Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Critical Care Medicine ; 51(1 Supplement):558, 2023.
Article in English | EMBASE | ID: covidwho-2190674

ABSTRACT

INTRODUCTION: Coronavirus disease 2019 (Covid-19) represents viral pneumonia from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In non-COVID ARDS patients, high driving pressure (DP = Plateau pressure - PEEP) has been associated with higher mortality. Pathophysiological features of COVID ARDS have been considered different from non-COVID ARDS, because of relatively preserved compliance of the respiratory system despite marked hypoxemia. The objective of this study is to evaluate the effect of DP on mortality in intubated COVID-19 ARDS patients. METHOD(S): All data were retrospectively collected from EHR of COVID-19 patients admitted to our ICU in2020. COVID ARDS patients in our institution are managed based on ARDS management guidelines that include lung protective ventilation strategy, PEEP protocol, and prone positioning for persistent PaO2/FiO2 (P/F) ratio < 150. Average P/F, Plateau pressure (PP), DP, and SOFA scores were collected and calculated on first three days of mechanical ventilation and correlated with mortality. RESULT(S): A total of 46 patients intubated with COVID-19 ARDS were included, with 25 survivors and 21 nonsurvivors (ICU mortality rate 46%). Compared to Non-survivor group, the survivor group had similar age (60 +/-12 vs 66 +/-12, p = 0.1), similar P/F ratios(D1: 147 +/-96 vs 136 +/-98, p = 0.7;D3: 136 +/-88 vs 128 +/-74, p = 0.7), similar PP (D1: 23 +/-7 vs 25 +/-6, p = 0.2;D2: 24 +/-6 vs 26 +/-7, p = 0.2;D3: 28 +/-7 vs 29 +/-7, p = 0.7), less number of comorbidities (1.7 +/-1.6 vs 3.2 +/-2.8, p = 0.03), better SOFA score change (SOFA D3 minus D1: 0.4 +/-0.5 vs 1.7 +/-1.7, p = 0.0006), and significantly less DP (D1: 11 +/-5 vs 15 +/-4, p = 0.006;D2: 12 +/-3 vs 15 +/-6, p = 0.01;D3: 14 +/-5 vs 19 +/-8, p = 0.02). CONCLUSION(S): Similar to other forms of ARDS, low driving pressure (less than 15 cmH2O) in COVID-19 ARDS is associated with lower mortality. These findings should be investigated in large multicenter prospective studies.

2.
Critical Care Medicine ; 51(1 Supplement):464, 2023.
Article in English | EMBASE | ID: covidwho-2190639

ABSTRACT

INTRODUCTION: Coronavirus disease 2019 (Covid-19) represents viral pneumonia from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In ARDS patients, positive fluid balance has been associated with prolonged mechanical ventilation, longer length of stay, and higher mortality. As a result, restrictive fluid strategies improved oxygenation and reduced duration of mechanical ventilation. Optimal fluid management strategy for invasively ventilated COVID-19 patients is lacking. The objective of this study is to evaluate the effect of fluid balance on need for proning and successful liberation of invasive mechanical ventilation (MV) in patients with COVID-19 ARDS. METHOD(S): All data were retrospectively collected from EHR of COVID-19 patients admitted to our ICU. COVID ARDS patients in our institution are managed based on ARDS management guidelines that include lung protective ventilation strategy, PEEP protocol, and prone positioning for persistent PaO2/FiO2 (P/F) ratio < 150. Fluid balance in ml was calculated on Day 1 (F1) and Day 7 (F7) of ICU admission. Groups were divided into those successfully liberated (L-group) and those unable to be liberated from MV (U-group). RESULT(S): A total of 57 patients intubated with COVID-19 ARDS were included, with 29 in the L-group and 28 in the U-group. Compared to U-group, L-group had similar age (64 +/- 13 vs 64 +/- 13, p = 1.0), number of comorbidities (2.3 +/- 2.2 vs 2.7 +/- 2.5, p = 0.5), P/F ratio on day 1 (D1, 144 +/- 110 vs 130 +/- 95, p = 0.6), D1 SOFA score (6.4 +/- 4.5 vs 5.9 +/- 4.3, p = 0.7), and F1 (434 +/- 1106 vs 413 +/- 1301, p = 0.9). F7 was significantly less for L-group than U-group (290 +/- 2500 vs 2000 +/- 4000, p = 0.05). [F7 - F1] was also significantly less for L-group compared to U-group (- 144 +/- 1400 vs 1600 +/- 2800, p = 0.004). There was less need for proning (38 % vs 72 %, p = 0.01), and lower mortality (24% vs 86 %, p < 0.001) in the L-group compared to the U-group. CONCLUSION(S): In a cohort of invasively ventilated patients with COVID-19 ARDS, a lower cumulative fluid balance was associated with less need for proning and more successful liberation of MV, indicating that restricted fluid management in these patients may be beneficial. These findings should be investigated in large multicenter prospective studies.

3.
mBio ; : e0230822, 2022 Oct 31.
Article in English | MEDLINE | ID: covidwho-2097925

ABSTRACT

Coronavirus disease 2019 (COVID-19) is frequently associated with neurological deficits, but how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces these effects remains unclear. Here, we show that astrocytes are readily infected by SARS-CoV-2, but surprisingly, neuropilin-1, not angiotensin-converting enzyme 2 (ACE2), serves as the principal receptor mediating cell entry. Infection is further positively modulated by the two-pore segment channel 2 (TPC2) protein that regulates membrane trafficking and endocytosis. Astrocyte infection produces a pathological response closely resembling reactive astrogliosis characterized by elevated type I interferon (IFN) production, increased inflammation, and the decreased expression of transporters of water, ions, choline, and neurotransmitters. These combined events initiated within astrocytes produce a hostile microenvironment that promotes the dysfunction and death of uninfected bystander neurons. IMPORTANCE SARS-CoV-2 infection primarily targets the lung but may also damage other organs, including the brain, heart, kidney, and intestine. Central nervous system (CNS) pathologies include loss of smell and taste, headache, delirium, acute psychosis, seizures, and stroke. Pathological loss of gray matter occurs in SARS-CoV-2 infection, but it is unclear whether this is due to direct viral infection, indirect effects associated with systemic inflammation, or both. Here, we used induced pluripotent stem cell (iPSC)-derived brain organoids and primary human astrocytes from the cerebral cortex to study direct SARS-CoV-2 infection. Our findings support a model where SARS-CoV-2 infection of astrocytes produces a panoply of changes in the expression of genes regulating innate immune signaling and inflammatory responses. The deregulation of these genes in astrocytes produces a microenvironment within the CNS that ultimately disrupts normal neuron function, promoting neuronal cell death and CNS deficits.

4.
Afr Health Sci ; 22(Spec Issue): 1-10, 2022 Aug.
Article in English | MEDLINE | ID: covidwho-2044107

ABSTRACT

The Infectious Diseases Institute (IDI), established in 2001, was the first autonomous institution of Makerere University set up as an example of what self-governing institutes can do in transforming the academic environment to become a rapidly progressive University addressing the needs of society This paper describes the success factors and lessons learned in development of sustainable centers of excellence to prepare academic institutions to respond appropriately to current and future challenges to global health. Key success factors included a) strong collaboration by local and international experts to combat the HIV pandemic, along with b) seed funding from Pfizer Inc., c) longstanding collaboration with Accordia Global Health Foundation to create and sustain institutional strengthening programs, d) development of a critical mass of multi-disciplinary research leaders and managers of the center, and e) a series of strong directors who built strong governance structures to execute the vision of the institute, with subsequent transition to local leadership. Conclusion: Twenty years of sustained investment in infrastructure, human capital, leadership, and collaborations present Makerere University and the sub-Saharan Africa region with an agile center of excellence with preparedness to meet the current and future challenges to global health.


Subject(s)
Capacity Building , Communicable Diseases , Humans , Universities , International Cooperation , Delivery of Health Care
5.
Sci Rep ; 12(1): 15451, 2022 09 14.
Article in English | MEDLINE | ID: covidwho-2028721

ABSTRACT

The spread of SARS-CoV-2 has led to a devastating pandemic, with infections resulting in a range of symptoms collectively known as COVID-19. The full repertoire of human tissues and organs susceptible to infection is an area of active investigation, and some studies have implicated the reproductive system. The effects of COVID-19 on human reproduction remain poorly understood, and particularly the impact on early embryogenesis and establishment of a pregnancy are not known. In this work, we explore the susceptibility of early human embryos to SARS-CoV-2 infection. By using RNA-seq and immunofluorescence, we note that ACE2 and TMPRSS2, two canonical cell entry factors for SARS-CoV-2, are co-expressed in cells of the trophectoderm in blastocyst-stage preimplantation embryos. For the purpose of viral entry studies, we used fluorescent reporter virions pseudotyped with Spike (S) glycoprotein from SARS-CoV-2, and we observe robust infection of trophectoderm cells. This permissiveness could be attenuated with blocking antibodies targeting S or ACE2. When exposing human blastocysts to the live, fully infectious SARS-CoV-2, we detected cases of infection that compromised embryo health. Therefore, we identify a new human target tissue for SARS-CoV-2 with potential medical implications for reproductive health during the COVID-19 pandemic and its aftermath.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Humans , Pandemics , Peptidyl-Dipeptidase A , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/genetics
6.
Nat Commun ; 13(1): 4422, 2022 07 30.
Article in English | MEDLINE | ID: covidwho-1967602

ABSTRACT

Studies are needed to evaluate the safety and effectiveness of mRNA SARS-CoV-2 vaccination during pregnancy, and the levels of protection provided to their newborns through placental transfer of antibodies. Here, we evaluate the transplacental transfer of mRNA vaccine products and functional anti-SARS-CoV-2 antibodies during pregnancy and early infancy in a cohort of 20 individuals vaccinated during late pregnancy. We find no evidence of mRNA vaccine products in maternal blood, placenta tissue, or cord blood at delivery. However, we find time-dependent efficient transfer of IgG and neutralizing antibodies to the neonate that persists during early infancy. Additionally, using phage immunoprecipitation sequencing, we find a vaccine-specific signature of SARS-CoV-2 Spike protein epitope binding that is transplacentally transferred during pregnancy. Timing of vaccination during pregnancy is critical to ensure transplacental transfer of protective antibodies during early infancy.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , Female , Humans , Immunoglobulin G , Infant, Newborn , Placenta , Pregnancy , RNA, Messenger , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus , Vaccines, Synthetic , mRNA Vaccines
7.
Nature ; 607(7918): 351-355, 2022 07.
Article in English | MEDLINE | ID: covidwho-1852428

ABSTRACT

SARS-CoV-2 Delta and Omicron are globally relevant variants of concern. Although individuals infected with Delta are at risk of developing severe lung disease, infection with Omicron often causes milder symptoms, especially in vaccinated individuals1,2. The question arises of whether widespread Omicron infections could lead to future cross-variant protection, accelerating the end of the pandemic. Here we show that without vaccination, infection with Omicron induces a limited humoral immune response in mice and humans. Sera from mice overexpressing the human ACE2 receptor and infected with Omicron neutralize only Omicron, but not other variants of concern, whereas broader cross-variant neutralization was observed after WA1 and Delta infections. Unlike WA1 and Delta, Omicron replicates to low levels in the lungs and brains of infected animals, leading to mild disease with reduced expression of pro-inflammatory cytokines and diminished activation of lung-resident T cells. Sera from individuals who were unvaccinated and infected with Omicron show the same limited neutralization of only Omicron itself. By contrast, Omicron breakthrough infections induce overall higher neutralization titres against all variants of concern. Our results demonstrate that Omicron infection enhances pre-existing immunity elicited by vaccines but, on its own, may not confer broad protection against non-Omicron variants in unvaccinated individuals.


Subject(s)
COVID-19 , Cross Protection , SARS-CoV-2 , Vaccination , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , Cross Protection/immunology , Cytokines , Humans , Mice , SARS-CoV-2/classification , SARS-CoV-2/immunology , Vaccination/statistics & numerical data
8.
JCI Insight ; 7(12)2022 06 22.
Article in English | MEDLINE | ID: covidwho-1846629

ABSTRACT

Pregnancy confers unique immune responses to infection and vaccination across gestation. To date, there are limited data comparing vaccine- and infection-induced neutralizing Abs (nAbs) against COVID-19 variants in mothers during pregnancy. We analyzed paired maternal and cord plasma samples from 60 pregnant individuals. Thirty women vaccinated with mRNA vaccines (from December 2020 through August 2021) were matched with 30 naturally infected women (from March 2020 through January 2021) by gestational age of exposure. Neutralization activity against the 5 SARS-CoV-2 spike sequences was measured by a SARS-CoV-2-pseudotyped spike virion assay. Effective nAbs against SARS-CoV-2 were present in maternal and cord plasma after both infection and vaccination. Compared with WT spike protein, these nAbs were less effective against the Delta and Mu spike variants. Vaccination during the third trimester induced higher cord-nAb levels at delivery than did infection during the third trimester. In contrast, vaccine-induced nAb levels were lower at the time of delivery compared with infection during the first trimester. The transfer ratio (cord nAb level divided by maternal nAb level) was greatest in mothers vaccinated in the second trimester. SARS-CoV-2 vaccination or infection in pregnancy elicits effective nAbs with differing neutralization kinetics that are influenced by gestational time of exposure.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , Female , Gestational Age , Humans , Mothers , Neutralization Tests , Vaccination
10.
Studia Universitatis Babes-Bolyai Musica ; 66(2):29-38, 2021.
Article in English | Web of Science | ID: covidwho-1638629

ABSTRACT

This case study presents the process of music therapy in a clinical setting, under the professional supervision of Dr. Catherine Warner, a music therapist with over 25 years of experience in this field. The music therapy sessions were conducted with a client displaying early-stage dementia symptoms. Over twelve weeks, with an eight-week hiatus due to the Covid-19 Pandemic lockdown, the client benefited from music therapy sessions which helped her navigate through the challenges of the disease, as well as providing a safe environment to express feelings and emotions. This case study presents the main themes of the work, such as combining poetry with singing, and listening to soothing music. This paper also illustrates the challenges of the sessions, especially in relation to memory loss and anxiety. Lastly, the therapist's reflections are presented to complete the overview of the work.

11.
medrxiv; 2021.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2021.12.09.21267557

ABSTRACT

Pregnancy confers unique immune responses to infection and vaccination across gestation. To date, there is limited data comparing vaccine versus infection-induced nAb to COVID-19 variants in mothers during pregnancy. We analyzed paired maternal and cord plasma samples from 60 pregnant individuals. Thirty women vaccinated with mRNA vaccines were matched with 30 naturally infected women by gestational age of exposure. Neutralization activity against the five SARS-CoV-2 Spike sequences was measured by a SARS-CoV-2 pseudotyped Spike virion assay. Effective nAbs against SARS-CoV-2 were present in maternal and cord plasma after both infection and vaccination. Compared to wild type or Alpha variant Spike, these nAbs were less effective against the Kappa, Delta, and Mu Spike variants. Vaccination during the third trimester induced higher nAb levels at delivery than infection during the third trimester. In contrast, vaccine-induced nAb levels were lower at the time of delivery compared to infection during the first trimester. The transfer ratio (cord nAb level/maternal nAb level) was greatest in mothers vaccinated in the second trimester. SARS-CoV-2 vaccination or infection in pregnancy elicit effective nAbs with differing neutralization kinetics that is impacted by gestational time of exposure. Vaccine induced neutralizing activity was reduced against the Delta, Mu, and Kappa variants.


Subject(s)
COVID-19 , Severe Acute Respiratory Syndrome
12.
Elife ; 102021 10 12.
Article in English | MEDLINE | ID: covidwho-1464010

ABSTRACT

While mRNA vaccines are proving highly efficacious against SARS-CoV-2, it is important to determine how booster doses and prior infection influence the immune defense they elicit, and whether they protect against variants. Focusing on the T cell response, we conducted a longitudinal study of infection-naïve and COVID-19 convalescent donors before vaccination and after their first and second vaccine doses, using a high-parameter CyTOF analysis to phenotype their SARS-CoV-2-specific T cells. Vaccine-elicited spike-specific T cells responded similarly to stimulation by spike epitopes from the ancestral, B.1.1.7 and B.1.351 variant strains, both in terms of cell numbers and phenotypes. In infection-naïve individuals, the second dose boosted the quantity and altered the phenotypic properties of SARS-CoV-2-specific T cells, while in convalescents the second dose changed neither. Spike-specific T cells from convalescent vaccinees differed strikingly from those of infection-naïve vaccinees, with phenotypic features suggesting superior long-term persistence and ability to home to the respiratory tract including the nasopharynx. These results provide reassurance that vaccine-elicited T cells respond robustly to emerging viral variants, confirm that convalescents may not need a second vaccine dose, and suggest that vaccinated convalescents may have more persistent nasopharynx-homing SARS-CoV-2-specific T cells compared to their infection-naïve counterparts.


Vaccination is one of the best ways to prevent severe COVID-19. Two doses of mRNA vaccine protect against serious illness caused by the coronavirus SARS-CoV-2. They do this, in part, by encouraging the immune system to make specialised proteins known as antibodies that recognise the virus. Most of the vaccine research so far has focussed on these antibodies, but they are only one part of the immune response. Vaccines also activate immune cells called T cells. These cells have two main roles, coordinating the immune response and killing cells infected with viruses. It is likely that they play a key role in preventing severe COVID-19. There are many kinds of T cells, each with a different role. Currently, the identity and characteristics of the T cells that protect against COVID-19 is unclear. Different types of T cells have unique proteins on their surface. Examining these proteins can reveal details about how the T cells work, which part of the virus they recognise, and which part of the body they protect. A tool called cytometry by time of flight allows researchers to measure these proteins, one cell at a time. Using this technique, Neidleman, Luo et al. investigated T cells from 11 people before vaccination and after their first and second doses. Five people had never had COVID-19 before, and six had already recovered from COVID-19. Neidleman, Luo et al. found that the T cells recognizing SARS-CoV-2 in the two groups differed. In people who had never had COVID-19 before, the second dose of vaccine improved the quality and quantity of the T cells. The same was not true for people who had already recovered from COVID-19. However, although their T cells did not improve further after a second vaccine dose, they did show signs that they might offer more protection overall. The proteins on the cells suggest that they might last longer, and that they might specifically protect the nose, throat and lungs. Neidleman, Luo et al. also found that, for both groups, T cells activated by vaccination responded in the same way to different variants of the virus. This work highlights the importance of getting both vaccine doses for people who have never had COVID-19. It also suggests that vaccination in people who have had COVID-19 may generate better T cells. Larger studies could show whether these patterns remain true across the wider population. If so, it is possible that delivering vaccines to the nose or throat could boost immunity by mimicking natural infection. This might encourage T cells to make the surface proteins that allow them to home to these areas.


Subject(s)
COVID-19 Vaccines/pharmacology , COVID-19/immunology , SARS-CoV-2/immunology , T-Lymphocytes/drug effects , Vaccines, Synthetic/pharmacology , Adult , Aged , COVID-19/prevention & control , COVID-19/virology , Female , Humans , Immunization, Secondary , Longitudinal Studies , Male , Middle Aged , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Young Adult
13.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.10.12.464152

ABSTRACT

Blood clots are a central feature of coronavirus disease-2019 (COVID-19) and can culminate in pulmonary embolism, stroke, and sudden death. However, it is not known how abnormal blood clots form in COVID-19 or why they occur even in asymptomatic and convalescent patients. Here we report that the Spike protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) binds to the blood coagulation factor fibrinogen and induces structurally abnormal blood clots with heightened proinflammatory activity. SARS-CoV-2 Spike virions enhanced fibrin-mediated microglia activation and induced fibrinogen-dependent lung pathology. COVID-19 patients had fibrin autoantibodies that persisted long after acute infection. Monoclonal antibody 5B8, targeting the cryptic inflammatory fibrin epitope, inhibited thromboinflammation. Our results reveal a procoagulant role for the SARS-CoV-2 Spike and propose fibrin-targeting interventions as a treatment for thromboinflammation in COVID-19.


Subject(s)
Pulmonary Embolism , Coronavirus Infections , Blood Coagulation Disorders , Severe Acute Respiratory Syndrome , Death, Sudden , COVID-19 , Stroke
14.
Cell Rep ; 36(3): 109414, 2021 07 20.
Article in English | MEDLINE | ID: covidwho-1283975

ABSTRACT

Although T cells are likely players in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunity, little is known about the phenotypic features of SARS-CoV-2-specific T cells associated with recovery from severe coronavirus disease 2019 (COVID-19). We analyze T cells from 34 individuals with COVID-19 with severity ranging from mild (outpatient) to critical, culminating in death. Relative to individuals who succumbed, individuals who recovered from severe COVID-19 harbor elevated and increasing numbers of SARS-CoV-2-specific T cells capable of homeostatic proliferation. In contrast, fatal COVID-19 cases display elevated numbers of SARS-CoV-2-specific regulatory T cells and a time-dependent escalation in activated bystander CXCR4+ T cells, as assessed by longitudinal sampling. Together with the demonstration of increased proportions of inflammatory CXCR4+ T cells in the lungs of individuals with severe COVID-19, these results support a model where lung-homing T cells activated through bystander effects contribute to immunopathology, whereas a robust, non-suppressive SARS-CoV-2-specific T cell response limits pathogenesis and promotes recovery from severe COVID-19.

15.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.05.12.443888

ABSTRACT

While mRNA vaccines are proving highly efficacious against SARS-CoV-2, it is important to determine how booster doses and prior infection influence the immune defense they elicit, and whether they protect against variants. Focusing on the T cell response, we conducted a longitudinal study of infection-naive and COVID-19 convalescent donors before vaccination and after their first and second vaccine doses, using a high-parameter CyTOF analysis to phenotype their SARS-CoV-2-specific T cells. Vaccine-elicited spike-specific T cells responded similarly to stimulation by spike epitopes from the ancestral, B.1.1.7 and B.1.351 variant strains, both in terms of cell numbers and phenotypes. In infection-naive individuals, the second dose boosted the quantity but not quality of the T cell response, while in convalescents the second dose helped neither. Spike-specific T cells from convalescent vaccinees differed strikingly from those of infection-naive vaccinees, with phenotypic features suggesting superior long-term persistence and ability to home to the respiratory tract including the nasopharynx. These results provide reassurance that vaccine-elicited T cells respond robustly to the B.1.1.7 and B.1.351 variants, confirm that convalescents may not need a second vaccine dose, and suggest that vaccinated convalescents may have more persistent nasopharynx-homing SARS-CoV-2-specific T cells compared to their infection-naive counterparts.


Subject(s)
COVID-19 , Infections
16.
medrxiv; 2021.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2021.01.22.21250054

ABSTRACT

Although T cells are likely players in SARS-CoV-2 immunity, little is known about the phenotypic features of SARS-CoV-2-specific T cells associated with recovery from severe COVID-19. We analyzed T cells from longitudinal specimens of 34 COVID-19 patients with severities ranging from mild (outpatient) to critical culminating in death. Relative to patients that succumbed, individuals that recovered from severe COVID-19 harbored elevated and increasing numbers of SARS-CoV-2-specific T cells capable of homeostatic proliferation. In contrast, fatal COVID-19 displayed elevated numbers of SARS-CoV-2-specific regulatory T cells and a time-dependent escalation in activated bystander CXCR4+ T cells. Together with the demonstration of increased proportions of inflammatory CXCR4+ T cells in the lungs of severe COVID-19 patients, these results support a model whereby lung-homing T cells activated through bystander effects contribute to immunopathology, while a robust, non-suppressive SARS-CoV-2-specific T cell response limits pathogenesis and promotes recovery from severe COVID-19.


Subject(s)
COVID-19 , Death
17.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.01.27.428543

ABSTRACT

Tremendous progress has been made to control the COVID-19 pandemic, including the development and approval of vaccines as well as the drug remdesivir, which inhibits the SARS-CoV-2 virus that causes COVID-19. However, remdesivir confers only mild benefits to a subset of patients, and additional effective therapeutic options are needed. Drug repurposing and drug combinations may represent practical strategies to address these urgent unmet medical needs. Viruses, including coronaviruses, are known to hijack the host metabolism to facilitate their own proliferation, making targeting host metabolism a promising antiviral approach. Here, we describe an integrated analysis of 12 published in vitro and human patient gene expression datasets on SARS-CoV-2 infection using genome-scale metabolic modeling (GEM). We find that SARS-CoV-2 infection can induce recurrent and complicated metabolic reprogramming spanning a wide range of metabolic pathways. We next applied the GEM-based metabolic transformation algorithm (MTA) to predict anti-SARS-CoV-2 targets that counteract the virus-induced metabolic changes. These predictions are enriched for validated targets from various published experimental drug and genetic screens. Further analyzing the RNA-sequencing data of remdesivir-treated Vero E6 cell samples that we generated, we predicted metabolic targets that act in combination with remdesivir. These predictions are enriched for previously reported synergistic drugs with remdesivir. Since our predictions are based in part on human patient data, they are likely to be clinically relevant. We provide our top high-confidence candidate targets for their evaluation in further studies, demonstrating host metabolism-targeting as a promising antiviral strategy.


Subject(s)
COVID-19
18.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.01.27.428516

ABSTRACT

The SARS-CoV-2 Spike glycoprotein mediates virus entry and is a major target for neutralizing antibodies. All current vaccines are based on the ancestral Spike with the goal of generating protective neutralizing antibodies. Several novel SARS-CoV-2 variants with multiple Spike mutations have emerged, and their rapid spread and potential for immune escape have raised concerns. One of these variants, first identified in the United Kingdom, B.1.1.7 (also called VUI202012/01), contains eight Spike mutations with potential to impact antibody therapy, vaccine efficacy and risk of reinfection. Here we employed a lentivirus-based pseudovirus assay to show that variant B.1.1.7 remains sensitive to neutralization, albeit at moderately reduced levels (~2-fold), by serum samples from convalescent individuals and recipients of two different vaccines based on ancestral Spike (mRNA-1273, Moderna, and protein nanoparticle (NVX-CoV2373, Novavax). Some monoclonal antibodies to the receptor binding domain (RBD) of Spike were less effective against the variant while others were largely unaffected. These findings indicate that B.1.1.7 is not a neutralization escape variant that would be a major concern for current vaccines, or for risk of reinfection.


Subject(s)
Severe Acute Respiratory Syndrome
19.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.01.21.427501

ABSTRACT

Vertical transmission of SARS-CoV-2, the virus responsible for COVID-19, from parents to early embryos during conception could be catastrophic, but is contingent on the susceptibility of cells of the embryo to infection. Because presence of the SARS-CoV-2 virus has been reported in the human reproductive system, we assessed whether pre-implantation embryos are permissive to SARS-CoV-2 entry. RNA-seq and immunostaining studies revealed presence of two key entry factors in the trophectoderm of blastocyst-stage embryos, the ACE2 receptor and the TMPRSS2 protease. Exposure of blastocysts to fluorescent reporter virions pseudotyped with the SARS-CoV-2 Spike (S) glycoprotein revealed S-ACE2 dependent entry and fusion. These results indicate that human pre-implantation embryos can be infected by SARS-CoV-2, a finding pertinent to natural human conceptions and assisted reproductive technologies during and after the COVID-19 pandemic.


Subject(s)
Severe Acute Respiratory Syndrome , COVID-19
20.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.01.21.427563

ABSTRACT

The coronavirus pandemic introduced many changes to our society, and deeply affected the established in biomedical sciences publication practices. In this article, we present a comprehensive study of the changes in scholarly publication landscape for biomedical sciences during the COVID-19 pandemic, with special emphasis on preprints posted on bioRxiv and medRxiv servers. We observe the emergence of a new category of preprint authors working in the fields of immunology, microbiology, infectious diseases, and epidemiology, who extensively used preprint platforms during the pandemic for sharing their immediate findings. The majority of these findings were works-in-progress unfitting for a prompt acceptance by refereed journals. The COVID-19 preprints that became peer-reviewed journal articles were often submitted to journals concurrently with the posting on a preprint server, and the entire publication cycle, from preprint to the online journal article, took on average 63 days. This included an expedited peer-review process of 43 days and journals production stage of 15 days, however there was a wide variation in publication delays between journals. Only one third of COVID-19 preprints posted during the first nine months of the pandemic appeared as peer-reviewed journal articles. These journal articles display high Altmetric Attention Scores further emphasizing a significance of COVID-19 research during 2020. This article will be relevant to editors, publishers, open science enthusiasts, and anyone interested in changes that the 2020 crisis transpired to publication practices and a culture of preprints in life sciences.


Subject(s)
COVID-19 , Fractures, Open , Communicable Diseases
SELECTION OF CITATIONS
SEARCH DETAIL