81 results on '"Boucau J"'
Search Results
2. Implications of post-translational modifications of IRF7 on pDC IFN-alpha response
- Author
-
Griesbeck M, Doyle E, Lindsay RJ, Boucau J, LeGall S, Altfeld M, and Chang JJ
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2012
- Full Text
- View/download PDF
3. Alteration of HIV epitope processing and presentation by HIV protease inhibitors
- Author
-
Kourjian G, Boucau J, Shimada M, Lai NY, and Le Gall S
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2012
- Full Text
- View/download PDF
4. Monoclonal antibody treatment drives rapid culture conversion in SARS-CoV-2 infection
- Author
-
Chew, K.W., Hughes, M.D., Choudhary, M.C., Deo, R., Ritz, J., Wohl, D.A., Greninger, A.L., Currier, J.S., Moser, C., Daar, E.S., Flynn, J.P., Javan, A.C., Eron, J.J., Klekotka, P., Boucau, J., Fischer II, W.A., Smith, D.M., Barczak, A.K., Li, J.Z., Crain, C.R., Regan, J., ACTIV-2/A5401 Study Team, Nirula, A., Dragavon, J.A., POSITIVES study team, and Coombs, R.W.
- Subjects
Clinical Trials and Supportive Activities ,mAbs ,Antibodies ,COVID therapies ,resistance ,Vaccine Related ,Clinical Research ,Biodefense ,Monoclonal ,Humans ,Neutralizing ,Humanized ,Lung ,COVID ,SARS-CoV-2 ,Prevention ,viral culture ,COVID-19 ,Pneumonia ,COVID-19 Drug Treatment ,Emerging Infectious Diseases ,Infectious Diseases ,Good Health and Well Being ,Pneumonia & Influenza ,POSITIVES study team ,ACTIV-2/A5401 Study Team ,Immunization ,monoclonal antibodies ,Infection ,Biotechnology - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) monoclonal antibodies (mAbs) are among the treatments recommended for high-risk ambulatory persons with coronavirus 2019 (COVID-19). Here, we study viral culture dynamics post-treatment in a subset of participants receiving the mAb bamlanivimab in the ACTIV-2 trial (ClinicalTrials.gov: NCT04518410). Viral load by qPCR and viral culture are performed from anterior nasal swabs collected on study days 0 (day of treatment), 1, 2, 3, and 7. Treatment with mAbs results in rapid clearance of culturable virus. One day after treatment, 0 of 28 (0%) participants receiving mAbs and 16 of 39 (41%) receiving placebo still have culturable virus (p
- Published
- 2022
5. Motifs within and outside epitopes define the efficiency of HIV epitope production in different subcellular compartments
- Author
-
Vaithilingam, A., Dinter, J., Zhang, S. -C., Kourjian, G., Boucau, J., Rucevic, M., and Le Gall, S.
- Published
- 2015
6. Drug-induced modulation of cellular activation during latency reversal changes antigen processing and peptide presentation in primary CD4 T cells
- Author
-
Boucau, J., primary, Sanchez-Bernabeu, A., additional, and Le Gall, S., additional
- Published
- 2017
- Full Text
- View/download PDF
7. Latency-reversing agents and cellular activation affect antigen processing in primary CD4 T cells
- Author
-
Boucau, J., primary, Madouasse, J., additional, Wambua, D., additional, Berberich, M.J., additional, and Le Gall, S., additional
- Published
- 2015
- Full Text
- View/download PDF
8. Crystal structure of Antigen 85C in presence of Ebselen
- Author
-
Favrot, L., primary, Grzegorzewicz, A.E., additional, Lajiness, D.H., additional, Marvin, R.K., additional, Boucau, J., additional, Isailovic, D., additional, Jackson, M., additional, and Ronning, D.R., additional
- Published
- 2013
- Full Text
- View/download PDF
9. Crystal structure of Antigen 85C-C209S mutant
- Author
-
Favrot, L., primary, Grzegorzewicz, A.E., additional, Lajiness, D.H., additional, Marvin, R.K., additional, Boucau, J., additional, Isailovic, D., additional, Jackson, M., additional, and Ronning, D.R., additional
- Published
- 2013
- Full Text
- View/download PDF
10. Crystal Structure of Antigen 85C and Glycerol
- Author
-
Boucau, J., primary, Sanki, A.K., additional, Umesiri, F.E., additional, Sucheck, S.J., additional, and Ronning, D.R., additional
- Published
- 2009
- Full Text
- View/download PDF
11. A New Bifunctional Catalyst - Asymmetric Co(II)-Mediated Wynberg Reaction
- Author
-
Lin, Y.-M., primary, Boucau, J., additional, Li, Z., additional, Casarotto, V., additional, Lin, J., additional, Nguyen, A., additional, and Ehrmantraut, J., additional
- Published
- 2007
- Full Text
- View/download PDF
12. The Design Analysis Re-engineering Tool—for design basis justification and safety related information management
- Author
-
Billington, A, primary, Blondiaux, P, additional, Boucau, J, additional, Cantineau, B, additional, and Mared, A, additional
- Published
- 1999
- Full Text
- View/download PDF
13. Abortive infection of bat fibroblasts with SARS-CoV-2.
- Author
-
Bisht P, Gallagher MD, Barrasa MI, Boucau J, Harding A, Déjosez M, Godoy-Parejo C, Bisher ME, de Nola G, Lytton-Jean AKR, Gehrke L, Zwaka TP, and Jaenisch R
- Subjects
- Humans, Animals, Induced Pluripotent Stem Cells virology, Induced Pluripotent Stem Cells metabolism, Virus Replication, Chiroptera virology, Fibroblasts virology, Fibroblasts metabolism, SARS-CoV-2 physiology, SARS-CoV-2 immunology, COVID-19 virology, COVID-19 immunology, Angiotensin-Converting Enzyme 2 metabolism, Angiotensin-Converting Enzyme 2 genetics
- Abstract
Bats are tolerant to highly pathogenic viruses such as Marburg, Ebola, and Nipah, suggesting the presence of a unique immune tolerance toward viral infection. Here, we compared severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of human and bat ( Rhinolophus ferrumequinum ) pluripotent cells and fibroblasts. Since bat cells do not express an angiotensin-converting enzyme 2 (ACE2) receptor that allows virus infection, we transduced the human ACE2 (hA) receptor into the cells and found that transduced cells can be infected with SARS-CoV-2. Compared to human embryonic stem cells-hA, infected bat induced Pluripotent Stem Cells (iPSCs)-hA produced about a 100-fold lower level of infectious virus and displayed lower toxicity. In contrast, bat embryonic fibroblast-hA produced no infectious virus while being infectable and synthesizing viral RNA and proteins, suggesting abortive infection. Indeed, electron microscopy failed to detect virus-like particles in infected bat fibroblasts in contrast to bat iPSCs or human cells, consistent with the latter producing infectious viruses. This suggests that bat somatic but not pluripotent cells have an effective mechanism to control virus replication. Consistent with previous results by others, we find that bat cells have a constitutively activated innate immune system, which might limit SARS-CoV-2 infection compared to human cells., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
14. Modeling suggests SARS-CoV-2 rebound after nirmatrelvir-ritonavir treatment is driven by target cell preservation coupled with incomplete viral clearance.
- Author
-
Phan T, Ribeiro RM, Edelstein GE, Boucau J, Uddin R, Marino C, Liew MY, Barry M, Choudhary MC, Tien D, Su K, Reynolds Z, Li Y, Sagar S, Vyas TD, Kawano Y, Sparks JA, Hammond SP, Wallace Z, Vyas JM, Li JZ, Siedner MJ, Barczak AK, Lemieux JE, and Perelson AS
- Abstract
In a subset of SARS-CoV-2 infected individuals treated with the oral antiviral nirmatrelvir-ritonavir, the virus rebounds following treatment. The mechanisms driving this rebound are not well understood. We used a mathematical model to describe the longitudinal viral load dynamics of 51 individuals treated with nirmatrelvir-ritonavir, 20 of whom rebounded. Target cell preservation, either by a robust innate immune response or initiation of nirmatrelvir-ritonavir near the time of symptom onset, coupled with incomplete viral clearance, appear to be the main factors leading to viral rebound. Moreover, the occurrence of viral rebound is likely influenced by time of treatment initiation relative to the progression of the infection, with earlier treatments leading to a higher chance of rebound. Finally, our model demonstrates that extending the course of nirmatrelvir-ritonavir treatment, in particular to a 10-day regimen, may greatly diminish the risk for rebound in people with mild-to-moderate COVID-19 and who are at high risk of progression to severe disease. Altogether, our results suggest that in some individuals, a standard 5-day course of nirmatrelvir-ritonavir starting around the time of symptom onset may not completely eliminate the virus. Thus, after treatment ends, the virus can rebound if an effective adaptive immune response has not fully developed. These findings on the role of target cell preservation and incomplete viral clearance also offer a possible explanation for viral rebounds following other antiviral treatments for SARS-CoV-2., Competing Interests: Competing interests: ASP owns stock in Pfizer. He was also on a Pfizer advisory committee and received an honorarium. The other authors declare that they have no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
15. Emerging SARS-CoV-2 Resistance After Antiviral Treatment.
- Author
-
Tamura TJ, Choudhary MC, Deo R, Yousuf F, Gomez AN, Edelstein GE, Boucau J, Glover OT, Barry M, Gilbert RF, Reynolds Z, Li Y, Tien D, Vyas TD, Passell E, Su K, Drapkin S, Abar EG, Kawano Y, Sparks JA, Wallace ZS, Vyas JM, Shafer RW, Siedner MJ, Barczak AK, Lemieux JE, and Li JZ
- Subjects
- Humans, Female, Male, Middle Aged, Adult, Mutation, COVID-19 epidemiology, Aged, Cohort Studies, Antiviral Agents therapeutic use, SARS-CoV-2, Drug Resistance, Viral genetics, Alanine analogs & derivatives, Alanine therapeutic use, COVID-19 Drug Treatment, Adenosine Monophosphate analogs & derivatives, Adenosine Monophosphate therapeutic use
- Abstract
Importance: Previous studies have identified mutations in SARS-CoV-2 strains that confer resistance to nirmatrelvir, yet how often this resistance arises and its association with posttreatment virologic rebound is not well understood., Objective: To examine the prevalence of emergent antiviral resistance after nirmatrelvir treatment and its association with virologic rebound., Design, Setting, and Participants: This cohort study enrolled outpatient adults with acute COVID-19 infection from May 2021 to October 2023. Participants were divided into those who received antiviral therapy and those who did not. The study was conducted at a multicenter health care system in Boston, Massachusetts., Exposure: Treatment regimen, including none, nirmatrelvir, and remdesivir., Main Outcomes and Measures: The primary outcome was emergent SARS-CoV-2 antiviral resistance, defined as the detection of antiviral resistance mutations, which were not present at baseline, were previously associated with decreased antiviral efficacy, and emerged during or after completion of a participant's treatment. Next-generation sequencing was used to detect low frequency mutations down to 1% of the total viral population., Results: Overall, 156 participants (114 female [73.1%]; median [IQR] age, 56 [38-69] years) were included. Compared with 63 untreated individuals, the 79 who received nirmatrelvir were older and more commonly immunosuppressed. After sequencing viral RNA from participants' anterior nasal swabs, nirmatrelvir resistance mutations were detected in 9 individuals who received nirmatrelvir (11.4%) compared with 2 of those who did not (3.2%) (P = .09). Among the individuals treated with nirmatrelvir, those who were immunosuppressed had the highest frequency of resistance emergence (5 of 22 [22.7%]), significantly greater than untreated individuals (2 of 63 [3.1%]) (P = .01). Similar rates of nirmatrelvir resistance were found in those who had virologic rebound (3 of 23 [13.0%]) vs those who did not (6 of 56 [10.7%]) (P = .86). Most of these mutations (10 of 11 [90.9%]) were detected at low frequencies (<20% of viral population) and reverted to the wild type at subsequent time points. Emerging remdesivir resistance mutations were only detected in immunosuppressed individuals (2 of 14 [14.3%]) but were similarly low frequency and transient. Global Initiative on Sharing All Influenza Data analysis showed no evidence of increased nirmatrelvir resistance in the United States after the authorization of nirmatrelvir., Conclusions and Relevance: In this cohort study of 156 participants, treatment-emergent nirmatrelvir resistance mutations were commonly detected, especially in individuals who were immunosuppressed. However, these mutations were generally present at low frequencies and were transient in nature, suggesting a low risk for the spread of nirmatrelvir resistance in the community with the current variants and drug usage patterns.
- Published
- 2024
- Full Text
- View/download PDF
16. A mouse model of TB-associated lung fibrosis reveals persistent inflammatory macrophage populations during treatment.
- Author
-
Boucau J, Naidoo T, Liu Y, Dasgupta S, Jain N, Castillo JR, Jacobson NE, Nargan K, Cimini BA, Eliceiri KW, Steyn AJC, and Barczak AK
- Abstract
Post-TB lung disease (PTLD) causes a significant burden of global disease. Fibrosis is a central component of many clinical features of PTLD. To date, we have a limited understanding of the mechanisms of TB-associated fibrosis and how these mechanisms are similar to or dissimilar from other fibrotic lung pathologies. We have adapted a mouse model of TB infection to facilitate the mechanistic study of TB-associated lung fibrosis. We find that the morphologies of fibrosis that develop in the mouse model are similar to the morphologies of fibrosis observed in human tissue samples. Using Second Harmonic Generation (SHG) microscopy, we are able to quantify a major component of fibrosis, fibrillar collagen, over time and with treatment. Inflammatory macrophage subpopulations persist during treatment; matrix remodeling enzymes and inflammatory gene signatures remain elevated. Our mouse model suggests that there is a therapeutic window during which adjunctive therapies could change matrix remodeling or inflammatory drivers of tissue pathology to improve functional outcomes after treatment for TB infection.
- Published
- 2024
- Full Text
- View/download PDF
17. Evasion of NKG2D-mediated cytotoxic immunity by sarbecoviruses.
- Author
-
Hartmann JA, Cardoso MR, Talarico MCR, Kenney DJ, Leone MR, Reese DC, Turcinovic J, O'Connell AK, Gertje HP, Marino C, Ojeda PE, De Paula EV, Orsi FA, Velloso LA, Cafiero TR, Connor JH, Ploss A, Hoelzemer A, Carrington M, Barczak AK, Crossland NA, Douam F, Boucau J, and Garcia-Beltran WF
- Subjects
- Humans, Histocompatibility Antigens Class I immunology, Histocompatibility Antigens Class I metabolism, Animals, Cytotoxicity, Immunologic, Down-Regulation, Lung immunology, Lung virology, Lung pathology, SARS-CoV-2 immunology, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, NK Cell Lectin-Like Receptor Subfamily K metabolism, COVID-19 immunology, COVID-19 virology, Immune Evasion
- Abstract
SARS-CoV-2 and other sarbecoviruses continue to threaten humanity, highlighting the need to characterize common mechanisms of viral immune evasion for pandemic preparedness. Cytotoxic lymphocytes are vital for antiviral immunity and express NKG2D, an activating receptor conserved among mammals that recognizes infection-induced stress ligands (e.g., MIC-A/B). We found that SARS-CoV-2 evades NKG2D recognition by surface downregulation of MIC-A/B via shedding, observed in human lung tissue and COVID-19 patient serum. Systematic testing of SARS-CoV-2 proteins revealed that ORF6, an accessory protein uniquely conserved among sarbecoviruses, was responsible for MIC-A/B downregulation via shedding. Further investigation demonstrated that natural killer (NK) cells efficiently killed SARS-CoV-2-infected cells and limited viral spread. However, inhibition of MIC-A/B shedding with a monoclonal antibody, 7C6, further enhanced NK-cell activity toward SARS-CoV-2-infected cells. Our findings unveil a strategy employed by SARS-CoV-2 to evade cytotoxic immunity, identify the culprit immunevasin shared among sarbecoviruses, and suggest a potential novel antiviral immunotherapy., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Enhancing antibody responses by multivalent antigen display on thymus-independent DNA origami scaffolds.
- Author
-
Wamhoff EC, Ronsard L, Feldman J, Knappe GA, Hauser BM, Romanov A, Case JB, Sanapala S, Lam EC, Denis KJS, Boucau J, Barczak AK, Balazs AB, Diamond MS, Schmidt AG, Lingwood D, and Bathe M
- Subjects
- Humans, Animals, Mice, Antibodies, Blocking, Antibodies, Neutralizing, DNA, Antibodies, Viral, Antibody Formation, Vaccines, Virus-Like Particle genetics, Spike Glycoprotein, Coronavirus
- Abstract
Protein-based virus-like particles (P-VLPs) are commonly used to spatially organize antigens and enhance humoral immunity through multivalent antigen display. However, P-VLPs are thymus-dependent antigens that are themselves immunogenic and can induce B cell responses that may neutralize the platform. Here, we investigate thymus-independent DNA origami as an alternative material for multivalent antigen display using the receptor binding domain (RBD) of the SARS-CoV-2 spike protein, the primary target of neutralizing antibody responses. Sequential immunization of mice with DNA-based VLPs (DNA-VLPs) elicits protective neutralizing antibodies to SARS-CoV-2 in a manner that depends on the valency of the antigen displayed and on T cell help. Importantly, the immune sera do not contain boosted, class-switched antibodies against the DNA scaffold, in contrast to P-VLPs that elicit strong B cell memory against both the target antigen and the scaffold. Thus, DNA-VLPs enhance target antigen immunogenicity without generating scaffold-directed immunity and thereby offer an important alternative material for particulate vaccine design., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
19. SARS-CoV-2 viral clearance and evolution varies by type and severity of immunodeficiency.
- Author
-
Li Y, Choudhary MC, Regan J, Boucau J, Nathan A, Speidel T, Liew MY, Edelstein GE, Kawano Y, Uddin R, Deo R, Marino C, Getz MA, Reynolds Z, Barry M, Gilbert RF, Tien D, Sagar S, Vyas TD, Flynn JP, Hammond SP, Novack LA, Choi B, Cernadas M, Wallace ZS, Sparks JA, Vyas JM, Seaman MS, Gaiha GD, Siedner MJ, Barczak AK, Lemieux JE, and Li JZ
- Subjects
- Humans, Prospective Studies, Kinetics, Immunosuppression Therapy, SARS-CoV-2, COVID-19
- Abstract
Despite vaccination and antiviral therapies, immunocompromised individuals are at risk for prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, but the immune defects that predispose an individual to persistent coronavirus disease 2019 (COVID-19) remain incompletely understood. In this study, we performed detailed viro-immunologic analyses of a prospective cohort of participants with COVID-19. The median times to nasal viral RNA and culture clearance in individuals with severe immunosuppression due to hematologic malignancy or transplant (S-HT) were 72 and 40 days, respectively, both of which were significantly longer than clearance rates in individuals with severe immunosuppression due to autoimmunity or B cell deficiency (S-A), individuals with nonsevere immunodeficiency, and nonimmunocompromised groups ( P < 0.01). Participants who were severely immunocompromised had greater SARS-CoV-2 evolution and a higher risk of developing resistance against therapeutic monoclonal antibodies. Both S-HT and S-A participants had diminished SARS-CoV-2-specific humoral responses, whereas only the S-HT group had reduced T cell-mediated responses. This highlights the varied risk of persistent COVID-19 across distinct immunosuppressive conditions and suggests that suppression of both B and T cell responses results in the highest contributing risk of persistent infection.
- Published
- 2024
- Full Text
- View/download PDF
20. SARS-CoV-2 Virologic Rebound With Nirmatrelvir-Ritonavir Therapy : An Observational Study.
- Author
-
Edelstein GE, Boucau J, Uddin R, Marino C, Liew MY, Barry M, Choudhary MC, Gilbert RF, Reynolds Z, Li Y, Tien D, Sagar S, Vyas TD, Kawano Y, Sparks JA, Hammond SP, Wallace Z, Vyas JM, Barczak AK, Lemieux JE, Li JZ, and Siedner MJ
- Subjects
- Adult, Humans, Ritonavir therapeutic use, COVID-19 Drug Treatment, SARS-CoV-2, COVID-19
- Abstract
Background: Data are conflicting regarding an association between treatment of acute COVID-19 with nirmatrelvir-ritonavir (N-R) and virologic rebound (VR)., Objective: To compare the frequency of VR in patients with and without N-R treatment for acute COVID-19., Design: Observational cohort study., Setting: Multicenter health care system in Boston, Massachusetts., Participants: Ambulatory adults with acute COVID-19 with and without use of N-R., Intervention: Receipt of 5 days of N-R treatment versus no COVID-19 therapy., Measurements: The primary outcome was VR, defined as either a positive SARS-CoV-2 viral culture result after a prior negative result or 2 consecutive viral loads above 4.0 log
10 copies/mL that were also at least 1.0 log10 copies/mL higher than a prior viral load below 4.0 log10 copies/mL., Results: Compared with untreated persons ( n = 55), those taking N-R ( n = 72) were older, received more COVID-19 vaccinations, and more commonly had immunosuppression. Fifteen participants (20.8%) taking N-R had VR versus 1 (1.8%) who was untreated (absolute difference, 19.0 percentage points [95% CI, 9.0 to 29.0 percentage points]; P = 0.001). All persons with VR had a positive viral culture result after a prior negative result. In multivariable models, only N-R use was associated with VR (adjusted odds ratio, 10.02 [CI, 1.13 to 88.74]; P = 0.038). Virologic rebound was more common among those who started therapy within 2 days of symptom onset (26.3%) than among those who started 2 or more days after symptom onset (0%) ( P = 0.030). Among participants receiving N-R, those who had VR had prolonged shedding of replication-competent virus compared with those who did not have VR (median, 14 vs. 3 days). Eight of 16 participants (50% [CI, 25% to 75%]) with VR also reported symptom rebound; 2 were completely asymptomatic. No post-VR resistance mutations were detected., Limitations: Observational study design with differences between the treated and untreated groups; positive viral culture result was used as a surrogate marker for risk for ongoing viral transmission., Conclusion: Virologic rebound occurred in approximately 1 in 5 people taking N-R, often without symptom rebound, and was associated with shedding of replication-competent virus., Primary Funding Source: National Institutes of Health., Competing Interests: Disclosures: Disclosures can be viewed at www.acponline.org/authors/icmje/ConflictOfInterestForms.do?msNum=M23-1756.- Published
- 2023
- Full Text
- View/download PDF
21. TLR2 is non-redundant in the population and subpopulation responses to Mycobacterium tuberculosis in macrophages and in vivo .
- Author
-
Jani C, Solomon SL, Peters JM, Pringle SC, Hinman AE, Boucau J, Bryson BD, and Barczak AK
- Subjects
- Cells, Cultured, Animals, Mice, Pathogen-Associated Molecular Pattern Molecules, Interferon Type I immunology, Microbial Viability, NF-kappa B immunology, Cellular Microenvironment immunology, Mycobacterium tuberculosis, Macrophages immunology, Macrophages microbiology, Tuberculosis immunology, Toll-Like Receptor 2 immunology, Host-Pathogen Interactions immunology
- Abstract
Tuberculosis (TB), caused by the pathogenic bacterium Mycobacterium tuberculosis (Mtb), is a global health threat. Targeting host pathways that modulate protective or harmful components of inflammation has been proposed as a therapeutic strategy that could aid sterilization or mitigate TB-associated permanent tissue damage. In purified form, many Mtb components can activate innate immune pathways. However, knowledge of the pathways that contribute most to the observed response to live Mtb is incomplete, limiting the possibility of precise intervention. We took a systematic, unbiased approach to define the pathways that drive the earliest immune response to Mtb. Using a macrophage model of infection, we compared the bulk transcriptional response to infection with the response to a panel of Mtb-derived putative innate immune ligands. We identified two axes of response: an NF-kB-dependent response similarly elicited by all Mtb pathogen-associated molecular patterns (PAMPs) and a type I interferon axis unique to cells infected with live Mtb. Consistent with growing literature data pointing to TLR2 as a dominant Mtb-associated PAMP, the TLR2 ligand PIM6 most closely approximated the NF-kB-dependent response to the intact bacterium. Quantitatively, the macrophage response to Mtb was slower and weaker than the response to purified PIM6. On a subpopulation level, the TLR2-dependent response was heterogeneously induced, with only a subset of infected cells expressing key inflammatory genes known to contribute to the control of infection. Despite potential redundancies in Mtb ligand/innate immune receptor interactions during in vivo infection, loss of the TLR2/PIM6 interaction impacted the cellular composition of both the innate and adaptive compartments. IMPORTANCE Tuberculosis (TB) is a leading cause of death globally. Drug resistance is outpacing new antibiotic discovery, and even after successful treatment, individuals are often left with permanent lung damage from the negative consequences of inflammation. Targeting host inflammatory pathways has been proposed as an approach that could either improve sterilization or improve post-treatment lung health. However, our understanding of the inflammatory pathways triggered by Mycobacterium tuberculosis (Mtb) in infected cells and lungs is incomplete, in part because of the complex array of potential molecular interactions between bacterium and host. Here, we take an unbiased approach to identify the pathways most central to the host response to Mtb. We examine how individual pathways are triggered differently by purified Mtb products or infection with the live bacterium and consider how these pathways inform the emergence of subpopulation responses in cell culture and in infected mice. Understanding how individual interactions and immune pathways contribute to inflammation in TB opens the door to the possibility of developing precise therapeutic interventions., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
22. Anamnestic humoral correlates of immunity across SARS-CoV-2 variants of concern.
- Author
-
McNamara RP, Maron JS, Boucau J, Roy V, Webb NE, Bertera HL, Barczak AK, Positives Study Staff T, Franko N, Logue JK, Kemp M, Li JZ, Zhou L, Hsieh CL, McLellan JS, Siedner MJ, Seaman MS, Lemieux JE, Chu HY, and Alter G
- Subjects
- Humans, SARS-CoV-2 genetics, Antibodies, Antibodies, Viral, Antibodies, Neutralizing, COVID-19
- Abstract
While immune correlates against SARS-CoV-2 are typically defined at peak immunogenicity following vaccination, immunologic responses that expand selectively during the anamnestic response following infection can provide mechanistic and detailed insights into the immune mechanisms of protection. Moreover, whether anamnestic correlates are conserved across variants of concern (VOC), including the Delta and more distant Omicron VOC, remains unclear. To define the anamnestic correlates of immunity, across VOCs, we deeply profiled the humoral immune response in individuals infected with sequence-confirmed Delta or Omicron VOC after completing the vaccination series. While limited acute N-terminal domain and receptor-binding domain (RBD)-specific immune expansion was observed following breakthrough infection, a significant immunodominant expansion of opsonophagocytic Spike-specific antibody responses focused largely on the conserved S2-domain of SARS-CoV-2 was observed. This S2-specific functional humoral response continued to evolve over 2-3 weeks following Delta or Omicron breakthrough, targeting multiple VOCs and common coronaviruses. Strong responses were observed on the fusion peptide (FP) region and the heptad repeat 1 (HR1) region adjacent to the RBD. Notably, the FP is highly conserved across SARS-related coronaviruses and even non-SARS-related betacoronavirus. Taken together, our results point to a critical role of highly conserved, functional S2-specific responses in the anamnestic antibody response to SARS-CoV-2 infection across VOCs. These humoral responses linked to virus clearance can guide next-generation vaccine-boosting approaches to confer broad protection against future SARS-related coronaviruses. IMPORTANCE The Spike protein of SARS-CoV-2 is the primary target of antibody-based recognition. Selective pressures, be it the adaption to human-to-human transmission or evasion of previously acquired immunity, have spurred the emergence of variants of the virus such as the Delta and Omicron lineages. Therefore, understanding how antibody responses are expanded in breakthrough cases of previously vaccinated individuals can provide insights into key correlates of protection against current and future variants. Here, we show that vaccinated individuals who had documented COVID-19 breakthrough showed anamnestic antibody expansions targeting the conserved S2 subdomain of Spike, particularly within the fusion peptide region. These S2-directed antibodies were highly leveraged for non-neutralizing, phagocytic functions and were similarly expanded independent of the variant. We propose that through deep profiling of anamnestic antibody responses in breakthrough cases, we can identify antigen targets susceptible to novel monoclonal antibody therapy or vaccination-boosting strategies., Competing Interests: Galit Alter is a founder/equity holder in Seromyx Systems and Leyden Labs. G.A. has served as a scientific advisor for Sanofi Vaccines. G.A. has collaborative agreements with GSK, Merck, Abbvie, Sanofi, Medicago, BioNtech, Moderna, BMS, Novavax, SK Biosciences, Gilead, and Sanaria.
- Published
- 2023
- Full Text
- View/download PDF
23. SARS-CoV-2 Viral Clearance and Evolution Varies by Extent of Immunodeficiency.
- Author
-
Li Y, Choudhary MC, Regan J, Boucau J, Nathan A, Speidel T, Liew MY, Edelstein GE, Kawano Y, Uddin R, Deo R, Marino C, Getz MA, Reynold Z, Barry M, Gilbert RF, Tien D, Sagar S, Vyas TD, Flynn JP, Hammond SP, Novack LA, Choi B, Cernadas M, Wallace ZS, Sparks JA, Vyas JM, Seaman MS, Gaiha GD, Siedner MJ, Barczak AK, Lemieux JE, and Li JZ
- Abstract
Despite vaccination and antiviral therapies, immunocompromised individuals are at risk for prolonged SARS-CoV-2 infection, but the immune defects that predispose to persistent COVID-19 remain incompletely understood. In this study, we performed detailed viro-immunologic analyses of a prospective cohort of participants with COVID-19. The median time to nasal viral RNA and culture clearance in the severe hematologic malignancy/transplant group (S-HT) were 72 and 40 days, respectively, which were significantly longer than clearance rates in the severe autoimmune/B-cell deficient (S-A), non-severe, and non-immunocompromised groups (P<0.001). Participants who were severely immunocompromised had greater SARS-CoV-2 evolution and a higher risk of developing antiviral treatment resistance. Both S-HT and S-A participants had diminished SARS-CoV-2-specific humoral, while only the S-HT group had reduced T cell-mediated responses. This highlights the varied risk of persistent COVID-19 across immunosuppressive conditions and suggests that suppression of both B and T cell responses results in the highest contributing risk of persistent infection.
- Published
- 2023
- Full Text
- View/download PDF
24. SARS-CoV-2 virologic rebound with nirmatrelvir-ritonavir therapy.
- Author
-
Edelstein GE, Boucau J, Uddin R, Marino C, Liew MY, Barry M, Choudhary MC, Gilbert RF, Reynolds Z, Li Y, Tien D, Sagar S, Vyas TD, Kawano Y, Sparks JA, Hammond SP, Wallace Z, Vyas JM, Barczak AK, Lemieux JE, Li JZ, and Siedner MJ
- Abstract
Objective: To compare the frequency of replication-competent virologic rebound with and without nirmatrelvir-ritonavir treatment for acute COVID-19. Secondary aims were to estimate the validity of symptoms to detect rebound and the incidence of emergent nirmatrelvir-resistance mutations after rebound., Design: Observational cohort study., Setting: Multicenter healthcare system in Boston, Massachusetts., Participants: We enrolled ambulatory adults with a positive COVID-19 test and/or a prescription for nirmatrelvir-ritonavir., Exposures: Receipt of 5 days of nirmatrelvir-ritonavir treatment versus no COVID-19 therapy., Main Outcome and Measures: The primary outcome was COVID-19 virologic rebound, defined as either (1) a positive SARS-CoV-2 viral culture following a prior negative culture or (2) two consecutive viral loads ≥4.0 log
10 copies/milliliter after a prior reduction in viral load to <4.0 log10 copies/milliliter., Results: Compared with untreated individuals (n=55), those taking nirmatrelvir-ritonavir (n=72) were older, received more COVID-19 vaccinations, and were more commonly immunosuppressed. Fifteen individuals (20.8%) taking nirmatrelvir-ritonavir experienced virologic rebound versus one (1.8%) of the untreated (absolute difference 19.0% [95%CI 9.0-29.0%], P=0.001). In multivariable models, only N-R was associated with VR (AOR 10.02, 95%CI 1.13-88.74). VR occurred more commonly among those with earlier nirmatrelvir-ritonavir initiation (29.0%, 16.7% and 0% when initiated days 0, 1, and ≥2 after diagnosis, respectively, P=0.089). Among participants on N-R, those experiencing rebound had prolonged shedding of replication-competent virus compared to those that did not rebound (median: 14 vs 3 days). Only 8/16 with virologic rebound reported worsening symptoms (50%, 95%CI 25%-75%); 2 were completely asymptomatic. We detected no post-rebound nirmatrelvir-resistance mutations in the NSP5 protease gene., Conclusions and Relevance: Virologic rebound occurred in approximately one in five people taking nirmatrelvir-ritonavir and often occurred without worsening symptoms. Because it is associated with replication-competent viral shedding, close monitoring and potential isolation of those who rebound should be considered.- Published
- 2023
- Full Text
- View/download PDF
25. Extrafollicular IgD - CD27 - CXCR5 - CD11c - DN3 B cells infiltrate inflamed tissues in autoimmune fibrosis and in severe COVID-19.
- Author
-
Allard-Chamard H, Kaneko N, Bertocchi A, Sun N, Boucau J, Kuo HH, Farmer JR, Perugino C, Mahajan VS, Murphy SJH, Premo K, Diefenbach T, Ghebremichael M, Yuen G, Kotta A, Akman Z, Lichterfeld M, Walker BD, Yu XG, Moriyama M, Maehara T, Nakamura S, Stone JH, Padera RF, and Pillai S
- Subjects
- Humans, Fibrosis, Immunoglobulin D, Inflammation, Receptors, CXCR5, COVID-19, Immunoglobulin G4-Related Disease, B-Lymphocyte Subsets metabolism, B-Lymphocyte Subsets pathology
- Abstract
Although therapeutic B cell depletion dramatically resolves inflammation in many diseases in which antibodies appear not to play a central role, distinct extrafollicular pathogenic B cell subsets that accumulate in disease lesions have hitherto not been identified. The circulating immunoglobulin D (IgD)
- CD27- CXCR5- CD11c+ DN2 B cell subset has been previously studied in some autoimmune diseases. A distinct IgD- CD27- CXCR5- CD11c- DN3 B cell subset accumulates in the blood both in IgG4-related disease, an autoimmune disease in which inflammation and fibrosis can be reversed by B cell depletion, and in severe COVID-19. These DN3 B cells prominently accumulate in the end organs of IgG4-related disease and in lung lesions in COVID-19, and double-negative B cells prominently cluster with CD4+ T cells in these lesions. Extrafollicular DN3 B cells may participate in tissue inflammation and fibrosis in autoimmune fibrotic diseases, as well as in COVID-19., Competing Interests: Declaration of interests S.P. is on the scientific advisory boards of Abpro, Inc., BeBiopharma, Inc., Paratus Biosciences, and Octagon Therapeutics., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
26. Enhancing antibody responses by multivalent antigen display on thymus-independent DNA origami scaffolds.
- Author
-
Wamhoff EC, Ronsard L, Feldman J, Knappe GA, Hauser BM, Romanov A, Lam E, Denis KS, Boucau J, Barczak AK, Balazs AB, Schmidt A, Lingwood D, and Bathe M
- Abstract
Multivalent antigen display is a well-established principle to enhance humoral immunity. Protein-based virus-like particles (VLPs) are commonly used to spatially organize antigens. However, protein-based VLPs are limited in their ability to control valency on fixed scaffold geometries and are thymus-dependent antigens that elicit neutralizing B cell memory themselves, which can distract immune responses. Here, we investigated DNA origami as an alternative material for multivalent antigen display in vivo, applied to the receptor binding domain (RBD) of SARS-CoV2 that is the primary antigenic target of neutralizing antibody responses. Icosahedral DNA-VLPs elicited neutralizing antibodies to SARS-CoV-2 in a valency-dependent manner following sequential immunization in mice, quantified by pseudo- and live-virus neutralization assays. Further, induction of B cell memory against the RBD required T cell help, but the immune sera did not contain boosted, class-switched antibodies against the DNA scaffold. This contrasted with protein-based VLP display of the RBD that elicited B cell memory against both the target antigen and the scaffold. Thus, DNA-based VLPs enhance target antigen immunogenicity without generating off-target, scaffold-directed immune memory, thereby offering a potentially important alternative material for particulate vaccine design., Competing Interests: Competing interests The Massachusetts Institute of Technology has filed a patent (US application number 16/752,394) covering the use of DNA origami as a vaccine platform on behalf of the co-inventors (E.-C. W. and M. B.).
- Published
- 2023
- Full Text
- View/download PDF
27. Characterization of Virologic Rebound Following Nirmatrelvir-Ritonavir Treatment for Coronavirus Disease 2019 (COVID-19).
- Author
-
Boucau J, Uddin R, Marino C, Regan J, Flynn JP, Choudhary MC, Chen G, Stuckwisch AM, Mathews J, Liew MY, Singh A, Reynolds Z, Iyer SL, Chamberlin GC, Vyas TD, Vyas JM, Turbett SE, Li JZ, Lemieux JE, Barczak AK, and Siedner MJ
- Subjects
- Humans, COVID-19 Drug Treatment, Ritonavir therapeutic use, Mutation, COVID-19
- Abstract
We enrolled 7 individuals with recurrent symptoms or antigen test conversion following nirmatrelvir-ritonavir treatment. High viral loads (median 6.1 log10 copies/mL) were detected after rebound for a median of 17 days after initial diagnosis. Three had culturable virus for up to 16 days after initial diagnosis. No known resistance-associated mutations were identified., Competing Interests: Potential conflicts of interest. J. E. L. reports grant to institution from the Centers for Disease Control and Prevention (CDC) outside of the submitted work; consulting fees from Sherlock Biosciences paid to individual; and honoraria paid to individual from Emerson Hospital and Virology Education. S. E. T. reports grants or contracts from the CDC for COVID-related work paid to the institution outside of the submitted work and travel support from the Duke Clinical Research Institute paid to self. All other authors report no conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2022. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
28. Cross-reactive SARS-CoV-2 epitope targeted across donors informs immunogen design.
- Author
-
Hauser BM, Feldman J, Sangesland M, Ronsard L, St Denis KJ, Sheehan ML, Cao Y, Boucau J, Windsor IW, Cheng AH, Vu ML, Cardoso MR, Kannegieter T, Balazs AB, Lingwood D, Garcia-Beltran WF, and Schmidt AG
- Subjects
- Animals, Humans, Mice, Epitopes genetics, COVID-19 Vaccines, Antibodies, SARS-CoV-2 genetics, COVID-19 prevention & control
- Abstract
The emergence of the antigenically distinct and highly transmissible Omicron variant highlights the possibility of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune escape due to viral evolution. This continued evolution, along with the possible introduction of new sarbecoviruses from zoonotic reservoirs, may evade host immunity elicited by current SARS-CoV-2 vaccines. Identifying cross-reactive antibodies and defining their epitope(s) can provide templates for rational immunogen design strategies for next-generation vaccines. Here, we characterize the receptor-binding-domain-directed, cross-reactive humoral repertoire across 10 human vaccinated donors. We identify cross-reactive antibodies from diverse gene rearrangements targeting two conserved receptor-binding domain epitopes. An engineered immunogen enriches antibody responses to one of these conserved epitopes in mice with pre-existing SARS-CoV-2 immunity; elicited responses neutralize SARS-CoV-2, variants, and related sarbecoviruses. These data show how immune focusing to a conserved epitope targeted by human cross-reactive antibodies may guide pan-sarbecovirus vaccine development, providing a template for identifying such epitopes and translating to immunogen design., Competing Interests: Declaration of interests B.M.H. and A.G.S. have filed a provisional patent for the described immunogens., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
29. Inhibition of major histocompatibility complex-I antigen presentation by sarbecovirus ORF7a proteins.
- Author
-
Zhang F, Zang TM, Stevenson EM, Lei X, Copertino DC, Mota TM, Boucau J, Garcia-Beltran WF, Jones RB, and Bieniasz PD
- Subjects
- Amino Acids, Humans, Major Histocompatibility Complex, Peptides, SARS-CoV-2, Antigen Presentation, CD8-Positive T-Lymphocytes immunology, COVID-19 immunology, Histocompatibility Antigens Class I immunology, Viral Proteins immunology
- Abstract
Viruses employ a variety of strategies to escape or counteract immune responses, including depletion of cell surface major histocompatibility complex class I (MHC-I), that would ordinarily present viral peptides to CD8
+ cytotoxic T cells. As part of a screen to elucidate biological activities associated with individual severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) viral proteins, we found that ORF7a reduced cell surface MHC-I levels by approximately fivefold. Nevertheless, in cells infected with SARS-CoV-2, surface MHC-I levels were reduced even in the absence of ORF7a, suggesting additional mechanisms of MHC-I down-regulation. ORF7a proteins from a sample of sarbecoviruses varied in their ability to induce MHC-I down-regulation and, unlike SARS-CoV-2, the ORF7a protein from SARS-CoV lacked MHC-I downregulating activity. A single amino acid at position 59 (T/F) that is variable among sarbecovirus ORF7a proteins governed the difference in MHC-I downregulating activity. SARS-CoV-2 ORF7a physically associated with the MHC-I heavy chain and inhibited the presentation of expressed antigen to CD8+ T cells. Specifically, ORF7a prevented the assembly of the MHC-I peptide loading complex and caused retention of MHC-I in the endoplasmic reticulum. The differential ability of ORF7a proteins to function in this way might affect sarbecovirus dissemination and persistence in human populations, particularly those with infection- or vaccine-elicited immunity.- Published
- 2022
- Full Text
- View/download PDF
30. Vaccine breakthrough infection leads to distinct profiles of neutralizing antibody responses by SARS-CoV-2 variant.
- Author
-
Seaman MS, Siedner MJ, Boucau J, Lavine CL, Ghantous F, Liew MY, Mathews JI, Singh A, Marino C, Regan J, Uddin R, Choudhary MC, Flynn JP, Chen G, Stuckwisch AM, Lipiner T, Kittilson A, Melberg M, Gilbert RF, Reynolds Z, Iyer SL, Chamberlin GC, Vyas TD, Vyas JM, Goldberg MB, Luban J, Li JZ, Barczak AK, and Lemieux JE
- Subjects
- Antibodies, Neutralizing, Antibodies, Viral, BCG Vaccine, COVID-19 Vaccines, Convalescence, Diphtheria-Tetanus-Pertussis Vaccine, Humans, Measles-Mumps-Rubella Vaccine, Neutralization Tests, SARS-CoV-2, AIDS Vaccines, COVID-19 prevention & control, Influenza Vaccines, Papillomavirus Vaccines, Respiratory Syncytial Virus Vaccines, SAIDS Vaccines
- Abstract
Protective immunity against SARS-CoV-2 infection after COVID-19 vaccination may differ by variant. We enrolled vaccinated (n = 39) and unvaccinated (n = 11) individuals with acute, symptomatic SARS-CoV-2 Delta or Omicron infection and performed SARS-CoV-2 viral load quantification, whole-genome sequencing, and variant-specific antibody characterization at the time of acute illness and convalescence. Viral load at the time of infection was inversely correlated with antibody binding and neutralizing antibody responses. Across all variants tested, convalescent neutralization titers in unvaccinated individuals were markedly lower than in vaccinated individuals. Increases in antibody titers and neutralizing activity occurred at convalescence in a variant-specific manner. For example, among individuals infected with the Delta variant, neutralizing antibody responses were weakest against BA.2, whereas infection with Omicron BA.1 variant generated a broader response against all tested variants, including BA.2.
- Published
- 2022
- Full Text
- View/download PDF
31. Permanent, Antimicrobial Coating to Rapidly Kill and Prevent Transmission of Bacteria, Fungi, Influenza, and SARS-CoV-2.
- Author
-
Ghosh S, Mukherjee R, Mahajan VS, Boucau J, Pillai S, and Haldar J
- Subjects
- Acrylates pharmacology, Anti-Bacterial Agents chemistry, Bacteria, Fungi, Humans, Nylons pharmacology, Polyethylenes pharmacology, Rubber, SARS-CoV-2, Anti-Infective Agents pharmacology, COVID-19 prevention & control, Disinfectants pharmacology, Influenza A Virus, H1N1 Subtype, Influenza, Human
- Abstract
Microbial adhesion and contamination on shared surfaces can lead to life-threatening infections with serious impacts on public health, economy, and clinical practices. The traditional use of chemical disinfectants for sanitization of surfaces, however, comes with its share of health risks, such as hazardous effects on the eyes, skin, and respiratory tract, carcinogenicity, as well as environmental toxicity. To address this, we have developed a nonleaching quaternary small molecule (QSM)-based sprayable coating which can be fabricated on a wide range of surfaces such as nylon, polyethylene, surgical mask, paper, acrylate, and rubber in a one-step, photocuring technique. This contact-active coating killed pathogenic bacteria and fungi including drug-resistant strains of Staphylococcus aureus and Candida albicans within 15-30 min of contact. QSM coatings withstood multiple washes, highlighting their durability. Interestingly, the coated surfaces exhibited rapid killing of pathogens, leading to the prevention of their transmission upon contact. The coating showed membrane disruption of bacterial cells in fluorescence and electron microscopic investigations. Along with bacteria and fungi, QSM-coated surfaces also showed the complete killing of high loads of influenza (H1N1) and SARS-CoV-2 viruses within 30 min of exposure. To our knowledge, this is the first report of a coating for multipurpose materials applied in high-touch public places, hospital equipment, and clinical consumables, rapidly killing drug-resistant bacteria, fungi, influenza virus, and SARS-CoV-2.
- Published
- 2022
- Full Text
- View/download PDF
32. Viral Kinetics of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Omicron Infection in mRNA-Vaccinated Individuals Treated and Not Treated with Nirmatrelvir-Ritonavir.
- Author
-
Dai EY, Lee KA, Nathanson AB, Leonelli AT, Petros BA, Brock-Fisher T, Dobbins ST, MacInnis BL, Capone A, Littlehale N, Boucau J, Marino C, Barczak AK, Sabeti PC, Springer M, and Stephenson KE
- Abstract
We measured viral kinetics of SARS-CoV-2 Omicron infection in 36 mRNA-vaccinated individuals, 11 of whom were treated with nirmatrelvir-ritonavir (NMV-r). We found that NMV-r was associated with greater incidence of viral rebound compared to no treatment. For those that did not rebound, NMV-r significantly reduced time to PCR conversion., Competing Interests: CONFLICT OF INTERESTS P.C.S. is a co-founder of, shareholder in, and scientific advisor to Sherlock Biosciences, Inc; she is also a Board member of and shareholder in Danaher Corporation. P.C.S. has filed IP related to genome sequencing and analysis. The authors declare no other conflicts of interests.
- Published
- 2022
- Full Text
- View/download PDF
33. Duration of Shedding of Culturable Virus in SARS-CoV-2 Omicron (BA.1) Infection.
- Author
-
Boucau J, Marino C, Regan J, Uddin R, Choudhary MC, Flynn JP, Chen G, Stuckwisch AM, Mathews J, Liew MY, Singh A, Lipiner T, Kittilson A, Melberg M, Li Y, Gilbert RF, Reynolds Z, Iyer SL, Chamberlin GC, Vyas TD, Goldberg MB, Vyas JM, Li JZ, Lemieux JE, Siedner MJ, and Barczak AK
- Subjects
- Animals, Chlorocebus aethiops, Humans, Spike Glycoprotein, Coronavirus, Vero Cells, COVID-19 virology, SARS-CoV-2 isolation & purification, SARS-CoV-2 physiology, Virus Cultivation methods, Virus Shedding physiology
- Published
- 2022
- Full Text
- View/download PDF
34. Monoclonal antibody treatment drives rapid culture conversion in SARS-CoV-2 infection.
- Author
-
Boucau J, Chew KW, Choudhary MC, Deo R, Regan J, Flynn JP, Crain CR, Hughes MD, Ritz J, Moser C, Dragavon JA, Javan AC, Nirula A, Klekotka P, Greninger AL, Coombs RW, Fischer WA 2nd, Daar ES, Wohl DA, Eron JJ, Currier JS, Smith DM, Li JZ, and Barczak AK
- Subjects
- Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Humanized, Antibodies, Neutralizing, Humans, SARS-CoV-2, COVID-19 Drug Treatment
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) monoclonal antibodies (mAbs) are among the treatments recommended for high-risk ambulatory persons with coronavirus 2019 (COVID-19). Here, we study viral culture dynamics post-treatment in a subset of participants receiving the mAb bamlanivimab in the ACTIV-2 trial (ClinicalTrials.gov: NCT04518410). Viral load by qPCR and viral culture are performed from anterior nasal swabs collected on study days 0 (day of treatment), 1, 2, 3, and 7. Treatment with mAbs results in rapid clearance of culturable virus. One day after treatment, 0 of 28 (0%) participants receiving mAbs and 16 of 39 (41%) receiving placebo still have culturable virus (p < 0.0001). Recrudescence of culturable virus is detected in three participants with emerging mAb resistance and viral RNA rebound. While further studies are necessary to fully define the relationship between shed culturable virus and transmission, these results raise the possibility that mAbs may offer immediate (household) and public-health benefits by reducing onward transmission., Competing Interests: Declaration of interests J.B., M.C.C., R.D., J. Regan, J.P.F., C.R.C., M.D.H., J. Ritz, C.M., J.A.D., A.C.J., R.W.C., J.S.C., and A.K.B. report no competing interests. K.W.C. reports research grant support to the institution from Merck Sharp & Dohme. A.N. and P.K. are employees and shareholders of Eli Lilly. A.L.G. declares central testing contracts with Abbott and research support from Gilead and Merck outside of the submitted work. W.A.F. reports research funding from Ridgeback Biopharmaceuticals and consultancy fees from Roche and Merck and serves on adjudication committees for Janssen and Syneos. E.S.D. reports consulting fees from Gilead Sciences and Merck and research support to the institution from Gilead Sciences and ViiV. J.J.E. reports serving as an ad hoc consultant to GSK/VIR and as data monitoring committee (DMC) chair for Adagio Phase III studies. D.M.S. reports consulting fees from the following companies: Fluxergy, Kiadis, Linear Therapies, Matrix BioMed, Arena Pharmaceuticals, VxBiosciences, Model Medicines, Bayer Pharmaceuticals, Signant Health, and Brio Clinical. J.Z.L. reports consulting for Abbvie and Recovery Therapeutics., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
35. FcγR-mediated SARS-CoV-2 infection of monocytes activates inflammation.
- Author
-
Junqueira C, Crespo Â, Ranjbar S, de Lacerda LB, Lewandrowski M, Ingber J, Parry B, Ravid S, Clark S, Schrimpf MR, Ho F, Beakes C, Margolin J, Russell N, Kays K, Boucau J, Das Adhikari U, Vora SM, Leger V, Gehrke L, Henderson LA, Janssen E, Kwon D, Sander C, Abraham J, Goldberg MB, Wu H, Mehta G, Bell S, Goldfeld AE, Filbin MR, and Lieberman J
- Subjects
- Caspase 1 metabolism, DNA-Binding Proteins, Humans, Inflammasomes metabolism, NLR Family, Pyrin Domain-Containing 3 Protein, Phosphate-Binding Proteins, Pore Forming Cytotoxic Proteins, COVID-19 virology, Inflammation metabolism, Inflammation virology, Monocytes metabolism, Monocytes virology, Receptors, IgG metabolism, SARS-CoV-2
- Abstract
SARS-CoV-2 can cause acute respiratory distress and death in some patients
1 . Although severe COVID-19 is linked to substantial inflammation, how SARS-CoV-2 triggers inflammation is not clear2 . Monocytes and macrophages are sentinel cells that sense invasive infection to form inflammasomes that activate caspase-1 and gasdermin D, leading to inflammatory death (pyroptosis) and the release of potent inflammatory mediators3 . Here we show that about 6% of blood monocytes of patients with COVID-19 are infected with SARS-CoV-2. Monocyte infection depends on the uptake of antibody-opsonized virus by Fcγ receptors. The plasma of vaccine recipients does not promote antibody-dependent monocyte infection. SARS-CoV-2 begins to replicate in monocytes, but infection is aborted, and infectious virus is not detected in the supernatants of cultures of infected monocytes. Instead, infected cells undergo pyroptosis mediated by activation of NLRP3 and AIM2 inflammasomes, caspase-1 and gasdermin D. Moreover, tissue-resident macrophages, but not infected epithelial and endothelial cells, from lung autopsies from patients with COVID-19 have activated inflammasomes. Taken together, these findings suggest that antibody-mediated SARS-CoV-2 uptake by monocytes and macrophages triggers inflammatory cell death that aborts the production of infectious virus but causes systemic inflammation that contributes to COVID-19 pathogenesis., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
36. Determining the Incidence of Asymptomatic SARS-CoV-2 Among Early Recipients of COVID-19 Vaccines (DISCOVER-COVID-19): A Prospective Cohort Study of Healthcare Workers Before, During and After Vaccination.
- Author
-
North CM, Barczak A, Goldstein RH, Healy BC, Finkelstein DM, Ding DD, Kim A, Boucau J, Shaw B, Gilbert RF, Vyas T, Reynolds Z, Siddle KJ, MacInnis BL, Regan J, Flynn JP, Choudhary MC, Vyas JM, Laskowski K, Dighe AS, Lemieux JE, Li JZ, Baden LR, Siedner MJ, Woolley AE, and Sacks CA
- Subjects
- COVID-19 Vaccines, Health Personnel, Humans, Incidence, Prospective Studies, SARS-CoV-2, Vaccination, COVID-19 epidemiology, COVID-19 prevention & control
- Abstract
The impact of coronavirus disease 2019 vaccination on viral characteristics of breakthrough infections is unknown. In this prospective cohort study, incidence of severe acute respiratory syndrome coronavirus 2 infection decreased following vaccination. Although asymptomatic positive tests were observed following vaccination, the higher cycle thresholds, repeat negative tests, and inability to culture virus raise questions about their clinical significance., (© The Author(s) 2021. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
37. Temporal changes in T cell subsets and expansion of cytotoxic CD4+ T cells in the lungs in severe COVID-19.
- Author
-
Kaneko N, Boucau J, Kuo HH, Perugino C, Mahajan VS, Farmer JR, Liu H, Diefenbach TJ, Piechocka-Trocha A, Lefteri K, Waring MT, Premo KR, Walker BD, Li JZ, Gaiha G, Yu XG, Lichterfeld M, Padera RF Jr, and Pillai S
- Subjects
- CD8-Positive T-Lymphocytes, Humans, Lung, T-Lymphocyte Subsets, T-Lymphocytes, Cytotoxic, CD4-Positive T-Lymphocytes, COVID-19
- Abstract
Many studies have been performed in severe COVID-19 on immune cells in the circulation and on cells obtained by bronchoalveolar lavage. Most studies have tended to provide relative information rather than a quantitative view, and it is a combination of approaches by various groups that is helping the field build a picture of the mechanisms that drive severe lung disease. Approaches employed to date have not revealed information on lung parenchymal T cell subsets in severe COVID-19. Therefore, we sought to examine early and late T cell subset alterations in the lungs and draining lymph nodes in severe COVID-19 using a rapid autopsy protocol and quantitative imaging approaches. Here, we have established that cytotoxic CD4+ T cells (CD4 + CTLs) increase in the lungs, draining lymph nodes and blood as COVID-19 progresses. CD4 + CTLs are prominently expanded in the lung parenchyma in severe COVID-19. In contrast CD8+ T cells are not prominent, exhibit increased PD-1 expression, and no obvious increase is seen in the number of Granzyme B+ CD8+ T cells in the lung parenchyma in severe COVID-19. Based on quantitative evidence for re-activation in the lung milieu, CD4 + CTLs may be as likely to drive viral clearance as CD8+ T cells and may also be contributors to lung inflammation and eventually to fibrosis in severe COVID-19., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
38. Vaccine Breakthrough Infection with the SARS-CoV-2 Delta or Omicron (BA.1) Variant Leads to Distinct Profiles of Neutralizing Antibody Responses.
- Author
-
Seaman MS, Siedner MJ, Boucau J, Lavine CL, Ghantous F, Liew MY, Mathews J, Singh A, Marino C, Regan J, Uddin R, Choudhary MC, Flynn JP, Chen G, Stuckwisch AM, Lipiner T, Kittilson A, Melberg M, Gilbert RF, Reynolds Z, Iyer SL, Chamberlin GC, Vyas TD, Vyas JM, Goldberg MB, Luban J, Li JZ, Barczak AK, and Lemieux JE
- Abstract
There is increasing evidence that the risk of SARS-CoV-2 infection among vaccinated individuals is variant-specific, suggesting that protective immunity against SARS-CoV-2 may differ by variant. We enrolled vaccinated (n = 39) and unvaccinated (n = 11) individuals with acute, symptomatic SARS-CoV-2 Delta or Omicron infection and performed SARS-CoV-2 viral load quantification, whole-genome sequencing, and variant-specific antibody characterization at the time of acute illness and convalescence. Viral load at the time of infection was inversely correlated with antibody binding and neutralizing antibody responses. Increases in antibody titers and neutralizing activity occurred at convalescence in a variant-specific manner. Across all variants tested, convalescent neutralization titers in unvaccinated individuals were markedly lower than in vaccinated individuals. For individuals infected with the Delta variant, neutralizing antibody responses were weakest against BA.2, whereas infection with Omicron BA.1 variant generated a broader response against all tested variants, including BA.2.
- Published
- 2022
- Full Text
- View/download PDF
39. Duration of viable virus shedding in SARS-CoV-2 omicron variant infection.
- Author
-
Boucau J, Marino C, Regan J, Uddin R, Choudhary MC, Flynn JP, Chen G, Stuckwisch AM, Mathews J, Liew MY, Singh A, Lipiner T, Kittilson A, Melberg M, Li Y, Gilbert RF, Reynolds Z, Iyer SL, Chamberlin GC, Vyas TD, Goldberg MB, Vyas JM, Li JZ, Lemieux JE, Siedner MJ, and Barczak AK
- Abstract
Clinical features of SARS-CoV-2 Omicron variant infection, including incubation period and transmission rates, distinguish this variant from preceding variants. However, whether the duration of shedding of viable virus differs between omicron and previous variants is not well understood. To characterize how variant and vaccination status impact shedding of viable virus, we serially sampled symptomatic outpatients newly diagnosed with COVID-19. Anterior nasal swabs were tested for viral load, sequencing, and viral culture. Time to PCR conversion was similar between individuals infected with the Delta and the Omicron variant. Time to culture conversion was also similar, with a median time to culture conversion of 6 days (interquartile range 4-8 days) in both groups. There were also no differences in time to PCR or culture conversion by vaccination status.
- Published
- 2022
- Full Text
- View/download PDF
40. Detection of the Omicron Variant Virus With the Abbott BinaxNow SARS-CoV-2 Rapid Antigen Assay.
- Author
-
Regan J, Flynn JP, Choudhary MC, Uddin R, Lemieux J, Boucau J, Bhattacharyya RP, Barczak AK, Li JZ, and Siedner MJ
- Abstract
We assessed the ability of the BinaxNow rapid test to detect severe acute respiratory syndrome coronavirus 2 antigen from 4 individuals with Omicron and Delta infections. We performed serial dilutions of nasal swab samples, and specimens with concentrations of ≥100
- Published
- 2022
- Full Text
- View/download PDF
41. Duration of viral shedding and culture positivity with postvaccination SARS-CoV-2 delta variant infections.
- Author
-
Siedner MJ, Boucau J, Gilbert RF, Uddin R, Luu J, Haneuse S, Vyas T, Reynolds Z, Iyer S, Chamberlin GC, Goldstein RH, North CM, Sacks CA, Regan J, Flynn JP, Choudhary MC, Vyas JM, Barczak AK, Lemieux JE, and Li JZ
- Subjects
- Adult, COVID-19 prevention & control, Female, Humans, Male, Middle Aged, Time Factors, COVID-19 genetics, COVID-19 metabolism, COVID-19 Vaccines administration & dosage, SARS-CoV-2 physiology, Virus Replication, Virus Shedding physiology
- Abstract
Isolation guidelines for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are largely derived from data collected prior to the emergence of the delta variant. We followed a cohort of ambulatory patients with postvaccination breakthrough SARS-CoV-2 infections with longitudinal collection of nasal swabs for SARS-CoV-2 viral load quantification, whole-genome sequencing, and viral culture. All delta variant infections in our cohort were symptomatic, compared with 64% of non-delta variant infections. Symptomatic delta variant breakthrough infections were characterized by higher initial viral load, longer duration of virologic shedding by PCR, greater likelihood of replication-competent virus at early stages of infection, and longer duration of culturable virus compared with non-delta variants. The duration of time since vaccination was also correlated with both duration of PCR positivity and duration of detection of replication-competent virus. Nonetheless, no individuals with symptomatic delta variant infections had replication-competent virus by day 10 after symptom onset or 24 hours after resolution of symptoms. These data support US CDC isolation guidelines as of November 2021, which recommend isolation for 10 days or until symptom resolution and reinforce the importance of prompt testing and isolation among symptomatic individuals with delta breakthrough infections. Additional data are needed to evaluate these relationships among asymptomatic and more severe delta variant breakthrough infections.
- Published
- 2022
- Full Text
- View/download PDF
42. Monoclonal antibody treatment drives rapid culture conversion in SARS-CoV-2 infection.
- Author
-
Boucau J, Chew KW, Choudhary M, Deo R, Regan J, Flynn JP, Crain CR, Hughes MD, Ritz J, Moser C, Dragavon JA, Javan AC, Nirula A, Klekotka P, Greninger AL, Coombs RW, Fischer WA 2nd, Daar ES, Wohl DA, Eron JJ, Currier JS, Smith DM, Li JZ, and Barczak AK
- Abstract
Monoclonal antibodies (mAbs) are the treatment of choice for high-risk ambulatory persons with mild to moderate COVID-19. We studied viral culture dynamics post-treatment in a subset of participants receiving the mAb bamlanivimab in the ACTIV-2 trial. Viral load by qPCR and viral culture were performed from anterior nasal swabs collected on study days 0 (day of treatment), 1, 2, 3, and 7. Treatment with mAb resulted in rapid clearance of culturable virus in participants without treatment-emergent resistance. One day after treatment, 0 of 28 (0%) participants receiving mAb and 16 of 39 (41%) receiving placebo still had culturable virus (p <0.0001); nasal viral loads were only modestly lower in the mAb-treated group at days 2 and 3. Recrudescence of culturable virus was detected in three participants with emerging mAb resistance and viral load rebound. The rapid reduction in shedding of viable SARS-CoV-2 after mAb treatment highlights the potential role of mAbs in preventing disease transmission.
- Published
- 2021
- Full Text
- View/download PDF
43. Virologic Features of Severe Acute Respiratory Syndrome Coronavirus 2 Infection in Children.
- Author
-
Yonker LM, Boucau J, Regan J, Choudhary MC, Burns MD, Young N, Farkas EJ, Davis JP, Moschovis PP, Bernard Kinane T, Fasano A, Neilan AM, Li JZ, and Barczak AK
- Subjects
- Adolescent, Child, Child, Preschool, Humans, Infant, Infant, Newborn, Pandemics, SARS-CoV-2 genetics, Young Adult, COVID-19 diagnosis, COVID-19 pathology, Viral Load
- Abstract
Background: Data on pediatric coronavirus disease 2019 (COVID-19) has lagged behind adults throughout the pandemic. An understanding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral dynamics in children would enable data-driven public health guidance., Methods: Respiratory swabs were collected from children with COVID-19. Viral load was quantified by reverse-transcription polymerase chain reaction (RT-PCR); viral culture was assessed by direct observation of cytopathic effects and semiquantitative viral titers. Correlations with age, symptom duration, and disease severity were analyzed. SARS-CoV-2 whole genome sequences were compared with contemporaneous sequences., Results: One hundred ten children with COVID-19 (median age, 10 years [range, 2 weeks-21 years]) were included in this study. Age did not impact SARS-CoV-2 viral load. Children were most infectious within the first 5 days of illness, and severe disease did not correlate with increased viral loads. Pediatric SARS-CoV-2 sequences were representative of those in the community and novel variants were identified., Conclusions: Symptomatic and asymptomatic children can carry high quantities of live, replicating SARS-CoV-2, creating a potential reservoir for transmission and evolution of genetic variants. As guidance around social distancing and masking evolves following vaccine uptake in older populations, a clear understanding of SARS-CoV-2 infection dynamics in children is critical for rational development of public health policies and vaccination strategies to mitigate the impact of COVID-19., (© The Author(s) 2021. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
44. Virologic features of SARS-CoV-2 infection in children.
- Author
-
Yonker LM, Boucau J, Regan J, Choudhary MC, Burns MD, Young N, Farkas EJ, Davis JP, Moschovis PP, Kinane TB, Fasano A, Neilan AM, Li JZ, and Barczak AK
- Abstract
Background: Data on pediatric COVID-19 has lagged behind adults throughout the pandemic. An understanding of SARS-CoV-2 viral dynamics in children would enable data-driven public health guidance., Methods: Respiratory swabs were collected from children with COVID-19. Viral load was quantified by RT-PCR; viral culture was assessed by direct observation of cytopathic effects and semiquantitative viral titers. Correlations with age, symptom duration, and disease severity were analyzed. SARS-CoV-2 whole genome sequences were compared with contemporaneous sequences., Results: 110 children with COVID-19 (median age 10 years, range 2 weeks-21 years) were included in this study. Age did not impact SARS-CoV-2 viral load. Children were most infectious within the first five days of illness, and severe disease did not correlate with increased viral loads. Pediatric SARS-CoV-2 sequences were representative of those in the community and novel variants were identified., Conclusions: Symptomatic and asymptomatic children can carry high quantities of live, replicating SARS-CoV-2, creating a potential reservoir for transmission and evolution of genetic variants. As guidance around social distancing and masking evolves following vaccine uptake in older populations, a clear understanding of SARS-CoV-2 infection dynamics in children is critical for rational development of public health policies and vaccination strategies to mitigate the impact of COVID-19., Competing Interests: Conflicts of Interest The authors do not report any conflicts of interest.
- Published
- 2021
- Full Text
- View/download PDF
45. Expansion of Cytotoxic CD4+ T cells in the lungs in severe COVID-19.
- Author
-
Kaneko N, Boucau J, Kuo HH, Perugino C, Mahajan VS, Farmer JR, Liu H, Diefenbach TJ, Piechocka-Trocha A, Lefteri K, Waring MT, Premo KR, Walker BD, Li JZ, Gaiha G, Yu XG, Lichterfeld M, Padera RF, and Pillai S
- Abstract
The contributions of T cells infiltrating the lungs to SARS-CoV-2 clearance and disease progression are poorly understood. Although studies of CD8+ T cells in bronchoalveolar lavage and blood have suggested that these cells are exhausted in severe COVID-19, CD4+ T cells have not been systematically interrogated within the lung parenchyma. We establish here that cytotoxic CD4+ T cells (CD4+CTLs) are prominently expanded in the COVID-19 lung infiltrate. CD4+CTL numbers in the lung increase with disease severity and progression is accompanied by widespread HLA-DR expression on lung epithelial and endothelial cells, increased apoptosis of epithelial cells and tissue remodeling. Based on quantitative evidence for re-activation in the lung milieu, CD4+ CTLs are as likely to drive viral clearance as CD8+ T cells and may also be contributors to lung inflammation and eventually to fibrosis in severe COVID-19., In Brief: In severe COVID-19 cytotoxic CD4+ T cells accumulate in draining lymph nodes and in the lungs during the resolving phase of the disease. Re-activated cytotoxic CD4+ T cells and cytotoxic CD8+ T cells are present in roughly equivalent numbers in the lungs at this stage and these cells likely collaborate to eliminate virally infected cells and potentially induce fibrosis. A large fraction of epithelial and endothelial cells in the lung express HLA class II in COVID-19 and there is temporal convergence between CD4+CTL accumulation and apoptosis in the lung., Highlights: In severe COVID-19, activated CD4+ CTLs accumulate in the lungs late in diseaseThese cells likely participate in SARS-CoV-2 clearance, collaborating with CD8+ T cells many of which exhibit an exhausted phenotypeT cells likely contribute to the late exacerbation of inflammationCD4+CTLs have been linked to fibrosis in many disorders and could also be responsible for the eventual induction of fibrosis in a subset of COVID-19 patients., Summary: The contributions of T cells infiltrating the lungs to SARS-CoV-2 clearance and disease progression are poorly understood. Although studies of CD8+ T cells in bronchoalveolar lavage and blood have suggested that these cells are exhausted in severe COVID-19, CD4+ T cells have not been systematically interrogated within the lung parenchyma. We establish here that cytotoxic CD4+ T cells (CD4+CTLs) are prominently expanded in the COVID-19 lung infiltrate. CD4+CTL numbers in the lung increase with disease severity and progression is accompanied by widespread HLA-DR expression on lung epithelial and endothelial cells, increased apoptosis of epithelial cells and tissue remodeling. Based on quantitative evidence for re-activation in the lung milieu, CD4+ CTLs are as likely to drive viral clearance as CD8+ T cells and may also be contributors to lung inflammation and eventually to fibrosis in severe COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
46. Persistence and Evolution of SARS-CoV-2 in an Immunocompromised Host.
- Author
-
Choi B, Choudhary MC, Regan J, Sparks JA, Padera RF, Qiu X, Solomon IH, Kuo HH, Boucau J, Bowman K, Adhikari UD, Winkler ML, Mueller AA, Hsu TY, Desjardins M, Baden LR, Chan BT, Walker BD, Lichterfeld M, Brigl M, Kwon DS, Kanjilal S, Richardson ET, Jonsson AH, Alter G, Barczak AK, Hanage WP, Yu XG, Gaiha GD, Seaman MS, Cernadas M, and Li JZ
- Subjects
- COVID-19 diagnosis, Fatal Outcome, Humans, Male, Middle Aged, SARS-CoV-2 isolation & purification, Viral Load, Antiphospholipid Syndrome complications, COVID-19 complications, Immunocompromised Host
- Published
- 2020
- Full Text
- View/download PDF
47. A natural polymorphism of Mycobacterium tuberculosis in the esxH gene disrupts immunodomination by the TB10.4-specific CD8 T cell response.
- Author
-
Sutiwisesak R, Hicks ND, Boyce S, Murphy KC, Papavinasasundaram K, Carpenter SM, Boucau J, Joshi N, Le Gall S, Fortune SM, Sassetti CM, and Behar SM
- Subjects
- Animals, Antigens, Bacterial genetics, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Epitopes, T-Lymphocyte immunology, Humans, Mice, Mice, Inbred C57BL, Tuberculosis immunology, Antigens, Bacterial immunology, Mycobacterium tuberculosis genetics, Mycobacterium tuberculosis immunology
- Abstract
CD8 T cells provide limited protection against Mycobacterium tuberculosis (Mtb) infection in the mouse model. As Mtb causes chronic infection in mice and humans, we hypothesize that Mtb impairs T cell responses as an immune evasion strategy. TB10.4 is an immunodominant antigen in people, nonhuman primates, and mice, which is encoded by the esxH gene. In C57BL/6 mice, 30-50% of pulmonary CD8 T cells recognize the TB10.44-11 epitope. However, TB10.4-specific CD8 T cells fail to recognize Mtb-infected macrophages. We speculate that Mtb elicits immunodominant CD8 T cell responses to antigens that are inefficiently presented by infected cells, thereby focusing CD8 T cells on nonprotective antigens. Here, we leverage naturally occurring polymorphisms in esxH, which frequently occur in lineage 1 strains, to test this "decoy hypothesis". Using the clinical isolate 667, which contains an EsxHA10T polymorphism, we observe a drastic change in the hierarchy of CD8 T cells. Using isogenic Erd.EsxHA10T and Erd.EsxHWT strains, we prove that this polymorphism alters the hierarchy of immunodominant CD8 T cell responses. Our data are best explained by immunodomination, a mechanism by which competition for APC leads to dominant responses suppressing subdominant responses. These results were surprising as the variant epitope can bind to H2-Kb and is recognized by TB10.4-specific CD8 T cells. The dramatic change in TB10.4-specific CD8 responses resulted from increased proteolytic degradation of A10T variant, which destroyed the TB10.44-11epitope. Importantly, this polymorphism affected T cell priming and recognition of infected cells. These data support a model in which nonprotective CD8 T cells become immunodominant and suppress subdominant responses. Thus, polymorphisms between clinical Mtb strains, and BCG or H37Rv sequence-based vaccines could lead to a mismatch between T cells that are primed by vaccines and the epitopes presented by infected cells. Reprograming host immune responses should be considered in the future design of vaccines., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
48. Loss of Bcl-6-Expressing T Follicular Helper Cells and Germinal Centers in COVID-19.
- Author
-
Kaneko N, Kuo HH, Boucau J, Farmer JR, Allard-Chamard H, Mahajan VS, Piechocka-Trocha A, Lefteri K, Osborn M, Bals J, Bartsch YC, Bonheur N, Caradonna TM, Chevalier J, Chowdhury F, Diefenbach TJ, Einkauf K, Fallon J, Feldman J, Finn KK, Garcia-Broncano P, Hartana CA, Hauser BM, Jiang C, Kaplonek P, Karpell M, Koscher EC, Lian X, Liu H, Liu J, Ly NL, Michell AR, Rassadkina Y, Seiger K, Sessa L, Shin S, Singh N, Sun W, Sun X, Ticheli HJ, Waring MT, Zhu AL, Alter G, Li JZ, Lingwood D, Schmidt AG, Lichterfeld M, Walker BD, Yu XG, Padera RF Jr, and Pillai S
- Subjects
- Aged, Aged, 80 and over, B-Lymphocytes immunology, COVID-19, Female, Germinal Center pathology, Humans, Male, Middle Aged, Pandemics, Proto-Oncogene Proteins c-bcl-6 genetics, Proto-Oncogene Proteins c-bcl-6 metabolism, Spleen immunology, Spleen pathology, Tumor Necrosis Factor-alpha metabolism, Coronavirus Infections immunology, Germinal Center immunology, Pneumonia, Viral immunology, T-Lymphocytes, Helper-Inducer immunology
- Abstract
Humoral responses in coronavirus disease 2019 (COVID-19) are often of limited durability, as seen with other human coronavirus epidemics. To address the underlying etiology, we examined post mortem thoracic lymph nodes and spleens in acute SARS-CoV-2 infection and observed the absence of germinal centers and a striking reduction in Bcl-6
+ germinal center B cells but preservation of AID+ B cells. Absence of germinal centers correlated with an early specific block in Bcl-6+ TFH cell differentiation together with an increase in T-bet+ TH1 cells and aberrant extra-follicular TNF-α accumulation. Parallel peripheral blood studies revealed loss of transitional and follicular B cells in severe disease and accumulation of SARS-CoV-2-specific "disease-related" B cell populations. These data identify defective Bcl-6+ TFH cell generation and dysregulated humoral immune induction early in COVID-19 disease, providing a mechanistic explanation for the limited durability of antibody responses in coronavirus infections, and suggest that achieving herd immunity through natural infection may be difficult., Competing Interests: Declaration of Interests S.P. is on the SAB of Abpro Inc. and Pulsar Biopharma. G.A. is founder of Seromyx Systems Inc., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
49. The Loss of Bcl-6 Expressing T Follicular Helper Cells and the Absence of Germinal Centers in COVID-19.
- Author
-
Kaneko N, Kuo HH, Boucau J, Farmer JR, Allard-Chamard H, Mahajan VS, Piechocka-Trocha A, Lefteri K, Osborn M, Bals J, Bartsch YC, Bonheur N, Caradonna TM, Chevalier J, Chowdhury F, Diefenbach TJ, Einkauf K, Fallon J, Feldman J, Finn KK, Garcia-Broncano P, Hartana CA, Hauser BM, Jiang C, Kaplonek P, Karpell M, Koscher EC, Lian X, Liu H, Liu J, Ly NL, Michell AR, Rassadkina Y, Seiger K, Sessa L, Shin S, Singh N, Sun W, Sun X, Ticheli HJ, Waring MT, Zhu AL, Li J, Lingwood D, Schmidt AG, Lichterfeld M, Walker BD, Yu X, Padera RF Jr, and Pillai S
- Abstract
Humoral responses in COVID-19 disease are often of limited durability, as seen with other human coronavirus epidemics. To address the underlying etiology, we examined postmortem thoracic lymph nodes and spleens in acute SARS-CoV-2 infection and observed the absence of germinal centers, a striking reduction in Bcl-6+ germinal center B cells but preservation of AID+ B cells. Absence of germinal centers correlated with an early specific block in Bcl-6+TFH cell differentiation together with an increase in T-bet+TH1 cells and aberrant extra-follicular TNF-a accumulation. Parallel peripheral blood studies revealed loss of transitional and follicular B cells in severe disease and accumulation of SARS-CoV-2-specific "disease-related" B cell populations. These data identify defective Bcl-6+TFH cell generation and dysregulated humoral immune induction early in COVID-19 disease, providing a mechanistic explanation for the limited durability of antibody responses in coronavirus infections and suggest that achieving herd immunity through natural infection may be difficult. Funding: This work was supported by NIH U19 AI110495 to SP, NIH R01 AI146779 to AGS, NIH R01AI137057 and DP2DA042422 to DL, BMH was supported by NIGMS T32 GM007753, TMC was supported by T32 AI007245. Funding for these studies from the Massachusetts Consortium of Pathogen Readiness, the Mark and Lisa Schwartz Foundation and Enid Schwartz is also acknowledged. Conflict of Interest: None. Ethical Approval: This study was performed with the approval of the Institutional Review Boards at the Massachusetts General Hospital and the Brigham and Women's Hospital.
- Published
- 2020
- Full Text
- View/download PDF
50. Latency reversal agents modulate HIV antigen processing and presentation to CD8 T cells.
- Author
-
Boucau J, Das J, Joshi N, and Le Gall S
- Subjects
- CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes pathology, CD8-Positive T-Lymphocytes pathology, HIV Infections drug therapy, HIV Infections pathology, Humans, Protein Kinase C immunology, Virus Latency immunology, Antigen Presentation, Antigens, Viral immunology, CD8-Positive T-Lymphocytes immunology, HIV Infections immunology, HIV-1 physiology, Histone Deacetylase Inhibitors pharmacology, Protein Kinase C antagonists & inhibitors, Virus Latency drug effects
- Abstract
Latency reversal agents (LRA) variably induce HIV re-expression in CD4 T cells but reservoirs are not cleared. Whether HIV epitope presentation is similar between latency reversal and initial infection of CD4 T cells is unknown yet crucial to define immune responses able to detect HIV-infected CD4 T cells after latency reversal. HIV peptides displayed by MHC comes from the intracellular degradation of proteins by proteasomes and post-proteasomal peptidases but the impact of LRAs on antigen processing is not known. Here we show that HDAC inhibitors (HDCAi) reduced cytosolic proteolytic activities while PKC agonists (PKCa) increased them to a lesser extent than that induced by TCR activation. During the cytosolic degradation of long HIV peptides in LRA-treated CD4 T cells extracts, HDACi and PKCa modulated degradation patterns of peptides and altered the production of HIV epitopes in often opposite ways. Beyond known HIV epitopes, HDACi narrowed the coverage of HIV antigenic fragments by 8-11aa degradation peptides while PKCa broadened it. LRAs altered HIV infection kinetics and modulated CD8 T cell activation in an epitope- and time-dependent manner. Interestingly the efficiency of endogenous epitope processing and presentation to CD8 T cells was increased by PKCa Ingenol at early time points despite low levels of antigens. LRA-induced modulations of antigen processing should be considered and exploited to enhance and broaden HIV peptide presentation by CD4 T cells and to improve immune recognition after latency reversal. This property of LRAs, if confirmed with other antigens, might be exploited to improve immune detection of diseased cells beyond HIV., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.