≥] 65 years (odds ratio [OR], 2.01; 95% CI 1.28-3.17), Hispanic ethnicity (OR, 1.71; 95% CI 1.13-2.57), elevated baseline creatinine (OR 2.80; 95% CI 1.63-4.80) or troponin (OR 1.89; 95% 1.03-3.47), baseline lymphopenia (OR 2.19; 95% CI 1.61-2.97), presence of infiltrate by chest imaging (OR 3.16; 95% CI 1.96-5.10), and high SARS-CoV2 viral load (OR 1.53; 95% CI 1.17-2.00). Fatal cases had the lowest ratio of SARS-CoV-2 antibody to viral load levels compared to other trajectories over time (p=0.001). 589 survivors (51%) completed at least one survey at follow-up with 305 (52%) having at least one symptom consistent with PASC, most commonly dyspnea (56% among symptomatic patients). Female sex was the only associated risk factor for PASC. InterpretationIntegration of PCR cycle threshold, and antibody values with demographics, comorbidities, and laboratory/radiographic findings identified risk factors for 28-day outcome severity, though only female sex was associated with PASC. Longitudinal clinical phenotyping offers important insights, and provides a framework for immunophenotyping for acute and long COVID-19. FundingNIH RESEARCH IN CONTEXTO_ST_ABSEvidence before this studyC_ST_ABSWe did a systematic search of the PubMed database from January 1st, 2020 until April 24th, 2022 using the search terms: "hospitalized" AND "SARS-CoV-2" OR "COVID-19" AND "Pro-spective" AND "Antibody" OR "PCR" OR "long term follow up" and applying the following filters: "Multicenter Study" AND "Observational Study". No language restrictions were applied. While clinical, laboratory, and radiographic features associated with severe COVID-19 in hospitalized adults have been described, description of the kinetics of SARS-CoV-2 specific assays available to clinicians (e.g. PCR and binding antibody) and their integration with other variables is scarce for both short and long term follow up. The current literature is comprised of several studies with small sample size, cross-sectional design with laboratory data typically only recorded at a single point in time (e.g., on admission), limited clinical characteristics, variable duration of follow up, single-center setting, retrospective analyses, kinetics of either PCR or antibody testing but not both, and outcomes such as death or, mechanical ventilation that do not allow delineation of variations in clinical course. Added value of this studyIn our large longitudinal multicenter cohort, the description of outcome severity, was not limited to survival versus death, but encompassed a clinical trajectory approach leveraging longitudinal data based on time in hospital, disease severity by ordinal scale based on degree of respiratory illness, and presence or absence of limitations at discharge. Fatal COVID-19 cases had the lowest ratio of antibody to viral load levels over time as compared to non-fatal cases. Integration of PCR cycle threshold and antibody values with demographics, baseline comorbidities, and laboratory/radiographic findings identified additional risk factors for outcome severity over the first 28 days. However, female sex was the only variable associated with persistence of symptoms over time. Persistence of symptoms was not associated with clinical trajectory over the first 28 days, nor with antibody/viral loads from the acute phase. Implications of all the available evidenceThe described calculated ratio (binding IgG/PCR Ct value) is unique compared to other studies, reflecting host pathogen interactions and representing an accessible approach for patient risk stratification. Integration of SARS-CoV-2 viral load and binding antibody kinetics with other laboratory as well as clinical characteristics in hospitalized COVID-19 patients can identify patients likely to have the most severe short-term outcomes, but is not predictive of symptom persistence at one year post-discharge.

2.
Preprint in English | bioRxiv | ID: ppbiorxiv-471539

ABSTRACT

The impact of coronavirus disease 2019 (COVID-19) mRNA vaccination on pregnancy and fertility has become a major topic of public interest. We investigated two of the most widely propagated claims to determine 1) whether COVID-19 mRNA vaccination of mice during early pregnancy is associated with an increased incidence of birth defects or growth abnormalities, and 2) whether COVID-19 mRNA-vaccinated human volunteers exhibit elevated levels of antibodies to the human placental protein syncytin-1. Using a mouse model, we found that intramuscular COVID-19 mRNA vaccination during early pregnancy at gestational age E7.5 did not lead to differences in fetal size by crown-rump length or weight at term, nor did we observe any gross birth defects. In contrast, injection of the TLR3 agonist and double-stranded RNA mimic polyinosinic-polycytidylic acid, or poly(I:C), impacted growth in utero leading to reduced fetal size. No overt maternal illness following either vaccination or poly(I:C) exposure was observed. We also found that term fetuses from vaccinated murine pregnancies exhibit high circulating levels of anti-Spike and anti-RBD antibodies to SARS-CoV-2 consistent with maternal antibody status, indicating transplacental transfer. Finally, we did not detect increased levels of circulating anti-syncytin-1 antibodies in a cohort of COVID-19 vaccinated adults compared to unvaccinated adults by ELISA. Our findings contradict popular claims associating COVID-19 mRNA vaccination with infertility and adverse neonatal outcomes.

3.
Preprint in English | medRxiv | ID: ppmedrxiv-21253992

ABSTRACT

Prior to the emergence of antigenically distinct SARS-CoV-2 variants, reinfections were reported infrequently - presumably due to the generation of durable and protective immune responses. However, case reports also suggested that rare, repeated infections may occur as soon as 48 days following initial disease onset. The underlying immunologic deficiencies enabling SARS-CoV-2 reinfections are currently unknown. Here we describe a renal transplant recipient who developed recurrent, symptomatic SARS-CoV-2 infection - confirmed by whole virus genome sequencing - 7 months after primary infection. To elucidate the immunological mechanisms responsible for SARS-CoV-2 reinfection, we performed longitudinal profiling of cellular and humoral responses during both primary and recurrent SARS-CoV-2 infection. We found that the patient responded to the primary infection with transient, poor-quality adaptive immune responses. The patients immune system was further compromised by intervening treatment for acute rejection of the renal allograft prior to reinfection. Importantly, we also identified the development of neutralizing antibodies and the formation of humoral memory responses prior to SARS-CoV-2 reinfection. However, these neutralizing antibodies failed to confer protection against reinfection, suggesting that additional factors are required for efficient prevention of SARS-CoV-2 reinfection. Further, we found no evidence supporting viral evasion of primary adaptive immune responses, suggesting that susceptibility to reinfection may be determined by host factors rather than pathogen adaptation in this patient. In summary, our study suggests that a low neutralizing antibody presence alone is not sufficient to confer resistance against reinfection. Thus, patients with solid organ transplantation, or patients who are otherwise immunosuppressed, who recover from infection with SARS-CoV-2 may not develop sufficient protective immunity and are at risk of reinfection.

4.
Preprint in English | medRxiv | ID: ppmedrxiv-21250946

ABSTRACT

There is an urgent need to expand testing for SARS-CoV-2 and other respiratory pathogens as the global community struggles to control the COVID-19 pandemic. Current diagnostic methods can be affected by supply chain bottlenecks and require the assistance of medical professionals, impeding the implementation of large-scale testing. Self-collection of saliva may solve these problems, as it can be completed without specialized training and uses generic materials. In this study, we observed thirty individuals who self-collected saliva using four different collection devices and analyzed their feedback. Two of these devices, a funnel and bulb pipette, were used to evaluate at-home saliva collection by 60 individuals. All devices enabled the safe, unsupervised self-collection of saliva. The quantity and quality of the samples received were acceptable for SARS-CoV-2 diagnostic testing, as determined by RNase P detection. Here, we demonstrate inexpensive, generic, buffer free collection devices suitable for unsupervised and home saliva self-collection.

5.
Preprint in English | medRxiv | ID: ppmedrxiv-21249236

ABSTRACT

While several clinical and immunological parameters correlate with disease severity and mortality in SARS-CoV-2 infection, work remains in identifying unifying correlates of coronavirus disease 2019 (COVID-19) that can be used to guide clinical practice. Here, we examine saliva and nasopharyngeal (NP) viral load over time and correlate them with patient demographics, and cellular and immune profiling. We found that saliva viral load was significantly higher in those with COVID-19 risk factors; that it correlated with increasing levels of disease severity and showed a superior ability over nasopharyngeal viral load as a predictor of mortality over time (AUC=0.90). A comprehensive analysis of immune factors and cell subsets revealed strong predictors of high and low saliva viral load, which were associated with increased disease severity or better overall outcomes, respectively. Saliva viral load was positively associated with many known COVID-19 inflammatory markers such as IL-6, IL-18, IL-10, and CXCL10, as well as type 1 immune response cytokines. Higher saliva viral loads strongly correlated with the progressive depletion of platelets, lymphocytes, and effector T cell subsets including circulating follicular CD4 T cells (cTfh). Anti-spike (S) and anti-receptor binding domain (RBD) IgG levels were negatively correlated with saliva viral load showing a strong temporal association that could help distinguish severity and mortality in COVID-19. Finally, patients with fatal COVID-19 exhibited higher viral loads, which correlated with the depletion of cTfh cells, and lower production of anti-RBD and anti-S IgG levels. Together these results demonstrated that viral load - as measured by saliva but not nasopharyngeal -- is a dynamic unifying correlate of disease presentation, severity, and mortality over time.

6.
Preprint in English | medRxiv | ID: ppmedrxiv-20248331

ABSTRACT

Recent studies have provided insights into innate and adaptive immune dynamics in coronavirus disease 2019 (COVID-19). Yet, the exact feature of antibody responses that governs COVID-19 disease outcomes remain unclear. Here, we analysed humoral immune responses in 209 asymptomatic, mild, moderate and severe COVID-19 patients over time to probe the nature of antibody responses in disease severity and mortality. We observed a correlation between anti-Spike (S) IgG levels, length of hospitalization and clinical parameters associated with worse clinical progression. While high anti-S IgG levels correlated with worse disease severity, such correlation was time-dependent. Deceased patients did not have higher overall humoral response than live discharged patients. However, they mounted a robust, yet delayed response, measured by anti-S, anti-RBD IgG, and neutralizing antibody (NAb) levels, compared to survivors. Delayed seroconversion kinetics correlated with impaired viral control in deceased patients. Finally, while sera from 89% of patients displayed some neutralization capacity during their disease course, NAb generation prior to 14 days of disease onset emerged as a key factor for recovery. These data indicate that COVID-19 mortality does not correlate with the cross-sectional antiviral antibody levels per se, but rather with the delayed kinetics of NAb production.

7.
Preprint in English | bioRxiv | ID: ppbiorxiv-383661

ABSTRACT

1The biomedical community is producing increasingly high dimensional datasets, integrated from hundreds of patient samples, which current computational techniques struggle to explore. To uncover biological meaning from these complex datasets, we present an approach called Multiscale PHATE, which learns abstracted biological features from data that can be directly predictive of disease. Built on a continuous coarse graining process called diffusion condensation, Multiscale PHATE creates a tree of data granularities that can be cut at coarse levels for high level summarizations of data, as well as at fine levels for detailed representations on subsets. We apply Multiscale PHATE to study the immune response to COVID-19 in 54 million cells from 168 hospitalized patients. Through our analysis of patient samples, we identify CD16hi CD66blo neutrophil and IFN{gamma}+GranzymeB+ Th17 cell responses enriched in patients who die. Further, we show that population groupings Multiscale PHATE discovers can be directly fed into a classifier to predict disease outcome. We also use Multiscale PHATE-derived features to construct two different manifolds of patients, one from abstracted flow cytometry features and another directly on patient clinical features, both associating immune subsets and clinical markers with outcome.

8.
Preprint in English | medRxiv | ID: ppmedrxiv-20175299

ABSTRACT

As COVID-19 continues to spread across the globe, the need for inexpensive, large-scale prevalence surveillance testing increases. We present a method for testing newborn dried blood spots (DBS) for anti-SARS-COV-2 IgG antibodies, and demonstrate its applicability as an easily accessible proxy for measuring maternal seroprevalence.

9.
Preprint in English | medRxiv | ID: ppmedrxiv-20167791

ABSTRACT

Current bottlenecks for improving accessibility and scalability of SARS-CoV-2 testing include diagnostic assay costs, complexity, and supply chain shortages. To resolve these issues, we developed SalivaDirect, which received Emergency Use Authorization (EUA) from the U.S. Food and Drug Administration on August 15th, 2020. The critical component of our approach is to use saliva instead of respiratory swabs, which enables non-invasive frequent sampling and reduces the need for trained healthcare professionals during collection. Furthermore, we simplified our diagnostic test by (1) not requiring nucleic acid preservatives at sample collection, (2) replacing nucleic acid extraction with a simple proteinase K and heat treatment step, and (3) testing specimens with a dualplex quantitative reverse transcription PCR (RT-qPCR) assay. We validated SalivaDirect with reagents and instruments from multiple vendors to minimize the risk for supply chain issues. Regardless of our tested combination of reagents and instruments from different vendors, we found that SalivaDirect is highly sensitive with a limit of detection of 6-12 SARS-CoV-2 copies/L. When comparing SalivaDirect to paired nasopharyngeal swabs using the authorized ThermoFisher Scientific TaqPath COVID-19 combo kit, we found high agreement in testing outcomes (>94%). In partnership with the National Basketball Association (NBA) and Players Association, we conducted a large-scale (n = 3,779) SalivaDirect usability study and comparison to standard nasal/oral tests for asymptomatic and presymptomatic SARS-CoV-2 detection. From this cohort of healthy NBA players, staff, and contractors, we found that 99.7% of samples were valid using our saliva collection techniques and a 89.5% positive and >99.9% negative test agreement to swabs, demonstrating that saliva is a valid and noninvasive alternative to swabs for large-scale SARS-CoV-2 testing. SalivaDirect is a flexible and inexpensive ($1.21-$4.39/sample in reagent costs) option to help improve SARS-CoV-2 testing capacity. Register to become a designated laboratory to use SalivaDirect under our FDA EUA on our website: publichealth.yale.edu/salivadirect/.

10.
Preprint in English | medRxiv | ID: ppmedrxiv-20149849

ABSTRACT

Introductory ParagraphThe COVID-19 pandemic has affected more than 10 million people worldwide with mortality exceeding half a million patients. Risk factors associated with severe disease and mortality include advanced age, hypertension, diabetes, and obesity.1 Clear mechanistic understanding of how these comorbidities converge to enable severe infection is lacking. Notably each of these risk factors pathologically disrupts the lipidome and this disruption may be a unifying feature of severe COVID-19.1-7 Here we provide the first in depth interrogation of lipidomic changes, including structural-lipids as well as the eicosanoids and docosanoids lipid mediators (LMs), that mark COVID-19 disease severity. Our data reveal that progression from moderate to severe disease is marked by a loss of specific immune regulatory LMs and increased pro-inflammatory species. Given the important immune regulatory role of LMs, these data provide mechanistic insight into the immune balance in COVID-19 and potential targets for therapy with currently approved pharmaceuticals.8

11.
Preprint in English | bioRxiv | ID: ppbiorxiv-169946

ABSTRACT

Although COVID-19 is considered to be primarily a respiratory disease, SARS-CoV-2 affects multiple organ systems including the central nervous system (CNS). Yet, there is no consensus whether the virus can infect the brain, or what the consequences of CNS infection are. Here, we used three independent approaches to probe the capacity of SARS-CoV-2 to infect the brain. First, using human brain organoids, we observed clear evidence of infection with accompanying metabolic changes in the infected and neighboring neurons. However, no evidence for the type I interferon responses was detected. We demonstrate that neuronal infection can be prevented either by blocking ACE2 with antibodies or by administering cerebrospinal fluid from a COVID-19 patient. Second, using mice overexpressing human ACE2, we demonstrate in vivo that SARS-CoV-2 neuroinvasion, but not respiratory infection, is associated with mortality. Finally, in brain autopsy from patients who died of COVID-19, we detect SARS-CoV-2 in the cortical neurons, and note pathologic features associated with infection with minimal immune cell infiltrates. These results provide evidence for the neuroinvasive capacity of SARS-CoV2, and an unexpected consequence of direct infection of neurons by SARS-CoV-2.

12.
Preprint in English | bioRxiv | ID: ppbiorxiv-155887

ABSTRACT

The current RT-qPCR assay recommended for SARS-CoV-2 testing in the United States requires analysis of three genomic targets per sample: two viral and one host. To simplify testing and reduce the volume of required reagents, we developed a multiplex RT-qPCR assay to detect SARS-CoV-2 in a single reaction. We used existing N1, N2, and RP primer and probe sets by the CDC, but substituted fluorophores to allow multiplexing of the assay. The cycle threshold (Ct) values of our multiplex RT-qPCR were comparable to those obtained by the singleplex assay adapted for research purposes. Low copies (>500 copies / reaction) of SARS-CoV-2 RNA were consistently detected by the multiplex RT-qPCR. Our novel multiplex RT-qPCR improves upon current singleplex diagnostics by saving reagents, costs, time and labor.

13.
Preprint in English | medRxiv | ID: ppmedrxiv-20138289

ABSTRACT

Recent studies have provided insights into the pathogenesis of coronavirus disease 2019 (COVID-19)1-4. Yet, longitudinal immunological correlates of disease outcome remain unclear. Here, we serially analysed immune responses in 113 COVID-19 patients with moderate (non-ICU) and severe (ICU) disease. Immune profiling revealed an overall increase in innate cell lineages with a concomitant reduction in T cell number. We identify an association between early, elevated cytokines and worse disease outcomes. Following an early increase in cytokines, COVID-19 patients with moderate disease displayed a progressive reduction in type-1 (antiviral) and type-3 (antifungal) responses. In contrast, patients with severe disease maintained these elevated responses throughout the course of disease. Moreover, severe disease was accompanied by an increase in multiple type 2 (anti-helminths) effectors including, IL-5, IL-13, IgE and eosinophils. Unsupervised clustering analysis of plasma and peripheral blood leukocyte data identified 4 immune signatures, representing (A) growth factors, (B) type-2/3 cytokines, (C) mixed type-1/2/3 cytokines, and (D) chemokines that correlated with three distinct disease trajectories of patients. The immune profile of patients who recovered with moderate disease was enriched in tissue reparative growth factor signature (A), while the profile for those with worsened disease trajectory had elevated levels of all four signatures. Thus, we identified development of a maladapted immune response profile associated with severe COVID-19 outcome and early immune signatures that correlate with divergent disease trajectories.

14.
Preprint in English | medRxiv | ID: ppmedrxiv-20123414

ABSTRACT

A growing body of evidence indicates sex differences in the clinical outcomes of coronavirus disease 2019 (COVID-19)1-4. However, whether immune responses against SARS-CoV-2 differ between sexes, and whether such differences explain male susceptibility to COVID-19, is currently unknown. In this study, we examined sex differences in viral loads, SARS-CoV-2-specific antibody titers, plasma cytokines, as well as blood cell phenotyping in COVID-19 patients. By focusing our analysis on patients with mild to moderate disease who had not received immunomodulatory medications, our results revealed that male patients had higher plasma levels of innate immune cytokines and chemokines including IL-8, IL-18, and CCL5, along with more robust induction of non-classical monocytes. In contrast, female patients mounted significantly more robust T cell activation than male patients during SARS-CoV-2 infection, which was sustained in old age. Importantly, we found that a poor T cell response negatively correlated with patients age and was predictive of worse disease outcome in male patients, but not in female patients. Conversely, higher innate immune cytokines in female patients associated with worse disease progression, but not in male patients. These findings reveal a possible explanation underlying observed sex biases in COVID-19, and provide important basis for the development of sex-based approach to the treatment and care of men and women with COVID-19.

15.
Preprint in English | medRxiv | ID: ppmedrxiv-20083907

ABSTRACT

BackgroundThe effects of Covid-19 in pregnancy remain relatively unknown. We present a case of second trimester pregnancy with symptomatic Covid-19 complicated by severe preeclampsia and placental abruption. MethodsWe analyzed placenta for the presence of SARS-CoV-2 through molecular and immunohistochemical assays and by and electron microscopy, and we measured the maternal antibody response in blood to this infection. ResultsSARS-CoV-2 localized predominantly to syncytiotrophoblast cells at the maternal-fetal interface of the placenta. Histological examination of the placenta revealed a dense macrophage infiltrate, but no evidence for vasculopathy typically associated with preeclampsia. ConclusionThis case demonstrates, for the first time, SARS-CoV-2 invasion of the placenta, highlighting the potential for severe morbidity among pregnant women with Covid-19.

16.
Preprint in English | medRxiv | ID: ppmedrxiv-20067835

ABSTRACT

Rapid and accurate SARS-CoV-2 diagnostic testing is essential for controlling the ongoing COVID-19 pandemic. The current gold standard for COVID-19 diagnosis is real-time RT-PCR detection of SARS-CoV-2 from nasopharyngeal swabs. Low sensitivity, exposure risks to healthcare workers, and global shortages of swabs and personal protective equipment, however, necessitate the validation of new diagnostic approaches. Saliva is a promising candidate for SARS-CoV-2 diagnostics because (1) collection is minimally invasive and can reliably be self-administered and (2) saliva has exhibited comparable sensitivity to nasopharyngeal swabs in detection of other respiratory pathogens, including endemic human coronaviruses, in previous studies. To validate the use of saliva for SARS-CoV-2 detection, we tested nasopharyngeal and saliva samples from confirmed COVID-19 patients and self-collected samples from healthcare workers on COVID-19 wards. When we compared SARS-CoV-2 detection from patient-matched nasopharyngeal and saliva samples, we found that saliva yielded greater detection sensitivity and consistency throughout the course of infection. Furthermore, we report less variability in self-sample collection of saliva. Taken together, our findings demonstrate that saliva is a viable and more sensitive alternative to nasopharyngeal swabs and could enable at-home self-administered sample collection for accurate large-scale SARS-CoV-2 testing.

17.
Preprint in English | medRxiv | ID: ppmedrxiv-20061168

ABSTRACT

BackgroundAs we contend with the massive SARS-CoV-2 pandemic, preventing infections among healthcare workers (HCWs) and patients is critical for delivering care to patients admitted for other purposes, and many standard scheduling practices require reassessment. In most academic hospitals in the United States, inpatient rotations are designed to deliver optimal patient care by staggering rotations of attendings and house-staff, and much emphasis is placed on HCW burnout, yet during a pandemic preventing further infection is the single most important factor. Our purpose was to model various inpatient rotation schedules of physicians and nurses to determine patterns associated with optimal workforce preservation and lower nosocomial infections in settings in which personal protective equipment is imperfect or unavailable. Summary of MethodsWe simulated the spread of COVID-19 in hospital wards using Monte Carlo methods. Universal model parameters for COVID-19 included incubation period distribution and latent period distribution. Situation-dependent COVID-19 model parameters included pre-admission infection probability, team member infection probability, physician-to-patient, nurse-to-patient, patient-to-physician, patient-to-nurse, and HCW-to-HCW transmission probabilities, team member absence after symptom onset, daily SARS-CoV-2 exposure probability of team members (e.g. via exposure to other staff), length of admission after COVID-19 symptoms, and length of simulation time. Model parameters that varied by hospital setting and service type included average patient load per team, average patient hospitalization, and number of physicians and nurses on a team and on duty. ResultsThe primary outcome measure was probability of team failure, defined as the likelihood that at some point there are insufficient attendings, house-staff or nurses to staff a fully functioning floor. In all of our simulations, physician and nurse rotation lengths of 1-3 days led to higher team failure rates. Nursing shifts of 12 versus 8 hours and avoiding staggering of physician rotations also decreased the chance of team failure. ConclusionsSimple changes in staff scheduling, such as lengthening nursing shifts or avoiding rotations that are either staggered or last fewer than three days, can result in improved workforce preservation. These workforce scheduling changes are easy to implement.

18.
Preprint in English | medRxiv | ID: ppmedrxiv-20048108

ABSTRACT

The recent spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exemplifies the critical need for accurate and rapid diagnostic assays to prompt clinical and public health interventions. Currently, several quantitative reverse-transcription polymerase chain reaction (qRT-PCR) assays are being used by clinical, research, and public health laboratories. However, it is currently unclear if results from different tests are comparable. Our goal was to evaluate the primer-probe sets used in four common diagnostic assays available on the World Health Organization (WHO) website. To facilitate this effort, we generated RNA transcripts to be used as assay standards and distributed them to other laboratories for internal validation. We then used (1) RNA transcript standards, (2) full-length SARS-CoV-2 RNA, (3) pre-COVID-19 nasopharyngeal swabs, and (4) clinical samples from COVID-19 patients to determine analytical efficiency and sensitivity of the qRT-PCR primer-probe sets. We show that all primer-probe sets can be used to detect SARS-CoV-2 at 500 virus copies per reaction, except for the RdRp-SARSr (Charite) confirmatory primer-probe set which has low sensitivity. Our findings characterize the limitations of currently used primer-probe sets and can assist other laboratories in selecting appropriate assays for the detection of SARS-CoV-2.

SELECTION OF CITATIONS
SEARCH DETAIL