Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
2.
J Infect Dis ; 226(11): 1897-1902, 2022 Nov 28.
Article in English | MEDLINE | ID: covidwho-2135321

ABSTRACT

BACKGROUND: The consequences of past coronavirus disease 2019 (COVID-19) infection for personal and population health are emerging, but accurately identifying distant infection is a challenge. Anti-spike antibodies rise after both vaccination and infection and anti-nucleocapsid antibodies rapidly decline. METHODS: We evaluated anti-membrane antibodies in COVID-19 naive, vaccinated, and convalescent subjects to determine if they persist and accurately detect distant infection. RESULTS: We found that anti-membrane antibodies persist for at least 1 year and are a sensitive and specific marker of past COVID-19 infection. CONCLUSIONS: Thus, anti-membrane and anti-spike antibodies together can differentiate between COVID-19 convalescent, vaccinated, and naive states to advance public health and research.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , Vaccination , Public Health , Virion , Antibodies, Viral , Spike Glycoprotein, Coronavirus
3.
Med (N Y) ; 2022 Sep 19.
Article in English | MEDLINE | ID: covidwho-2031561

ABSTRACT

BACKGROUND: Universities are vulnerable to infectious disease outbreaks, making them ideal environments to study transmission dynamics and evaluate mitigation and surveillance measures. Here, we analyze multimodal COVID-19-associated data collected during the 2020-2021 academic year at Colorado Mesa University and introduce a SARS-CoV-2 surveillance and response framework. METHODS: We analyzed epidemiological and sociobehavioral data (demographics, contact tracing, and WiFi-based co-location data) alongside pathogen surveillance data (wastewater and diagnostic testing, and viral genomic sequencing of wastewater and clinical specimens) to characterize outbreak dynamics and inform policy. We applied relative risk, multiple linear regression, and social network assortativity to identify attributes or behaviors associated with contracting SARS-CoV-2. To characterize SARS-CoV-2 transmission, we used viral sequencing, phylogenomic tools, and functional assays. FINDINGS: Athletes, particularly those on high-contact teams, had the highest risk of testing positive. On average, individuals who tested positive had more contacts and longer interaction durations than individuals who never tested positive. The distribution of contacts per individual was overdispersed, although not as overdispersed as the distribution of phylogenomic descendants. Corroboration via technical replicates was essential for identification of wastewater mutations. CONCLUSIONS: Based on our findings, we formulate a framework that combines tools into an integrated disease surveillance program that can be implemented in other congregate settings with limited resources. FUNDING: This work was supported by the National Science Foundation, the Hertz Foundation, the National Institutes of Health, the Centers for Disease Control and Prevention, the Massachusetts Consortium on Pathogen Readiness, the Howard Hughes Medical Institute, the Flu Lab, and the Audacious Project.

4.
Sci Transl Med ; 14(657): eabm4908, 2022 08 10.
Article in English | MEDLINE | ID: covidwho-1846321

ABSTRACT

The SARS-CoV-2 B.1.621 (Mu) variant emerged in January 2021 and was categorized as a variant of interest by the World Health Organization in August 2021. This designation prompted us to study the sensitivity of this variant to antibody neutralization. In a live virus neutralization assay with serum samples from individuals vaccinated with the Pfizer/BioNTech or Moderna mRNA vaccines, we measured neutralization antibody titers against B.1.621, an early isolate (spike 614D), and a variant of concern (B.1.351, Beta variant). We observed reduced neutralizing antibody titers against the B.1.621 variant (3.4- to 7-fold reduction, depending on the serum sample and time after the second vaccination) compared to the early isolate and a similar reduction when compared to B.1.351. Likewise, convalescent serum from hamsters previously infected with an early isolate neutralized B.1.621 to a lower degree. Despite this antibody titer reduction, hamsters could not be efficiently rechallenged with the B.1.621 variant, suggesting that the immune response to the first infection is adequate to provide protection against a subsequent infection with the B.1.621 variant.


Subject(s)
COVID-19 , Viral Envelope Proteins , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/therapy , Humans , Immunization, Passive , Membrane Glycoproteins/genetics , Neutralization Tests , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/genetics , Vaccination , Viral Envelope Proteins/genetics , COVID-19 Serotherapy
5.
Nat Med ; 28(5): 1083-1094, 2022 05.
Article in English | MEDLINE | ID: covidwho-1671607

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has demonstrated a clear need for high-throughput, multiplexed and sensitive assays for detecting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other respiratory viruses and their emerging variants. Here, we present a cost-effective virus and variant detection platform, called microfluidic Combinatorial Arrayed Reactions for Multiplexed Evaluation of Nucleic acids (mCARMEN), which combines CRISPR-based diagnostics and microfluidics with a streamlined workflow for clinical use. We developed the mCARMEN respiratory virus panel to test for up to 21 viruses, including SARS-CoV-2, other coronaviruses and both influenza strains, and demonstrated its diagnostic-grade performance on 525 patient specimens in an academic setting and 166 specimens in a clinical setting. We further developed an mCARMEN panel to enable the identification of 6 SARS-CoV-2 variant lineages, including Delta and Omicron, and evaluated it on 2,088 patient specimens with near-perfect concordance to sequencing-based variant classification. Lastly, we implemented a combined Cas13 and Cas12 approach that enables quantitative measurement of SARS-CoV-2 and influenza A viral copies in samples. The mCARMEN platform enables high-throughput surveillance of multiple viruses and variants simultaneously, enabling rapid detection of SARS-CoV-2 variants.


Subject(s)
COVID-19 , Influenza, Human , COVID-19/diagnosis , Humans , Microfluidics , SARS-CoV-2/genetics
6.
Cell ; 185(3): 485-492.e10, 2022 02 03.
Article in English | MEDLINE | ID: covidwho-1588148

ABSTRACT

An outbreak of over 1,000 COVID-19 cases in Provincetown, Massachusetts (MA), in July 2021-the first large outbreak mostly in vaccinated individuals in the US-prompted a comprehensive public health response, motivating changes to national masking recommendations and raising questions about infection and transmission among vaccinated individuals. To address these questions, we combined viral genomic and epidemiological data from 467 individuals, including 40% of outbreak-associated cases. The Delta variant accounted for 99% of cases in this dataset; it was introduced from at least 40 sources, but 83% of cases derived from a single source, likely through transmission across multiple settings over a short time rather than a single event. Genomic and epidemiological data supported multiple transmissions of Delta from and between fully vaccinated individuals. However, despite its magnitude, the outbreak had limited onward impact in MA and the US overall, likely due to high vaccination rates and a robust public health response.


Subject(s)
COVID-19/epidemiology , COVID-19/immunology , COVID-19/transmission , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19/virology , Child , Child, Preschool , Contact Tracing/methods , Disease Outbreaks , Female , Genome, Viral , Humans , Infant , Infant, Newborn , Male , Massachusetts/epidemiology , Middle Aged , Molecular Epidemiology , Phylogeny , SARS-CoV-2/classification , Vaccination , Whole Genome Sequencing , Young Adult
7.
Emerg Infect Dis ; 27(11): 2776-2785, 2021 11.
Article in English | MEDLINE | ID: covidwho-1444021

ABSTRACT

University settings have demonstrated potential for coronavirus disease (COVID-19) outbreaks; they combine congregate living, substantial social activity, and a young population predisposed to mild illness. Using genomic and epidemiologic data, we describe a COVID-19 outbreak at the University of Wisconsin-Madison, Madison, Wisconsin, USA. During August-October 2020, a total of 3,485 students, including 856/6,162 students living in dormitories, tested positive. Case counts began rising during move-in week, August 25-31, 2020, then rose rapidly during September 1-11, 2020. The university initiated multiple prevention efforts, including quarantining 2 dormitories; a subsequent decline in cases was observed. Genomic surveillance of cases from Dane County, in which the university is located, did not find evidence of transmission from a large cluster of cases in the 2 quarantined dorms during the outbreak. Coordinated implementation of prevention measures can reduce COVID-19 spread in university settings and may limit spillover to the surrounding community.


Subject(s)
COVID-19 , Universities , Disease Outbreaks , Humans , SARS-CoV-2 , Wisconsin/epidemiology
8.
Clin Infect Dis ; 73(6): e1329-e1336, 2021 09 15.
Article in English | MEDLINE | ID: covidwho-1411883

ABSTRACT

BACKGROUND: Healthcare personnel (HCP) are at increased risk of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We posit that current infection control guidelines generally protect HCP from SARS-CoV-2 infection in a healthcare setting. METHODS: In this retrospective case series, we used viral genomics to investigate the likely source of SARS-CoV-2 infection in HCP at a major academic medical institution in the Upper Midwest of the United States between 25 March and 27 December 2020. We obtained limited epidemiological data through informal interviews and review of the electronic health record and combined this information with healthcare-associated viral sequences and viral sequences collected in the broader community to infer the most likely source of infection in HCP. RESULTS: We investigated SARS-CoV-2 infection clusters involving 95 HCP and 137 possible patient contact sequences. The majority of HCP infections could not be linked to a patient or coworker (55 of 95 [57.9%]) and were genetically similar to viruses circulating concurrently in the community. We found that 10.5% of HCP infections (10 of 95) could be traced to a coworker. Strikingly, only 4.2% (4 of 95) could be traced to a patient source. CONCLUSIONS: Infections among HCP add further strain to the healthcare system and put patients, HCP, and communities at risk. We found no evidence for healthcare-associated transmission in the majority of HCP infections evaluated. Although we cannot rule out the possibility of cryptic healthcare-associated transmission, it appears that HCP most commonly become infected with SARS-CoV-2 via community exposure. This emphasizes the ongoing importance of mask wearing, physical distancing, robust testing programs, and rapid distribution of vaccines.


Subject(s)
COVID-19 , SARS-CoV-2 , Delivery of Health Care , Health Personnel , Humans , Retrospective Studies , United States/epidemiology
9.
Dissertation Abstracts International: Section B: The Sciences and Engineering ; 82(12-B):No Pagination Specified, 2021.
Article in English | APA PsycInfo | ID: covidwho-1411301

ABSTRACT

The rapid worldwide spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) took the world by storm in early 2020, amassing over 153 million con-firmed cases as of May 5, 2021. Whole-genome sequencing of confirmed SARS-CoV-2 cases has been used to infer patterns of viral transmission, identify putative introductions of the virus, and provide surveillance on established transmission chains. Aside from tracking the spread of SARS-CoV-2, sequencing data can provide insights into the efficacy of mitigation efforts and forecast the future trajectory of local outbreaks. In this dissertation I focus on exploring the factors that influenced SARS-CoV-2 introduction and spread in southern Wisconsin. Chapter 1 reviews the current state of the pandemic and takes a deeper look into what is known about SARS-CoV-2 replication, the factors that influence SARS-CoV-2 spread, and the preventative measure that can be implemented to slow the spread of SARS-CoV-2.In chapter 2, I describe our work characterizing the initial introduction and spread of SARS-CoV-2 in Dane and Milwaukee counties. These counties are the two most populous counties in southern Wisconsin, separated by only ~100km. Dane County had the 12th confirmed SARS-CoV-2 in the United States, but this did not lead to de-scendant cryptic spread, posited as the cause of many local outbreaks early in the pandemic. Instead, the Dane County outbreak was seeded by multiple later introductions, followed by limited community spread. In contrast, relatively few introductions in Milwaukee County led to extensive community spread. We characterized the efficacy of Wisconsin's "Safer at Home" order and found that viral spread decreased by at least 40% in both Dane and Milwaukee counties. This suggested that, at least early in the pandemic, patterns of SARS-CoV-2 transmission varied substantially even in nearby communities.In chapter 3, I describe our work tracing patterns of SARS-CoV-2 spread in the context of hospitals and clinics. Despite the use of personal protective equipment (PPE) and other strategies to decrease risk of infection, front-line healthcare workers (HCWs) are at increased risk for infection with SARS-CoV-2 compared to the general population. Using a computational contact tracing tool, informal interviews, and genomic sequencing of HCWs and patient contacts, we evaluate the efficacy of current PPE protocols and other preventative measures put in place at a major academic biomedical institution in the American Upper Midwest. We found that the majority of HCWs evaluated, had viral sequences that could be traced to the outside community, suggesting that PPE protocols and preventative measures in place were effective at suppressing viral spread within the institution evaluated here.In chapter 4, I describe our work characterizing an outbreak of SARS-CoV-2 in two residence halls on the University of Wisconsin-Madison's campus. Universities represent high-risk settings for COVID-19 outbreaks, as they combine congregate living situations with high levels of social activity, and a young population that is unlikely to experience severe disease. The University of Wisconsin-Madison had an outbreak of SARS-CoV-2 during Fall 2020 resulting in which 3,485 students tested positive during August-October 2020. This rapid uptick in cases led the university to quarantine two residence halls to contain further SARS-CoV-2 spread. We combine genomic sequencing from these two residence halls, and with 5% of community sequences from Dane County to reveal limited downstream transmission from the residence hall viruses. Our results suggest that containment strategies including large-scale quarantines in congregate living situations (such as in dorms) and suspension of on-campus activities may be effective when experiencing large-scale outbreaks, when implemented rapidly and effectively. (PsycInfo Database Record (c) 2021 APA, all rights reserved)

10.
PLoS Pathog ; 17(8): e1009849, 2021 08.
Article in English | MEDLINE | ID: covidwho-1369568

ABSTRACT

The emergence of divergent SARS-CoV-2 lineages has raised concern that novel variants eliciting immune escape or the ability to displace circulating lineages could emerge within individual hosts. Though growing evidence suggests that novel variants arise during prolonged infections, most infections are acute. Understanding how efficiently variants emerge and transmit among acutely-infected hosts is therefore critical for predicting the pace of long-term SARS-CoV-2 evolution. To characterize how within-host diversity is generated and propagated, we combine extensive laboratory and bioinformatic controls with metrics of within- and between-host diversity to 133 SARS-CoV-2 genomes from acutely-infected individuals. We find that within-host diversity is low and transmission bottlenecks are narrow, with very few viruses founding most infections. Within-host variants are rarely transmitted, even among individuals within the same household, and are rarely detected along phylogenetically linked infections in the broader community. These findings suggest that most variation generated within-host is lost during transmission.


Subject(s)
COVID-19/virology , Genetic Variation , SARS-CoV-2/genetics , Acute Disease , COVID-19/transmission , Evolution, Molecular , Genome, Viral , Humans , Phylogeny , SARS-CoV-2/pathogenicity , Time Factors
11.
Clin Infect Dis ; 73(Suppl 1): S45-S53, 2021 07 15.
Article in English | MEDLINE | ID: covidwho-1315688

ABSTRACT

BACKGROUND: High-frequency, rapid-turnaround severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) testing continues to be proposed as a way of efficiently identifying and mitigating transmission in congregate settings. However, 2 SARS-CoV-2 outbreaks occurred among intercollegiate university athletic programs during the fall 2020 semester, despite mandatory directly observed daily antigen testing. METHODS: During the fall 2020 semester, athletes and staff in both programs were tested daily using Quidel's Sofia SARS Antigen Fluorescent Immunoassay, with positive antigen results requiring confirmatory testing with real-time reverse-transcription polymerase chain reaction. We used genomic sequencing to investigate transmission dynamics in these 2 outbreaks. RESULTS: In the first outbreak, 32 confirmed cases occurred within a university athletics program after the index patient attended a meeting while infectious, despite a negative antigen test on the day of the meeting. Among isolates sequenced from that outbreak, 24 (92%) of 26 were closely related, suggesting sustained transmission following an initial introduction event. In the second outbreak, 12 confirmed cases occurred among athletes from 2 university programs that faced each other in an athletic competition, despite receipt of negative antigen test results on the day of the competition. Sequences from both teams were closely related and distinct from viruses circulating in the community for team 1, suggesting transmission during intercollegiate competition in the community for team 2. CONCLUSIONS: These findings suggest that antigen testing alone, even when mandated and directly observed, may not be sufficient as an intervention to prevent SARS-CoV-2 outbreaks in congregate settings, and they highlight the importance of vaccination to prevent SARS-CoV-2 outbreak in congregate settings.


Subject(s)
COVID-19 , Sports , Humans , Immunologic Tests , SARS-CoV-2 , Universities
12.
PLoS Pathog ; 17(2): e1009373, 2021 02.
Article in English | MEDLINE | ID: covidwho-1105836

ABSTRACT

The evolutionary mechanisms by which SARS-CoV-2 viruses adapt to mammalian hosts and, potentially, undergo antigenic evolution depend on the ways genetic variation is generated and selected within and between individual hosts. Using domestic cats as a model, we show that SARS-CoV-2 consensus sequences remain largely unchanged over time within hosts, while dynamic sub-consensus diversity reveals processes of genetic drift and weak purifying selection. We further identify a notable variant at amino acid position 655 in Spike (H655Y), which was previously shown to confer escape from human monoclonal antibodies. This variant arises rapidly and persists at intermediate frequencies in index cats. It also becomes fixed following transmission in two of three pairs. These dynamics suggest this site may be under positive selection in this system and illustrate how a variant can quickly arise and become fixed in parallel across multiple transmission pairs. Transmission of SARS-CoV-2 in cats involved a narrow bottleneck, with new infections founded by fewer than ten viruses. In RNA virus evolution, stochastic processes like narrow transmission bottlenecks and genetic drift typically act to constrain the overall pace of adaptive evolution. Our data suggest that here, positive selection in index cats followed by a narrow transmission bottleneck may have instead accelerated the fixation of S H655Y, a potentially beneficial SARS-CoV-2 variant. Overall, our study suggests species- and context-specific adaptations are likely to continue to emerge. This underscores the importance of continued genomic surveillance for new SARS-CoV-2 variants as well as heightened scrutiny for signatures of SARS-CoV-2 positive selection in humans and mammalian model systems.


Subject(s)
COVID-19/veterinary , Cat Diseases/virology , SARS-CoV-2/physiology , Adaptation, Biological , Animals , Biological Evolution , COVID-19/transmission , COVID-19/virology , Cats , Evolution, Molecular , Genetic Variation , Humans , Phylogeny , Selection, Genetic
13.
Nat Commun ; 11(1): 5558, 2020 11 03.
Article in English | MEDLINE | ID: covidwho-910229

ABSTRACT

Evidence-based public health approaches that minimize the introduction and spread of new SARS-CoV-2 transmission clusters are urgently needed in the United States and other countries struggling with expanding epidemics. Here we analyze 247 full-genome SARS-CoV-2 sequences from two nearby communities in Wisconsin, USA, and find surprisingly distinct patterns of viral spread. Dane County had the 12th known introduction of SARS-CoV-2 in the United States, but this did not lead to descendant community spread. Instead, the Dane County outbreak was seeded by multiple later introductions, followed by limited community spread. In contrast, relatively few introductions in Milwaukee County led to extensive community spread. We present evidence for reduced viral spread in both counties following the statewide "Safer at Home" order, which went into effect 25 March 2020. Our results suggest patterns of SARS-CoV-2 transmission may vary substantially even in nearby communities. Understanding these local patterns will enable better targeting of public health interventions.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Genome, Viral/genetics , Pneumonia, Viral/epidemiology , Pneumonia, Viral/transmission , COVID-19 , Coronavirus Infections/prevention & control , Geography , Humans , Mass Screening/methods , Molecular Epidemiology/methods , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Psychological Distance , Respiratory Protective Devices , SARS-CoV-2 , United States/epidemiology , Wisconsin/epidemiology
14.
Emerg Infect Dis ; 26(10): 2489-2491, 2020 Oct.
Article in English | MEDLINE | ID: covidwho-696601

ABSTRACT

Whether a healthcare worker's severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is community or hospital acquired affects prevention practices. We used virus sequencing to determine that infection of a healthcare worker who cared for 2 SARS-CoV-2-infected patients was probably community acquired. Appropriate personal protective equipment may have protected against hospital-acquired infection.


Subject(s)
Betacoronavirus , Community-Acquired Infections/virology , Coronavirus Infections/transmission , Infectious Disease Transmission, Patient-to-Professional/prevention & control , Occupational Diseases/prevention & control , Pneumonia, Viral/transmission , RNA, Viral/analysis , COVID-19 , Coronavirus Infections/virology , Health Personnel , Humans , Pandemics , Personal Protective Equipment , Pneumonia, Viral/virology , SARS-CoV-2 , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL