443 results on '"Douek DC"'
Search Results
2. Delayed booster dosing improves human antigen-specific Ig and B cell responses to the RH5.1/AS01B malaria vaccine
- Author
-
Nielsen, CM, primary, Barrett, JR, additional, Davis, C, additional, Fallon, JK, additional, Goh, C, additional, Michell, AR, additional, Griffin, C, additional, Kwok, A, additional, Loos, C, additional, Darko, S, additional, Laboune, F, additional, Silk, SE, additional, Tekman, M, additional, Francica, JR, additional, Ransier, A, additional, Payne, RO, additional, Minassian, AM, additional, Lauffenburger, DA, additional, Seder, RA, additional, Douek, DC, additional, Alter, G, additional, and Draper, SJ, additional
- Published
- 2022
- Full Text
- View/download PDF
3. Tuning antiviral CD8 T-cell response via proline-altered peptide ligand vaccination
- Author
-
Douek, DC, Duru, AD, Sun, R, Allerbring, EB, Chadderton, J, Kadri, N, Han, X, Peqini, K, Uchtenhagen, H, Madhurantakam, C, Pellegrino, S, Sandalova, T, Nygren, P-A, Turner, SJ, Achour, A, Douek, DC, Duru, AD, Sun, R, Allerbring, EB, Chadderton, J, Kadri, N, Han, X, Peqini, K, Uchtenhagen, H, Madhurantakam, C, Pellegrino, S, Sandalova, T, Nygren, P-A, Turner, SJ, and Achour, A
- Abstract
Viral escape from CD8+ cytotoxic T lymphocyte responses correlates with disease progression and represents a significant challenge for vaccination. Here, we demonstrate that CD8+ T cell recognition of the naturally occurring MHC-I-restricted LCMV-associated immune escape variant Y4F is restored following vaccination with a proline-altered peptide ligand (APL). The APL increases MHC/peptide (pMHC) complex stability, rigidifies the peptide and facilitates T cell receptor (TCR) recognition through reduced entropy costs. Structural analyses of pMHC complexes before and after TCR binding, combined with biophysical analyses, revealed that although the TCR binds similarly to all complexes, the p3P modification alters the conformations of a very limited amount of specific MHC and peptide residues, facilitating efficient TCR recognition. This approach can be easily introduced in peptides restricted to other MHC alleles, and can be combined with currently available and future vaccination protocols in order to prevent viral immune escape.
- Published
- 2020
4. The peripheral differentiation of human natural killer T cells
- Author
-
Liu, J, Hill, BJ, Darko, S, Song, K, Quigley, MF, Asher, TE, Morita, Y, Greenaway, HY, Venturi, V, Douek, DC, Davenport, MP, Price, DA, Roederer, M, Liu, J, Hill, BJ, Darko, S, Song, K, Quigley, MF, Asher, TE, Morita, Y, Greenaway, HY, Venturi, V, Douek, DC, Davenport, MP, Price, DA, and Roederer, M
- Abstract
The peripheral maturation of human CD1d-restricted natural killer T (NKT) cells has not been well described. In this study, we identified four major subsets of NKT cells in adults, distinguished by the expression of CD4, CD8 and CCR5. Phenotypic analysis suggested a hierarchical pattern of differentiation, whereby immature CD4+CD8−CCR5− cells progressed to an intermediate CD4+CD8−CCR5+ stage, which remained less differentiated than the CD4−CD8− and CD4−CD8+ subsets, both of which expressed CCR5. This interpretation was supported by functional data, including clonogenic potential and cytokine secretion profiles, as well as T-cell receptor (TCR) excision circle analysis. Moreover, conventional and high-throughput sequencing of the corresponding TCR repertoires demonstrated significant clonotypic overlap within individuals, especially between the more differentiated CD4−CD8− and CD4−CD8+ subsets. Collectively, these results mapped a linear differentiation pathway across the post-thymic landscape of human CD1d-restricted NKT cells.
- Published
- 2019
5. Design of Nanoparticulate Group 2 Influenza Virus Hemagglutinin Stem Antigens That Activate Unmutated Ancestor B Cell Receptors of Broadly Neutralizing Antibody Lineages
- Author
-
Subbarao, K, Corbett, KS, Moin, SM, Yassine, HM, Cagigi, A, Kanekiyo, M, Boyoglu-Barnum, S, Myers, SI, Tsybovsky, Y, Wheatley, AK, Schramm, CA, Gillespie, RA, Shi, W, Wang, L, Zhang, Y, Andrews, SF, Joyce, MG, Crank, MC, Douek, DC, McDermott, AB, Mascola, JR, Graham, BS, Boyington, JC, Subbarao, K, Corbett, KS, Moin, SM, Yassine, HM, Cagigi, A, Kanekiyo, M, Boyoglu-Barnum, S, Myers, SI, Tsybovsky, Y, Wheatley, AK, Schramm, CA, Gillespie, RA, Shi, W, Wang, L, Zhang, Y, Andrews, SF, Joyce, MG, Crank, MC, Douek, DC, McDermott, AB, Mascola, JR, Graham, BS, and Boyington, JC
- Abstract
Influenza vaccines targeting the highly conserved stem of the hemagglutinin (HA) surface glycoprotein have the potential to protect against pandemic and drifted seasonal influenza viruses not covered by current vaccines. While HA stem-based immunogens derived from group 1 influenza A viruses have been shown to induce intragroup heterosubtypic protection, HA stem-specific antibody lineages originating from group 2 may be more likely to possess broad cross-group reactivity. We report the structure-guided development of mammalian-cell-expressed candidate vaccine immunogens based on influenza A virus group 2 H3 and H7 HA stem trimers displayed on self-assembling ferritin nanoparticles using an iterative, multipronged approach involving helix stabilization, loop optimization, disulfide bond addition, and side-chain repacking. These immunogens were thermostable, formed uniform and symmetric nanoparticles, were recognized by cross-group-reactive broadly neutralizing antibodies (bNAbs) with nanomolar affinity, and elicited protective, homosubtypic antibodies in mice. Importantly, several immunogens were able to activate B cells expressing inferred unmutated common ancestor (UCA) versions of cross-group-reactive human bNAbs from two multidonor classes, suggesting they could initiate elicitation of these bNAbs in humans.IMPORTANCE Current influenza vaccines are primarily strain specific, requiring annual updates, and offer minimal protection against drifted seasonal or pandemic strains. The highly conserved stem region of hemagglutinin (HA) of group 2 influenza A virus subtypes is a promising target for vaccine elicitation of broad cross-group protection against divergent strains. We used structure-guided protein engineering employing multiple protein stabilization methods simultaneously to develop group 2 HA stem-based candidate influenza A virus immunogens displayed as trimers on self-assembling nanoparticles. Characterization of antigenicity, thermostability, and particle
- Published
- 2019
6. VDJdb: A curated database of T-cell receptor sequences with known antigen specificity
- Author
-
Shugay, M, Bagaev, DV, Zvyagin, IV, Vroomans, RM, Crawford, JC, Dolton, G, Komech, EA, Sycheva, AL, Koneva, AE, Egorov, ES, Eliseev, AV, Van Dyk, E, Dash, P, Attaf, M, Rius, C, Ladell, K, McLaren, JE, Matthews, KK, Clemens, EB, Douek, DC, Luciani, F, Van Baarle, D, Kedzierska, K, Kesmir, C, Thomas, PG, Price, DA, Sewell, AK, Chudakov, DM, Shugay, M, Bagaev, DV, Zvyagin, IV, Vroomans, RM, Crawford, JC, Dolton, G, Komech, EA, Sycheva, AL, Koneva, AE, Egorov, ES, Eliseev, AV, Van Dyk, E, Dash, P, Attaf, M, Rius, C, Ladell, K, McLaren, JE, Matthews, KK, Clemens, EB, Douek, DC, Luciani, F, Van Baarle, D, Kedzierska, K, Kesmir, C, Thomas, PG, Price, DA, Sewell, AK, and Chudakov, DM
- Abstract
© The Author(s) 2017. Published by Oxford University Press on behalf of Nucleic Acids Research. The ability to decode antigen specificities encapsulated in the sequences of rearranged T-cell receptor (TCR) genes is critical for our understanding of the adaptive immune system and promises significant advances in the field of translational medicine. Recent developments in high-throughput sequencing methods (immune repertoire sequencing technology, or RepSeq) and single-cell RNA sequencing technology have allowed us to obtain huge numbers of TCR sequences from donor samples and link them to T-cell phenotypes. However, our ability to annotate these TCR sequences still lags behind, owing to the enormous diversity of the TCR repertoire and the scarcity of available data on T-cell specificities. In this paper, we present VDJdb, a database that stores and aggregates the results of published T-cell specificity assays and provides a universal platform that couples antigen specificities with TCR sequences. We demonstrate that VDJdb is a versatile instrument for the annotation of TCR repertoire data, enabling a concatenated view of antigen-specific TCR sequence motifs. VDJdb can be accessed at https://vdjdb.cdr3.net and https://github.com/antigenomics/vdjdb-db.
- Published
- 2018
7. Reproducibility and reuse of adaptive immune receptor repertoire data
- Author
-
Breden, F, Luning Prak, ET, Peters, B, Rubelt, F, Schramm, CA, Busse, CE, Vander Heiden, JA, Christley, S, Bukhari, SAC, Thorogood, A, Matsen, FA, Wine, Y, Laserson, U, Klatzmann, D, Douek, DC, Lefranc, MP, Collins, AM, Bubela, T, Kleinstein, SH, Watson, CT, Cowell, LG, Scott, JK, Kepler, TB, Breden, F, Luning Prak, ET, Peters, B, Rubelt, F, Schramm, CA, Busse, CE, Vander Heiden, JA, Christley, S, Bukhari, SAC, Thorogood, A, Matsen, FA, Wine, Y, Laserson, U, Klatzmann, D, Douek, DC, Lefranc, MP, Collins, AM, Bubela, T, Kleinstein, SH, Watson, CT, Cowell, LG, Scott, JK, and Kepler, TB
- Abstract
© 2017 Breden, Luning Prak, Peters, Rubelt, Schramm, Busse, Vander Heiden, Christley, Bukhari, Thorogood, Matsen IV, Wine, Laserson, Klatzmann, Douek, Lefranc, Collins, Bubela, Kleinstein, Watson, Cowell, Scott and Kepler. High-throughput sequencing (HTS) of immunoglobulin (B-cell receptor, antibody) and T-cell receptor repertoires has increased dramatically since the technique was introduced in 2009 (1-3). This experimental approach explores the maturation of the adaptive immune system and its response to antigens, pathogens, and disease conditions in exquisite detail. It holds significant promise for diagnostic and therapy-guiding applications. New technology often spreads rapidly, sometimes more rapidly than the understanding of how to make the products of that technology reliable, reproducible, or usable by others. As complex technologies have developed, scientific communities have come together to adopt common standards, protocols, and policies for generating and sharing data sets, such as the MIAME protocols developed for microarray experiments. The Adaptive Immune Receptor Repertoire (AIRR) Community formed in 2015 to address similar issues for HTS data of immune repertoires. The purpose of this perspective is to provide an overview of the AIRR Community's founding principles and present the progress that the AIRR Community has made in developing standards of practice and data sharing protocols. Finally, and most important, we invite all interested parties to join this effort to facilitate sharing and use of these powerful data sets (join@airr-community.org).
- Published
- 2017
8. A Cure for HIV Infection: 'Not in My Lifetime' or 'Just Around the Corner'?
- Author
-
Lederman, MM, Cannon, PM, Currier, JS, June, CH, Kiem, HP, Kuritzkes, DR, Lewin, SR, Margolis, DM, McCune, JM, Mellors, JW, Schacker, TW, Sekaly, RP, Tebas, P, Walker, BD, Douek, DC, Lederman, MM, Cannon, PM, Currier, JS, June, CH, Kiem, HP, Kuritzkes, DR, Lewin, SR, Margolis, DM, McCune, JM, Mellors, JW, Schacker, TW, Sekaly, RP, Tebas, P, Walker, BD, and Douek, DC
- Abstract
With the advent and stunning success of combination antiretroviral therapy (ART) to prolong and improve quality of life for persons with HIV infection, HIV research has been afforded the opportunity to pivot towards studies aimed at finding "a cure." The mere idea that cure of HIV might be possible has energized researchers and the community towards achieving this goal. Funding agencies, both governmental and private, have targeted HIV cure as a high priority; many in the field have responded to these initiatives and the cure research agenda is robust. In this "salon" two editors of Pathogens and Immunity, Michael Lederman and Daniel Douek ask whether curing HIV is a realistic, scalable objective. We start with an overview perspective and have asked a number of prominent HIV researchers to add to the discussion.
- Published
- 2016
9. CD4+ T Cells Expressing PD-1, TIGIT and LAG-3 Contribute to HIV Persistence during ART
- Author
-
Douek, DC, Fromentin, R, Bakeman, W, Lawani, MB, Khoury, G, Hartogensis, W, DaFonseca, S, Killian, M, Epling, L, Hoh, R, Sinclair, E, Hecht, FM, Bacchetti, P, Deeks, SG, Lewin, SR, Sekaly, R-P, Chomont, N, Douek, DC, Fromentin, R, Bakeman, W, Lawani, MB, Khoury, G, Hartogensis, W, DaFonseca, S, Killian, M, Epling, L, Hoh, R, Sinclair, E, Hecht, FM, Bacchetti, P, Deeks, SG, Lewin, SR, Sekaly, R-P, and Chomont, N
- Abstract
HIV persists in a small pool of latently infected cells despite antiretroviral therapy (ART). Identifying cellular markers expressed at the surface of these cells may lead to novel therapeutic strategies to reduce the size of the HIV reservoir. We hypothesized that CD4+ T cells expressing immune checkpoint molecules would be enriched in HIV-infected cells in individuals receiving suppressive ART. Expression levels of 7 immune checkpoint molecules (PD-1, CTLA-4, LAG-3, TIGIT, TIM-3, CD160 and 2B4) as well as 4 markers of HIV persistence (integrated and total HIV DNA, 2-LTR circles and cell-associated unspliced HIV RNA) were measured in PBMCs from 48 virally suppressed individuals. Using negative binomial regression models, we identified PD-1, TIGIT and LAG-3 as immune checkpoint molecules positively associated with the frequency of CD4+ T cells harboring integrated HIV DNA. The frequency of CD4+ T cells co-expressing PD-1, TIGIT and LAG-3 independently predicted the frequency of cells harboring integrated HIV DNA. Quantification of HIV genomes in highly purified cell subsets from blood further revealed that expressions of PD-1, TIGIT and LAG-3 were associated with HIV-infected cells in distinct memory CD4+ T cell subsets. CD4+ T cells co-expressing the three markers were highly enriched for integrated viral genomes (median of 8.2 fold compared to total CD4+ T cells). Importantly, most cells carrying inducible HIV genomes expressed at least one of these markers (median contribution of cells expressing LAG-3, PD-1 or TIGIT to the inducible reservoir = 76%). Our data provide evidence that CD4+ T cells expressing PD-1, TIGIT and LAG-3 alone or in combination are enriched for persistent HIV during ART and suggest that immune checkpoint blockers directed against these receptors may represent valuable tools to target latently infected cells in virally suppressed individuals.
- Published
- 2016
10. Systemic inflammation and liver damage in HIV/hepatitis C virus coinfection
- Author
-
Shmagel, KV, primary, Saidakova, EV, additional, Shmagel, NG, additional, Korolevskaya, LB, additional, Chereshnev, VA, additional, Robinson, J, additional, Grivel, J-C, additional, Douek, DC, additional, Margolis, L, additional, Anthony, DD, additional, and Lederman, MM, additional
- Published
- 2016
- Full Text
- View/download PDF
11. Loss of Circulating CD4 T Cells with B Cell Helper Function during Chronic HIV Infection
- Author
-
Silvestri, G, Boswell, KL, Paris, R, Boritz, E, Ambrozak, D, Yamamoto, T, Darko, S, Wloka, K, Wheatley, A, Narpala, S, McDermott, A, Roederer, M, Haubrich, R, Connors, M, Ake, J, Douek, DC, Kim, J, Petrovas, C, Koup, RA, Silvestri, G, Boswell, KL, Paris, R, Boritz, E, Ambrozak, D, Yamamoto, T, Darko, S, Wloka, K, Wheatley, A, Narpala, S, McDermott, A, Roederer, M, Haubrich, R, Connors, M, Ake, J, Douek, DC, Kim, J, Petrovas, C, and Koup, RA
- Abstract
The interaction between follicular T helper cells (TFH) and B cells in the lymph nodes and spleen has a major impact on the development of antigen-specific B cell responses during infection or vaccination. Recent studies described a functional equivalent of these cells among circulating CD4 T cells, referred to as peripheral TFH cells. Here, we characterize the phenotype and in vitro B cell helper activity of peripheral TFH populations, as well as the effect of HIV infection on these populations. In co-culture experiments we confirmed CXCR5+ cells from HIV-uninfected donors provide help to B cells and more specifically, we identified a CCR7(high)CXCR5(high)CCR6(high)PD-1(high) CD4 T cell population that secretes IL-21 and enhances isotype-switched immunoglobulin production. This population is significantly decreased in treatment-naïve, HIV-infected individuals and can be recovered after anti-retroviral therapy. We found impaired immunoglobulin production in co-cultures from HIV-infected individuals and found no correlation between the frequency of peripheral TFH cells and memory B cells, or with neutralization activity in untreated HIV infection in our cohort. Furthermore, we found that within the peripheral TFH population, the expression level of TFH-associated genes more closely resembles a memory, non-TFH population, as opposed to a TFH population. Overall, our data identify a heterogeneous population of circulating CD4 T cells that provides in vitro help to B cells, and challenges the origin of these cells as memory TFH cells.
- Published
- 2014
12. Protective Efficacy of Cross-Reactive CD8+ T Cells Recognising Mutant Viral Epitopes Depends on Peptide-MHC-I Structural Interactions and T Cell Activation Threshold
- Author
-
Douek, DC, Valkenburg, SA, Gras, S, Guillonneau, C, La Gruta, NL, Thomas, PG, Purcell, AW, Rossjohn, J, Doherty, PC, Turner, SJ, Kedzierska, K, Douek, DC, Valkenburg, SA, Gras, S, Guillonneau, C, La Gruta, NL, Thomas, PG, Purcell, AW, Rossjohn, J, Doherty, PC, Turner, SJ, and Kedzierska, K
- Abstract
Emergence of a new influenza strain leads to a rapid global spread of the virus due to minimal antibody immunity. Pre-existing CD8(+) T-cell immunity directed towards conserved internal viral regions can greatly ameliorate the disease. However, mutational escape within the T cell epitopes is a substantial issue for virus control and vaccine design. Although mutations can result in a loss of T cell recognition, some variants generate cross-reactive T cell responses. In this study, we used reverse genetics to modify the influenza NP(336-374) peptide at a partially-solvent exposed residue (N->A, NPN3A mutation) to assess the availability, effectiveness and mechanism underlying influenza-specific cross-reactive T cell responses. The engineered virus induced a diminished CD8(+) T cell response and selected a narrowed T cell receptor (TCR) repertoire within two V beta regions (V beta 8.3 and V beta 9). This can be partially explained by the H-2D(b)NPN3A structure that showed a loss of several contacts between the NPN3A peptide and H-2D(b), including a contact with His155, a position known to play an important role in mediating TCR-pMHC-I interactions. Despite these differences, common cross-reactive TCRs were detected in both the naïve and immune NPN3A-specific TCR repertoires. However, while the NPN3A epitope primes memory T-cells that give an equivalent recall response to the mutant or wild-type (wt) virus, both are markedly lower than wt->wt challenge. Such decreased CD8(+) responses elicited after heterologous challenge resulted in delayed viral clearance from the infected lung. Furthermore, mice first exposed to the wt virus give a poor, low avidity response following secondary infection with the mutant. Thus, the protective efficacy of cross-reactive CD8(+) T cells recognising mutant viral epitopes depend on peptide-MHC-I structural interactions and functional avidity. Our study does not support vaccine strategies that include immunization against commonly selected
- Published
- 2010
13. Public clonotype usage identifies protective Gag-specific CD8+ T cell responses in SIV infection
- Author
-
Price, DA, Asher, TE, Wilson, NA, Nason, MC, Brenchley, JM, Metzler, IS, Venturi, V, Gostick, E, Chattopadhyay, PK, Roederer, M, Davenport, MP, Watkins, DI, Douek, DC, Price, DA, Asher, TE, Wilson, NA, Nason, MC, Brenchley, JM, Metzler, IS, Venturi, V, Gostick, E, Chattopadhyay, PK, Roederer, M, Davenport, MP, Watkins, DI, and Douek, DC
- Abstract
Despite the pressing need for an AIDS vaccine, the determinants of protective immunity to HIV remain concealed within the complexity of adaptive immune responses. We dissected immunodominant virus-specific CD8+ T cell populations in Mamu-A01+ rhesus macaques with primary SIV infection to elucidate the hallmarks of effective immunity at the level of individual constituent clonotypes, which were identified according to the expression of distinct T cell receptors (TCRs). The number of public clonotypes, defined as those that expressed identical TCR β-chain amino acid sequences and recurred in multiple individuals, contained within the acute phase CD8 + T cell population specific for the biologically constrained Gag CM9 (CTPYDINQM; residues 181-189) epitope correlated negatively with the virus load set point. This independent molecular signature of protection was confirmed in a prospective vaccine trial, in which clonotype engagement was governed by the nature of the antigen rather than the context of exposure and public clonotype usage was associated with enhanced recognition of epitope variants. Thus, the pattern of antigen-specific clonotype recruitment within a protective CD8 + T cell population is a prognostic indicator of vaccine efficacy and biological outcome in an AIDS virus infection.
- Published
- 2009
14. P11-01. Extensive intestinal damage underlies microbial translocation in the GI tract of chronically SIV-infected Rhesus macaques
- Author
-
Harris, LD, primary, Estes, JD, additional, Klatt, NR, additional, Taft, B, additional, Barclay, R, additional, Douek, DC, additional, Silvestri, G, additional, Liffson, J, additional, and Brenchley, J, additional
- Published
- 2009
- Full Text
- View/download PDF
15. Glutamic acid decarboxylase T lymphocyte responses associated with susceptibility or resistance to type I diabetes: analysis in disease discordant human twins, non-obese diabetic mice and HLA-DQ transgenic mice.
- Author
-
Boyton, RJ, Lohmann, T, Londei, M, Kalbacher, H, Halder, T, Frater, AJ, Douek, DC, Leslie, RDG, Flavell, RA, and Altmann, DM
- Abstract
Glutamic acid decarboxylase (GAD65) has been implicated as a targeted self antigen in the immune destruction of pancreatic β cells. T cell responses to GAD65 peptides have been detected in both patients with type I diabetes and in the non-obese diabetic (NOD) mouse. To establish which GAD65 epitopes are important in the immunopathogenesis of disease we initially compared T cell responses to GAD65 epitopes in conditions of disease susceptibility and protection. T cell responses to GAD65 peptides were measured in monozygotic twin pairs selected on the basis of disease discordance and T cell recognition of immunogenic regions of GAD65. Peptides of interest were then used to immunize susceptible NOD mice and H2-E transgenic NOD mice which are protected from diabetes. A differential response to the epitope GAD65 521-535 discriminated diabetic from non-diabetic human twins as well as susceptible from protected mice. This epitope as well as GAD 505-519 induces T cell responses despite binding the type I diabetes associated HLA-DQA1*301/DQB1*0302 product with low affinity. Since DQ-restricted T cell responses are difficult to study in humans, HLA-DQ8 transgenic mice were then used: GAD epitopes 521-535 and 505-519 induced responses in DQ8 transgenic mice and T cells were established. Long-term T cell lines against GAD 505-519 were HLA-DQ restricted, and responded to peptide with a strong IFN-γ and IL-10 response. The findings implicate GAD 521-535 as a possible target peptide in pathogenesis and are compatible with a model whereby self-reactive T cells specific for low-affinity peptide -MHC complexes may escape thymic negative selection. [ABSTRACT FROM PUBLISHER]
- Published
- 1998
- Full Text
- View/download PDF
16. The contribution of the thymus to immune reconstitution after hematopoietic stem-cell transplantation.
- Author
-
Douek, DC
- Subjects
- *
THYMUS , *LYMPHOID tissue , *IMMUNITY , *DRUG therapy , *STEM cell transplantation , *CELL transplantation - Abstract
Discusses research on the contribution of the adult thymus to immune reconstitution after myeloablative chemotherapy and autologous hematopoietic stem cell transplantation. Details of an assay that can be used to estimate thymic output by measuring T-cell receptor rearrangement excision circles; Occurrence of thymopoiesis.
- Published
- 2002
- Full Text
- View/download PDF
17. Determinants of protection among HIV‐exposed seronegative persons: an overview.
- Author
-
Lederman MM, Alter G, Daskalakis DC, Rodriguez B, Sieg SF, Hardy G, Cho M, Anthony D, Harding C, Weinberg A, Silverman RH, Douek DC, Margolis L, Goldstein DB, Carrington M, Goedert JJ, Lederman, Michael M, Alter, Galit, Daskalakis, Demetre C, and Rodriguez, Benigno
- Abstract
Both clinical experience and a growing medical literature indicate that some persons who have been exposed to human immunodeficiency virus (HIV) infection remain uninfected. Although in some instances this may represent good fortune, cohorts of uninfected persons have been reported who are considered at high risk for infection. In these cohorts a variety of characteristics have been proposed as mediating protection, but to date only the 32–base pair deletion in the chemokine (C‐C motif) receptor 5 gene, which results in complete failure of cell surface expression of this coreceptor, has been associated with high‐level protection from HIV infection. With this in mind, there are probably many other factors that may individually or in combination provide some level of protection from acquisition of HIV infection. Because some of these factors are probably incompletely protective or inconsistently active, identifying them with confidence will be difficult. Nonetheless, clarifying the determinants of protection against HIV infection is a high priority that will require careful selection of high‐risk uninfected cohorts, who should undergo targeted studies of plausible mediators and broad screening for unexpected determinants of protection. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
18. Mechanisms of disease: where does HIV live?
- Author
-
Stebbing J, Gazzard B, and Douek DC
- Published
- 2004
19. Temporal Dynamics of CD8+ T Cell Effector Responses during Primary HIV Infection
- Author
-
Leigh Anne Eller, Hannah Kibuuka, Marcus Buggert, Barton F. Haynes, Nicole F. Bernard, Korey Demers, Nilu Goonetilleke, Sarah J. Ratcliffe, Chris Ka-fai Li, Sorachai Nitayaphan, Lucas Maganga, George Makedonas, Mark K. Slifka, Jean-Pierre Routy, Merlin L. Robb, Michael R. Betts, Andrew J. McMichael, Michael A. Eller, Kathleen Rono, and Douek, DC
- Subjects
0301 basic medicine ,RNA viruses ,Male ,HIV Infections ,CD8-Positive T-Lymphocytes ,Pathology and Laboratory Medicine ,Memory T cells ,Interleukin 21 ,White Blood Cells ,Cognition ,Learning and Memory ,Immunodeficiency Viruses ,Animal Cells ,Medicine and Health Sciences ,Cytotoxic T cell ,lcsh:QH301-705.5 ,education.field_of_study ,Immunity, Cellular ,biology ,Effector ,T Cells ,virus diseases ,Viral Load ,Middle Aged ,3. Good health ,medicine.anatomical_structure ,Medical Microbiology ,Viral Pathogens ,Viruses ,Infectious diseases ,Female ,Cellular Types ,Pathogens ,Research Article ,lcsh:Immunologic diseases. Allergy ,Adult ,T cell ,Immune Cells ,Population ,Immunology ,Eomesodermin ,Cytotoxic T cells ,Viral diseases ,Research and Analysis Methods ,Microbiology ,03 medical and health sciences ,Memory ,Virology ,Retroviruses ,Genetics ,medicine ,Humans ,education ,Molecular Biology Techniques ,Microbial Pathogens ,Molecular Biology ,Cell Proliferation ,Blood Cells ,Perforin ,Lentivirus ,Organisms ,Biology and Life Sciences ,HIV ,Cell Biology ,030104 developmental biology ,lcsh:Biology (General) ,Chronic Disease ,biology.protein ,Cognitive Science ,Parasitology ,lcsh:RC581-607 ,T-Box Domain Proteins ,CD8 ,Viral Transmission and Infection ,Neuroscience ,Cloning - Abstract
The loss of HIV-specific CD8+ T cell cytolytic function is a primary factor underlying progressive HIV infection, but whether HIV-specific CD8+ T cells initially possess cytolytic effector capacity, and when and why this may be lost during infection, is unclear. Here, we assessed CD8+ T cell functional evolution from primary to chronic HIV infection. We observed a profound expansion of perforin+ CD8+ T cells immediately following HIV infection that quickly waned after acute viremia resolution. Selective expression of the effector-associated transcription factors T-bet and eomesodermin in cytokine-producing HIV-specific CD8+ T cells differentiated HIV-specific from bulk memory CD8+ T cell effector expansion. As infection progressed expression of perforin was maintained in HIV-specific CD8+ T cells with high levels of T-bet, but not necessarily in the population of T-betLo HIV-specific CD8+ T cells that expand as infection progresses. Together, these data demonstrate that while HIV-specific CD8+ T cells in acute HIV infection initially possess cytolytic potential, progressive transcriptional dysregulation leads to the reduced CD8+ T cell perforin expression characteristic of chronic HIV infection., Author Summary Previous studies have demonstrated that HIV-specific CD8+ T cells are critical for the initial control of HIV infection. However, this control is typically incomplete, being able to neither clear infection nor maintain plasma viremia below undetectable levels. Mounting evidence has implicated CD8+ T cell cytotoxic capacity as a critical component of the HIV-specific response associated with spontaneous long-term control of HIV replication. CD8+ T cell cytotoxic responses are largely absent in the vast majority of HIV chronically infected individuals and it is unclear when or why this functionality is lost. In this study we show that HIV-specific CD8+ T cells readily express the cytolytic protein perforin during the acute phase of chronic progressive HIV infection but rapidly lose the ability to upregulate this molecule following resolution of peak viremia. Maintenance of perforin expression by HIV-specific CD8+ T cells appears to be associated with the expression level of the transcription factor T-bet, but not with the T-bet paralogue, Eomes. These findings further delineate qualitative attributes of CD8+ T cell-mediated immunity that may serve as targets for future HIV vaccine and therapeutic research.
- Published
- 2016
20. FIND-seq: high-throughput nucleic acid cytometry for rare single-cell transcriptomics.
- Author
-
Shin SW, Mudvari P, Thaploo S, Wheeler MA, Douek DC, Quintana FJ, Boritz EA, Abate AR, and Clark IC
- Subjects
- Humans, High-Throughput Nucleotide Sequencing methods, Transcriptome, Gene Expression Profiling methods, Flow Cytometry methods, Single-Cell Analysis methods
- Abstract
Rare cells have an important role in development and disease, and methods for isolating and studying cell subsets are therefore an essential part of biology research. Such methods traditionally rely on labeled antibodies targeted to cell surface proteins, but large public databases and sophisticated computational approaches increasingly define cell subsets on the basis of genomic, epigenomic and transcriptomic sequencing data. Methods for isolating cells on the basis of nucleic acid sequences powerfully complement these approaches by providing experimental access to cell subsets discovered in cell atlases, as well as those that cannot be otherwise isolated, including cells infected with pathogens, with specific DNA mutations or with unique transcriptional or splicing signatures. We recently developed a nucleic acid cytometry platform called 'focused interrogation of cells by nucleic acid detection and sequencing' (FIND-seq), capable of isolating rare cells on the basis of RNA or DNA markers, followed by bulk or single-cell transcriptomic analysis. This platform has previously been used to characterize the splicing-dependent activation of the transcription factor XBP1 in astrocytes and HIV persistence in memory CD4 T cells from people on long-term antiretroviral therapy. Here, we outline the molecular and microfluidic steps involved in performing FIND-seq, including protocol updates that allow detection and whole transcriptome sequencing of rare HIV-infected cells that harbor genetically intact virus genomes. FIND-seq requires knowledge of microfluidics, optics and molecular biology. We expect that FIND-seq, and this comprehensive protocol, will enable mechanistic studies of rare HIV
+ cells, as well as other cell subsets that were previously difficult to recover and sequence., (© 2024. Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
21. Potent and broad HIV-1 neutralization in fusion peptide-primed SHIV-infected macaques.
- Author
-
Wang H, Cheng C, Dal Santo JL, Shen CH, Bylund T, Henry AR, Howe CA, Hwang J, Morano NC, Morris DJ, Pletnev S, Roark RS, Zhou T, Hansen BT, Hoyt FH, Johnston TS, Wang S, Zhang B, Ambrozak DR, Becker JE, Bender MF, Changela A, Chaudhary R, Corcoran M, Corrigan AR, Foulds KE, Guo Y, Lee M, Li Y, Lin BC, Liu T, Louder MK, Mandolesi M, Mason RD, McKee K, Nair V, O'Dell S, Olia AS, Ou L, Pegu A, Raju N, Rawi R, Roberts-Torres J, Sarfo EK, Sastry M, Schaub AJ, Schmidt SD, Schramm CA, Schwartz CL, Smith SC, Stephens T, Stuckey J, Teng IT, Todd JP, Tsybovsky Y, Van Wazer DJ, Wang S, Doria-Rose NA, Fischer ER, Georgiev IS, Karlsson Hedestam GB, Sheng Z, Woodward RA, Douek DC, Koup RA, Pierson TC, Shapiro L, Shaw GM, Mascola JR, and Kwong PD
- Abstract
An antibody-based HIV-1 vaccine will require the induction of potent cross-reactive HIV-1-neutralizing responses. To demonstrate feasibility toward this goal, we combined vaccination targeting the fusion-peptide site of vulnerability with infection by simian-human immunodeficiency virus (SHIV). In four macaques with vaccine-induced neutralizing responses, SHIV infection boosted plasma neutralization to 45%-77% breadth (geometric mean 50% inhibitory dilution [ID
50 ] ∼100) on a 208-strain panel. Molecular dissection of these responses by antibody isolation and cryo-electron microscopy (cryo-EM) structure determination revealed 15 of 16 antibody lineages with cross-clade neutralization to be directed toward the fusion-peptide site of vulnerability. In each macaque, isolated antibodies from memory B cells recapitulated the plasma-neutralizing response, with fusion-peptide-binding antibodies reaching breadths of 40%-60% (50% inhibitory concentration [IC50 ] < 50 μg/mL) and total lineage-concentrations estimates of 50-200 μg/mL. Longitudinal mapping indicated that these responses arose prior to SHIV infection. Collectively, these results provide in vivo molecular examples for one to a few B cell lineages affording potent, broadly neutralizing plasma responses., Competing Interests: Declaration of interests The authors declare no competing interests., (Published by Elsevier Inc.)- Published
- 2024
- Full Text
- View/download PDF
22. A multispecific antibody against SARS-CoV-2 prevents immune escape in vitro and confers prophylactic protection in vivo.
- Author
-
Misasi J, Wei RR, Wang L, Pegu A, Wei CJ, Oloniniyi OK, Zhou T, Moliva JI, Zhao B, Choe M, Yang ES, Zhang Y, Boruszczak M, Chen M, Leung K, Li J, Yang ZY, Andersen H, Carlton K, Godbole S, Harris DR, Henry AR, Ivleva VB, Lei QP, Liu C, Longobardi L, Merriam JS, Nase D, Olia AS, Pessaint L, Porto M, Shi W, Wallace SM, Wolff JJ, Douek DC, Suthar MS, Gall JG, Koup RA, Kwong PD, Mascola JR, Nabel GJ, and Sullivan NJ
- Subjects
- Animals, Humans, Mice, Epitopes immunology, Mesocricetus, Cricetinae, Antibodies, Bispecific immunology, Antibodies, Bispecific pharmacology, SARS-CoV-2 immunology, COVID-19 immunology, COVID-19 prevention & control, COVID-19 virology, Antibodies, Neutralizing immunology, Antibodies, Neutralizing therapeutic use, Spike Glycoprotein, Coronavirus immunology, Spike Glycoprotein, Coronavirus chemistry, Antibodies, Viral immunology, Immune Evasion
- Abstract
Despite effective countermeasures, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) persists worldwide because of its ability to diversify and evade human immunity. This evasion stems from amino acid substitutions, particularly in the receptor binding domain (RBD) of the spike protein that confers resistance to vaccine-induced antibodies and antibody therapeutics. To constrain viral escape through resistance mutations, we combined antibody variable regions that recognize different RBD sites into multispecific antibodies. Here, we describe multispecific antibodies, including a trivalent trispecific antibody that potently neutralized diverse SARS-CoV-2 variants and prevented virus escape more effectively than single antibodies or mixtures of the parental antibodies. Despite being generated before the appearance of Omicron, this trispecific antibody neutralized all major Omicron variants through BA.4/BA.5 at nanomolar concentrations. Negative stain electron microscopy suggested that synergistic neutralization was achieved by engaging different epitopes in specific orientations that facilitated binding across more than one spike protein. Moreover, a tetravalent trispecific antibody containing the same variable regions as the trivalent trispecific antibody also protected Syrian hamsters against Omicron variants BA.1, BA.2, and BA.5 challenge, each of which uses different amino acid substitutions to mediate escape from therapeutic antibodies. These results demonstrated that multispecific antibodies have the potential to provide broad SARS-CoV-2 coverage, decrease the likelihood of escape, simplify treatment, and provide a strategy for antibody therapies that could help eliminate pandemic spread for this and other pathogens.
- Published
- 2024
- Full Text
- View/download PDF
23. Mucosal adenovirus vaccine boosting elicits IgA and durably prevents XBB.1.16 infection in nonhuman primates.
- Author
-
Gagne M, Flynn BJ, Andrew SF, Marquez J, Flebbe DR, Mychalowych A, Lamb E, Davis-Gardner ME, Burnett MR, Serebryannyy LA, Lin BC, Ziff ZE, Maule E, Carroll R, Naisan M, Jethmalani Y, Pessaint L, Todd JM, Doria-Rose NA, Case JB, Dmitriev IP, Kashentseva EA, Ying B, Dodson A, Kouneski K, O'Dell S, Wali B, Ellis M, Godbole S, Laboune F, Henry AR, Teng IT, Wang D, Wang L, Zhou Q, Zouantchangadou S, Van Ry A, Lewis MG, Andersen H, Kwong PD, Curiel DT, Roederer M, Nason MC, Foulds KE, Suthar MS, Diamond MS, Douek DC, and Seder RA
- Subjects
- Animals, Spike Glycoprotein, Coronavirus immunology, Spike Glycoprotein, Coronavirus genetics, Macaca mulatta, Adenoviridae immunology, Adenoviridae genetics, Immunity, Mucosal, Adenovirus Vaccines immunology, Adenovirus Vaccines administration & dosage, Female, Lung virology, Lung immunology, B-Lymphocytes immunology, Immunoglobulin G immunology, Immunoglobulin G blood, Antibodies, Neutralizing immunology, Antibodies, Neutralizing blood, Administration, Intranasal, Vaccination methods, Humans, Immunoglobulin A immunology, SARS-CoV-2 immunology, COVID-19 prevention & control, COVID-19 immunology, COVID-19 virology, Antibodies, Viral immunology, Antibodies, Viral blood, Immunization, Secondary, COVID-19 Vaccines immunology, COVID-19 Vaccines administration & dosage
- Abstract
A mucosal route of vaccination could prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication at the site of infection and limit transmission. We compared protection against heterologous XBB.1.16 challenge in nonhuman primates (NHPs) ~5 months following intramuscular boosting with bivalent mRNA encoding WA1 and BA.5 spike proteins or mucosal boosting with a WA1-BA.5 bivalent chimpanzee adenoviral-vectored vaccine delivered by intranasal or aerosol device. NHPs boosted by either mucosal route had minimal virus replication in the nose and lungs, respectively. By contrast, protection by intramuscular mRNA was limited to the lower airways. The mucosally delivered vaccine elicited durable airway IgG and IgA responses and, unlike the intramuscular mRNA vaccine, induced spike-specific B cells in the lungs. IgG, IgA and T cell responses correlated with protection in the lungs, whereas mucosal IgA alone correlated with upper airway protection. This study highlights differential mucosal and serum correlates of protection and how mucosal vaccines can durably prevent infection against SARS-CoV-2., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
24. Clonal succession after prolonged antiretroviral therapy rejuvenates CD8 + T cell responses against HIV-1.
- Author
-
White E, Papagno L, Samri A, Sugata K, Hejblum B, Henry AR, Rogan DC, Darko S, Recordon-Pinson P, Dudoit Y, Llewellyn-Lacey S, Chakrabarti LA, Buseyne F, Migueles SA, Price DA, Andreola MA, Satou Y, Thiebaut R, Katlama C, Autran B, Douek DC, and Appay V
- Subjects
- Humans, Male, Middle Aged, Female, Antiretroviral Therapy, Highly Active, Anti-Retroviral Agents therapeutic use, Single-Cell Analysis, Cell Differentiation immunology, CD8-Positive T-Lymphocytes immunology, HIV-1 immunology, HIV-1 physiology, HIV Infections drug therapy, HIV Infections immunology, HIV Infections virology, Virus Replication drug effects
- Abstract
Human immunodeficiency virus 1 (HIV-1) infection is characterized by a dynamic and persistent state of viral replication that overwhelms the host immune system in the absence of antiretroviral therapy (ART). The impact of prolonged treatment on the antiviral efficacy of HIV-1-specific CD8
+ T cells has nonetheless remained unknown. Here, we used single-cell technologies to address this issue in a cohort of aging individuals infected early during the pandemic and subsequently treated with continuous ART. Our data showed that long-term ART was associated with a process of clonal succession, which effectively rejuvenated HIV-1-specific CD8+ T cell populations in the face of immune senescence. Tracking individual transcriptomes further revealed that initially dominant CD8+ T cell clonotypes displayed signatures of exhaustion and terminal differentiation, whereas newly dominant CD8+ T cell clonotypes displayed signatures of early differentiation and stemness associated with natural control of viral replication. These findings reveal a degree of immune resilience that could inform adjunctive treatments for HIV-1., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
25. Administration of anti-HIV-1 broadly neutralizing monoclonal antibodies with increased affinity to Fcγ receptors during acute SHIV AD8-EO infection.
- Author
-
Dias J, Fabozzi G, Fourati S, Chen X, Liu C, Ambrozak DR, Ransier A, Laboune F, Hu J, Shi W, March K, Maximova AA, Schmidt SD, Samsel J, Talana CA, Ernste K, Ko SH, Lucas ME, Radecki PE, Boswell KL, Nishimura Y, Todd JP, Martin MA, Petrovas C, Boritz EA, Doria-Rose NA, Douek DC, Sékaly RP, Lifson JD, Asokan M, Gama L, Mascola JR, Pegu A, and Koup RA
- Subjects
- Animals, Antibodies, Monoclonal immunology, Lymph Nodes immunology, CD8-Positive T-Lymphocytes immunology, Antibody Affinity immunology, NF-kappa B metabolism, NF-kappa B immunology, Humans, HIV Infections immunology, HIV Infections virology, Killer Cells, Natural immunology, Broadly Neutralizing Antibodies immunology, Receptors, IgG immunology, Receptors, IgG metabolism, Simian Acquired Immunodeficiency Syndrome immunology, Simian Acquired Immunodeficiency Syndrome virology, HIV-1 immunology, Simian Immunodeficiency Virus immunology, Macaca mulatta, Antibodies, Neutralizing immunology, HIV Antibodies immunology
- Abstract
Anti-HIV-1 broadly neutralizing antibodies (bNAbs) have the dual potential of mediating virus neutralization and antiviral effector functions through their Fab and Fc domains, respectively. So far, bNAbs with enhanced Fc effector functions in vitro have only been tested in NHPs during chronic simian-HIV (SHIV) infection. Here, we investigate the effects of administering in acute SHIV
AD8-EO infection either wild-type (WT) bNAbs or bNAbs carrying the S239D/I332E/A330L (DEL) mutation, which increases binding to FcγRs. Emergence of virus in plasma and lymph nodes (LNs) was delayed by bNAb treatment and occurred earlier in monkeys given DEL bNAbs than in those given WT bNAbs, consistent with faster clearance of DEL bNAbs from plasma. DEL bNAb-treated monkeys had higher levels of circulating virus-specific IFNγ single-producing CD8+ CD69+ T cells than the other groups. In LNs, WT bNAbs were evenly distributed between follicular and extrafollicular areas, but DEL bNAbs predominated in the latter. At week 8 post-challenge, LN monocytes and NK cells from DEL bNAb-treated monkeys upregulated proinflammatory signaling pathways and LN T cells downregulated TNF signaling via NF-κB. Overall, bNAbs with increased affinity to FcγRs shape innate and adaptive cellular immunity, which may be important to consider in future strategies of passive bNAb therapy., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2024
- Full Text
- View/download PDF
26. Recruitment and retention of pediatric participants for pandemic preparedness research: Experience from the PREMISE EV-D68 Pilot Study.
- Author
-
Nguyen-Tran H, Spaulding AB, Messacar K, Vogt MR, Permaul P, Douek DC, Mittelman A, Thompson C, Grubbs S, Magana C, and Hernandez ML
- Abstract
Recruitment and retention are challenges for prospective pediatric cohort studies, particularly those involving serial venipunctures. We investigated factors underlying enrollment and retention in the Pandemic Response Repository through Microbial and Immune Surveillance and Epidemiology (PREMISE) Enterovirus D68 (EV-D68) Pilot Study, a multicenter prospective longitudinal cohort study assessing the utility of immunologic surveillance for pandemic preparedness. This study enrolls children ≤10 years for two blood draws, pre- and post-EV-D68 season, separated by 6-18 months. Overall, 174 children were enrolled in Cohort 1 of the study and 120 (69 %) of children completed the study, with follow-up blood samples obtained from 101 (58 %) of participants. Families were primarily motivated to participate by a desire to help other children, advance science, and better prepare for the next pandemic. Adding research blood draws to clinically indicated blood draws improved enrollment, and multiple study touch points facilitated retention. These findings can be applied to improve recruitment and retention in future pandemic preparedness efforts and longitudinal pediatric cohort studies., Competing Interests: The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: HN-T received funding from the Centers for Disease Control and Prevention for her involvement in the “Duration of Enterovirus D68 Shedding from the Upper Respiratory Tract” study and received support from the Congenital and Perinatal Infections Consortium (CPIC) and Cambridge Innovation Institute for conference attendance at IDWeek (2022, 2023) and Next Generation Dx Summit (2023), respectively. AS and AM receive support from the NIH, NIAID Vaccine Research Center. MRV receives support from the following grants/contracts: K08AI156125, 75N91019D00024, R01AI169461; received honorariums for lectures from NIAID, Uniformed Services University, University of Maryland-Baltimore, Duke University, Baylor College of Medicine; and has a patent application pending (PCT/US2020/043,415). MLH receives support from the NIH, NIAID Vaccine Research Center; reports the following grants: NHLBI-R01, NCATS-CTSA K12; NCATS-UM1; NSF-SCH; participates in the GSK Advisory Board for asthma; and is the AAAAI EORD Interest Section Vice Chair., (© 2024 The Authors. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
27. Variant-proof high affinity ACE2 antagonist limits SARS-CoV-2 replication in upper and lower airways.
- Author
-
Gagne M, Flynn BJ, Honeycutt CC, Flebbe DR, Andrew SF, Provost SJ, McCormick L, Van Ry A, McCarthy E, Todd JM, Bao S, Teng IT, Marciano S, Rudich Y, Li C, Jain S, Wali B, Pessaint L, Dodson A, Cook A, Lewis MG, Andersen H, Zahradník J, Suthar MS, Nason MC, Foulds KE, Kwong PD, Roederer M, Schreiber G, Seder RA, and Douek DC
- Subjects
- Animals, Humans, Male, Chlorocebus aethiops, COVID-19 Drug Treatment, Disease Models, Animal, Macaca mulatta, Spike Glycoprotein, Coronavirus metabolism, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Vero Cells, Angiotensin-Converting Enzyme 2 antagonists & inhibitors, Antiviral Agents pharmacology, COVID-19 virology, COVID-19 immunology, COVID-19 prevention & control, SARS-CoV-2 drug effects, SARS-CoV-2 physiology, SARS-CoV-2 immunology, Virus Replication drug effects
- Abstract
SARS-CoV-2 has the capacity to evolve mutations that escape vaccine- and infection-acquired immunity and antiviral drugs. A variant-agnostic therapeutic agent that protects against severe disease without putting selective pressure on the virus would thus be a valuable biomedical tool that would maintain its efficacy despite the ongoing emergence of new variants. Here, we challenge male rhesus macaques with SARS-CoV-2 Delta-the most pathogenic variant in a highly susceptible animal model. At the time of challenge, we also treat the macaques with aerosolized RBD-62, a protein developed through multiple rounds of in vitro evolution of SARS-CoV-2 RBD to acquire 1000-fold enhanced ACE2 binding affinity. RBD-62 treatment equivalently suppresses virus replication in both upper and lower airways, a phenomenon not previously observed with clinically approved vaccines. Importantly, RBD-62 does not block the development of virus-specific T- and B-cell responses and does not elicit anti-drug immunity. These data provide proof-of-concept that RBD-62 can prevent severe disease from a highly virulent variant., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
28. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques.
- Author
-
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore I, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, and Bosinger SE
- Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n=6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines., Competing Interests: Conflicting Interests: RA, TFR, and DRB are listed as inventors on pending patent applications describing the SARS-CoV-2 and HCoV-HKU1 S cross-reactive antibodies. DRB and RA are listed as inventors on a pending patent application describing the S2 stem epitope immunogens identified in this study. DRB is a consultant for IAVI. All other authors declare that they have no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
29. Immunotoxin-mediated depletion of Gag-specific CD8+ T cells undermines natural control of SIV.
- Author
-
Simpson J, Starke CE, Ortiz AM, Ransier A, Darko S, Llewellyn-Lacey S, Fennessey CM, Keele BF, Douek DC, Price DA, and Brenchley JM
- Subjects
- Animals, Gene Products, gag immunology, Virus Replication immunology, Virus Replication drug effects, Lymphocyte Depletion methods, CD8-Positive T-Lymphocytes immunology, Simian Immunodeficiency Virus immunology, Simian Acquired Immunodeficiency Syndrome immunology, Macaca mulatta, Immunotoxins immunology, Immunotoxins pharmacology
- Abstract
Antibody-mediated depletion studies have demonstrated that CD8+ T cells are required for effective immune control of SIV. However, this approach is potentially confounded by several factors, including reactive CD4+ T cell proliferation, and provides no information on epitope specificity, a likely determinant of CD8+ T cell efficacy. We circumvented these limitations by selectively depleting CD8+ T cells specific for the Gag epitope CTPYDINQM (CM9) via the administration of immunotoxin-conjugated tetrameric complexes of CM9/Mamu-A*01. Immunotoxin administration effectively depleted circulating but not tissue-localized CM9-specific CD8+ T cells, akin to the bulk depletion pattern observed with antibodies directed against CD8. However, we found no evidence to indicate that circulating CM9-specific CD8+ T cells suppressed viral replication in Mamu-A*01+ rhesus macaques during acute or chronic progressive infection with a pathogenic strain of SIV. This observation extended to macaques with established infection during and after continuous antiretroviral therapy. In contrast, natural controller macaques experienced dramatic increases in plasma viremia after immunotoxin administration, highlighting the importance of CD8+ T cell-mediated immunity against CM9. Collectively, these data showed that CM9-specific CD8+ T cells were necessary but not sufficient for robust immune control of SIV in a nonhuman primate model and, more generally, validated an approach that could inform the design of next-generation vaccines against HIV-1.
- Published
- 2024
- Full Text
- View/download PDF
30. Imprinting of serum neutralizing antibodies by Wuhan-1 mRNA vaccines.
- Author
-
Liang CY, Raju S, Liu Z, Li Y, Asthagiri Arunkumar G, Case JB, Scheaffer SM, Zost SJ, Acreman CM, Gagne M, Andrew SF, Carvalho Dos Anjos DC, Foulds KE, McLellan JS, Crowe JE Jr, Douek DC, Whelan SPJ, Elbashir SM, Edwards DK, and Diamond MS
- Subjects
- Adult, Animals, Female, Humans, Male, Mice, 2019-nCoV Vaccine mRNA-1273 administration & dosage, 2019-nCoV Vaccine mRNA-1273 immunology, China, Cross Reactions immunology, Epitopes, B-Lymphocyte immunology, Spike Glycoprotein, Coronavirus immunology, Spike Glycoprotein, Coronavirus genetics, Vaccination, Antibodies, Neutralizing immunology, Antibodies, Neutralizing blood, Antibodies, Viral immunology, Antibodies, Viral blood, COVID-19 immunology, COVID-19 prevention & control, COVID-19 virology, COVID-19 Vaccines administration & dosage, COVID-19 Vaccines genetics, COVID-19 Vaccines immunology, Immunization, Secondary, mRNA Vaccines administration & dosage, mRNA Vaccines genetics, mRNA Vaccines immunology, SARS-CoV-2 classification, SARS-CoV-2 genetics, SARS-CoV-2 immunology
- Abstract
Immune imprinting is a phenomenon in which prior antigenic experiences influence responses to subsequent infection or vaccination
1,2 . The effects of immune imprinting on serum antibody responses after boosting with variant-matched SARS-CoV-2 vaccines remain uncertain. Here we characterized the serum antibody responses after mRNA vaccine boosting of mice and human clinical trial participants. In mice, a single dose of a preclinical version of mRNA-1273 vaccine encoding Wuhan-1 spike protein minimally imprinted serum responses elicited by Omicron boosters, enabling generation of type-specific antibodies. However, imprinting was observed in mice receiving an Omicron booster after two priming doses of mRNA-1273, an effect that was mitigated by a second booster dose of Omicron vaccine. In both SARS-CoV-2-infected and uninfected humans who received two Omicron-matched boosters after two or more doses of the prototype mRNA-1273 vaccine, spike-binding and neutralizing serum antibodies cross-reacted with Omicron variants as well as more distantly related sarbecoviruses. Because serum neutralizing responses against Omicron strains and other sarbecoviruses were abrogated after pre-clearing with Wuhan-1 spike protein, antibodies induced by XBB.1.5 boosting in humans focus on conserved epitopes targeted by the antecedent mRNA-1273 primary series. Thus, the antibody response to Omicron-based boosters in humans is imprinted by immunizations with historical mRNA-1273 vaccines, but this outcome may be beneficial as it drives expansion of cross-neutralizing antibodies that inhibit infection of emerging SARS-CoV-2 variants and distantly related sarbecoviruses., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
31. The Gut and the Translocated Microbiomes in HIV Infection: Current Concepts and Future Avenues.
- Author
-
Nganou-Makamdop K and Douek DC
- Abstract
It is widely acknowledged that HIV infection results in disruption of the gut's mucosal integrity partly due a profound loss of gastrointestinal CD4
+ T cells that are targets of the virus. In addition, systemic inflammation and immune activation that drive disease pathogenesis are reduced but not normalized by antiretroviral therapy (ART). It has long been postulated that through the process of microbial translocation, the gut microbiome acts as a key driver of systemic inflammation and immune recovery in HIV infection. As such, many studies have aimed at characterizing the gut microbiota in order to unravel its influence in people with HIV and have reported an association between various bacterial taxa and inflammation. This review assesses both contra-dictory and consistent findings among several studies in order to clarify the overall mechanisms by which the gut microbiota in adults may influence immune recovery in HIV infection. Independently of the gut microbiome, observations made from analysis of microbial products in the blood provide direct insight into how the translocated microbiome may drive immune recovery. To help better understand strengths and limitations of the findings reported, this review also highlights the numerous factors that can influence microbiome studies, be they experimental methodologies, and host-intrinsic or host-extrinsic factors. Altogether, a fuller understanding of the interplay between the gut microbiome and immunity in HIV infection may contribute to preventive and therapeutic approaches., Competing Interests: Competing interests: The authors declare that they have no competing interests., (Copyright © 2024 Pathogens and Immunity.)- Published
- 2024
- Full Text
- View/download PDF
32. Comparative analysis of SARS-CoV-2 neutralization titers reveals consistency between human and animal model serum and across assays.
- Author
-
Mühlemann B, Wilks SH, Baracco L, Bekliz M, Carreño JM, Corman VM, Davis-Gardner ME, Dejnirattisai W, Diamond MS, Douek DC, Drosten C, Eckerle I, Edara VV, Ellis M, Fouchier RAM, Frieman M, Godbole S, Haagmans B, Halfmann PJ, Henry AR, Jones TC, Katzelnick LC, Kawaoka Y, Kimpel J, Krammer F, Lai L, Liu C, Lusvarghi S, Meyer B, Mongkolsapaya J, Montefiori DC, Mykytyn A, Netzl A, Pollett S, Rössler A, Screaton GR, Shen X, Sigal A, Simon V, Subramanian R, Supasa P, Suthar MS, Türeli S, Wang W, Weiss CD, and Smith DJ
- Subjects
- Animals, Humans, Mice, Cricetinae, Disease Models, Animal, SARS-CoV-2 immunology, COVID-19 immunology, COVID-19 blood, COVID-19 virology, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Neutralization Tests, Antibodies, Viral blood, Antibodies, Viral immunology
- Abstract
The evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires ongoing monitoring to judge the ability of newly arising variants to escape the immune response. A surveillance system necessitates an understanding of differences in neutralization titers measured in different assays and using human and animal serum samples. We compared 18 datasets generated using human, hamster, and mouse serum and six different neutralization assays. Datasets using animal model serum samples showed higher titer magnitudes than datasets using human serum samples in this comparison. Fold change in neutralization of variants compared to ancestral SARS-CoV-2, immunodominance patterns, and antigenic maps were similar among serum samples and assays. Most assays yielded consistent results, except for differences in fold change in cytopathic effect assays. Hamster serum samples were a consistent surrogate for human first-infection serum samples. These results inform the transition of surveillance of SARS-CoV-2 antigenic variation from dependence on human first-infection serum samples to the utilization of serum samples from animal models.
- Published
- 2024
- Full Text
- View/download PDF
33. Harnessing the power of IFN for therapeutic approaches to COVID-19.
- Author
-
Viox EG, Bosinger SE, Douek DC, Schreiber G, and Paiardini M
- Subjects
- Animals, Humans, Virus Replication drug effects, Antiviral Agents therapeutic use, COVID-19 immunology, COVID-19 virology, COVID-19 therapy, COVID-19 Drug Treatment, Interferons therapeutic use, Interferons immunology, SARS-CoV-2 drug effects, SARS-CoV-2 immunology
- Abstract
Interferons (IFNs) are essential for defense against viral infections but also drive recruitment of inflammatory cells to sites of infection, a key feature of severe COVID-19. Here, we explore the complexity of the IFN response in COVID-19, examine the effects of manipulating IFN on SARS-CoV-2 viral replication and pathogenesis, and highlight pre-clinical and clinical studies evaluating the therapeutic efficacy of IFN in limiting COVID-19 severity., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
34. A family of olfactory receptors uniquely expanded in marsupial and monotreme genomes are expressed by a T cell subset also unique to marsupials and monotremes.
- Author
-
Sampson JM, Morrissey KA, Douek DC, and Miller RD
- Subjects
- Female, Pregnancy, Animals, Placenta, Genome genetics, Mammals genetics, T-Lymphocyte Subsets, Marsupialia genetics, Receptors, Odorant genetics
- Abstract
Olfactory receptors (OR), expressed on olfactory neurons, mediate the sense of smell. Recently, OR have also been shown to be expressed in non-olfactory tissues, including cells of the immune system. An analysis of single-cell transcriptomes of splenocytes of the grey short-tailed opossum (Monodelphis domestica) found OR are expressed on a subset of T cells, the γμ T cells, that are unique to marsupials and monotremes. A majority of opossum γμ T cells transcriptomes contain OR family 14 transcripts, specifically, from the OR14C subfamily. Amongst the mammals, the OR14 gene family is expanded in the genomes of marsupials and monotremes, and rarer or absent in placental mammals. In summary, here we demonstrate the intriguing correlation that a family of OR genes, abundant in the genomes of marsupials and monotremes, are ectopically expressed in a particular subset of T cells unique to the marsupials and monotremes., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
35. Immunological imprinting shapes the specificity of human antibody responses against SARS-CoV-2 variants.
- Author
-
Johnston TS, Li SH, Painter MM, Atkinson RK, Douek NR, Reeg DB, Douek DC, Wherry EJ, and Hensley SE
- Subjects
- Humans, Antibody Formation, Antibodies, Epitopes, Antibodies, Neutralizing, Antibodies, Viral, SARS-CoV-2, COVID-19
- Abstract
The spike glycoprotein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) continues to accumulate substitutions, leading to breakthrough infections of vaccinated individuals. It remains unclear if exposures to antigenically distant SARS-CoV-2 variants can overcome memory B cell biases established by initial SARS-CoV-2 encounters. We determined the specificity and functionality of antibody and B cell responses following exposure to BA.5 and XBB variants in individuals who received ancestral SARS-CoV-2 mRNA vaccines. BA.5 exposures elicited antibody responses that targeted epitopes conserved between the BA.5 and ancestral spike. XBB exposures also elicited antibody responses that primarily targeted epitopes conserved between the XBB.1.5 and ancestral spike. However, unlike BA.5, a single XBB exposure elicited low frequencies of XBB.1.5-specific antibodies and B cells in some individuals. Pre-existing cross-reactive B cells and antibodies were correlated with stronger overall responses to XBB but weaker XBB-specific responses, suggesting that baseline immunity influences the activation of variant-specific SARS-CoV-2 responses., Competing Interests: Declaration of interests E.J.W. is a member of the Parker Institute for Cancer Immunotherapy. E.J.W. is an advisor for Arsenal Biosciences, Coherus, Danger Bio, IpiNovyx, Janssen, New Limit, Marengo, Pluto Immunotherapeutics Related Sciences, Santa Ana Bio, and Synthekine. E.J.W. is a founder of and holds stock in Coherus, Danger Bio, and Arsenal Biosciences. S.E.H. reports receiving consulting fees from Sanofi Vaccines, Lumen, Novavax, and Merck. S.E.H. is a co-inventor on patents that describe the use of nucleoside-modified mRNA as a vaccine platform., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
36. Predicting the impact of COVID-19 non-pharmaceutical intervention on short- and medium-term dynamics of enterovirus D68 in the US.
- Author
-
Park SW, Messacar K, Douek DC, Spaulding AB, Metcalf CJE, and Grenfell BT
- Subjects
- Humans, Disease Outbreaks, Enterovirus D, Human, COVID-19 epidemiology, Neuromuscular Diseases epidemiology, Myelitis epidemiology, Enterovirus Infections epidemiology, Enterovirus Infections prevention & control, Central Nervous System Viral Diseases
- Abstract
Recent outbreaks of enterovirus D68 (EV-D68) infections, and their causal linkage with acute flaccid myelitis (AFM), continue to pose a serious public health concern. During 2020 and 2021, the dynamics of EV-D68 and other pathogens have been significantly perturbed by non-pharmaceutical interventions against COVID-19; this perturbation presents a powerful natural experiment for exploring the dynamics of these endemic infections. In this study, we analyzed publicly available data on EV-D68 infections, originally collected through the New Vaccine Surveillance Network, to predict their short- and long-term dynamics following the COVID-19 interventions. Although long-term predictions are sensitive to our assumptions about underlying dynamics and changes in contact rates during the NPI periods, the likelihood of a large outbreak in 2023 appears to be low. Comprehensive surveillance data are needed to accurately characterize future dynamics of EV-D68. The limited incidence of AFM cases in 2022, despite large EV-D68 outbreaks, poses further questions for the timing of the next AFM outbreaks., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
37. A non-human primate model for human norovirus infection.
- Author
-
Rimkute I, Chaimongkol N, Woods KD, Nagata BM, Darko S, Gudbole S, Henry AR, Sosnovtsev SV, Olia AS, Verardi R, Bok K, Todd JP, Woodward R, Kwong PD, Douek DC, Alves DA, Green KY, and Roederer M
- Subjects
- Humans, Animals, Macaca mulatta, Intestine, Small, Norovirus, Caliciviridae Infections, Vaccines
- Abstract
Norovirus infection can cause gastrointestinal disease in humans. Development of therapies and vaccines against norovirus have been limited by the lack of a suitable and reliable animal model. Here we established rhesus macaques as an animal model for human norovirus infection. We show that rhesus macaques are susceptible to oral infection with human noroviruses from two different genogroups. Variation in duration of virus shedding (days to weeks) between animals, evolution of the virus over the time of infection, induction of virus-specific adaptive immune responses, susceptibility to reinfection and preferential replication of norovirus in the jejunum of rhesus macaques was similar to infection reported in humans. We found minor pathological signs and changes in epithelial cell surface glycosylation patterns in the small intestine during infection. Detection of viral protein and RNA in intestinal biopsies confirmed the presence of the virus in chromogranin A-expressing epithelial cells, as it does in humans. Thus, rhesus macaques are a promising non-human primate model to evaluate vaccines and therapeutics against norovirus disease., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
38. The immunopathological landscape of human pre-TCRα deficiency: From rare to common variants.
- Author
-
Materna M, Delmonte OM, Bosticardo M, Momenilandi M, Conrey PE, Charmeteau-De Muylder B, Bravetti C, Bellworthy R, Cederholm A, Staels F, Ganoza CA, Darko S, Sayed S, Le Floc'h C, Ogishi M, Rinchai D, Guenoun A, Bolze A, Khan T, Gervais A, Krüger R, Völler M, Palterer B, Sadeghi-Shabestari M, Langlois de Septenville A, Schramm CA, Shah S, Tello-Cajiao JJ, Pala F, Amini K, Campos JS, Lima NS, Eriksson D, Lévy R, Seeleuthner Y, Jyonouchi S, Ata M, Al Ali F, Stittrich A, Deswarte C, Pereira A, Mégret J, Le Voyer T, Bastard P, Berteloot L, Dussiot M, Vladikine N, Cardenas PP, Jouanguy E, Alqahtani M, Hasan A, Thanaraj TA, Rosain J, Al Qureshah F, Sabato V, Alyanakian MA, Leruez-Ville M, Rozenberg F, Haddad E, Regueiro JR, Toribio ML, Kelsen JR, Salehi M, Nasiri S, Torabizadeh M, Rokni-Zadeh H, Changi-Ashtiani M, Vatandoost N, Moravej H, Akrami SM, Mazloomrezaei M, Cobat A, Meyts I, Toyofuku E, Nishimura M, Moriya K, Mizukami T, Imai K, Abel L, Malissen B, Al-Mulla F, Alkuraya FS, Parvaneh N, von Bernuth H, Beetz C, Davi F, Douek DC, Cheynier R, Langlais D, Landegren N, Marr N, Morio T, Shahrooei M, Schrijvers R, Henrickson SE, Luche H, Notarangelo LD, Casanova JL, and Béziat V
- Subjects
- Humans, Cell Differentiation, Homozygote, Loss of Function Mutation, Lymphocyte Count, Alleles, Infections immunology, Lymphoproliferative Disorders immunology, Pedigree, Male, Female, Middle Aged, Aged, Aged, 80 and over, Autoimmunity genetics, Intraepithelial Lymphocytes immunology, Receptors, Antigen, T-Cell, alpha-beta genetics, Membrane Glycoproteins genetics
- Abstract
We describe humans with rare biallelic loss-of-function PTCRA variants impairing pre-α T cell receptor (pre-TCRα) expression. Low circulating naive αβ T cell counts at birth persisted over time, with normal memory αβ and high γδ T cell counts. Their TCRα repertoire was biased, which suggests that noncanonical thymic differentiation pathways can rescue αβ T cell development. Only a minority of these individuals were sick, with infection, lymphoproliferation, and/or autoimmunity. We also report that 1 in 4000 individuals from the Middle East and South Asia are homozygous for a common hypomorphic PTCRA variant. They had normal circulating naive αβ T cell counts but high γδ T cell counts. Although residual pre-TCRα expression drove the differentiation of more αβ T cells, autoimmune conditions were more frequent in these patients compared with the general population.
- Published
- 2024
- Full Text
- View/download PDF
39. Publisher Correction: A non-human primate model for human norovirus infection.
- Author
-
Rimkute I, Chaimongkol N, Woods KD, Nagata BM, Darko S, Gudbole S, Henry AR, Sosnovtsev SV, Olia AS, Verardi R, Bok K, Todd JP, Woodward R, Kwong PD, Douek DC, Alves DA, Green KY, and Roederer M
- Published
- 2024
- Full Text
- View/download PDF
40. Systems immunology of transcriptional responses to viral infection identifies conserved antiviral pathways across macaques and humans.
- Author
-
Ratnasiri K, Zheng H, Toh J, Yao Z, Duran V, Donato M, Roederer M, Kamath M, Todd JM, Gagne M, Foulds KE, Francica JR, Corbett KS, Douek DC, Seder RA, Einav S, Blish CA, and Khatri P
- Subjects
- Animals, Humans, Immunoinformatics, Macaca, Antiviral Agents, Orthomyxoviridae Infections, Filoviridae
- Abstract
Viral pandemics and epidemics pose a significant global threat. While macaque models of viral disease are routinely used, it remains unclear how conserved antiviral responses are between macaques and humans. Therefore, we conducted a cross-species analysis of transcriptomic data from over 6,088 blood samples from macaques and humans infected with one of 31 viruses. Our findings demonstrate that irrespective of primate or viral species, there are conserved antiviral responses that are consistent across infection phase (acute, chronic, or latent) and viral genome type (DNA or RNA viruses). Leveraging longitudinal data from experimental challenges, we identify virus-specific response kinetics such as host responses to Coronaviridae and Orthomyxoviridae infections peaking 1-3 days earlier than responses to Filoviridae and Arenaviridae viral infections. Our results underscore macaque studies as a powerful tool for understanding viral pathogenesis and immune responses that translate to humans, with implications for viral therapeutic development and pandemic preparedness., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
41. XBB.1.5 monovalent booster improves antibody binding and neutralization against emerging SARS-CoV-2 Omicron variants.
- Author
-
Jain S, Kumar S, Lai L, Linderman S, Malik AA, Ellis ML, Godbole S, Solis D, Sahoo MK, Bechnak K, Paredes I, Tanios R, Kazzi B, Dib SM, Litvack MB, Wimalasena ST, Ciric C, Rostad C, West R, Teng IT, Wang D, Edupuganti S, Kwong PD, Rouphael N, Pinsky BA, Douek DC, Wrammert J, Moreno A, and Suthar MS
- Abstract
The rapid emergence of divergent SARS-CoV-2 variants has led to an update of the COVID-19 booster vaccine to a monovalent version containing the XBB.1.5 spike. To determine the neutralization breadth following booster immunization, we collected blood samples from 24 individuals pre- and post-XBB.1.5 mRNA booster vaccination (∼1 month). The XBB.1.5 booster improved both neutralizing activity against the ancestral SARS-CoV-2 strain (WA1) and the circulating Omicron variants, including EG.5.1, HK.3, HV.1, XBB.1.5 and JN.1. Relative to the pre-boost titers, the XBB.1.5 monovalent booster induced greater total IgG and IgG subclass binding, particular IgG4, to the XBB.1.5 spike as compared to the WA1 spike. We evaluated antigen-specific memory B cells (MBCs) using either spike or receptor binding domain (RBD) probes and found that the monovalent booster largely increases non-RBD cross-reactive MBCs. These data suggest that the XBB.1.5 monovalent booster induces cross-reactive antibodies that neutralize XBB.1.5 and related Omicron variants.
- Published
- 2024
- Full Text
- View/download PDF
42. Cleavage-intermediate Lassa virus trimer elicits neutralizing responses, identifies neutralizing nanobodies, and reveals an apex-situated site-of-vulnerability.
- Author
-
Gorman J, Cheung CS, Duan Z, Ou L, Wang M, Chen X, Cheng C, Biju A, Sun Y, Wang P, Yang Y, Zhang B, Boyington JC, Bylund T, Charaf S, Chen SJ, Du H, Henry AR, Liu T, Sarfo EK, Schramm CA, Shen CH, Stephens T, Teng IT, Todd JP, Tsybovsky Y, Verardi R, Wang D, Wang S, Wang Z, Zheng CY, Zhou T, Douek DC, Mascola JR, Ho DD, Ho M, and Kwong PD
- Subjects
- Animals, Guinea Pigs, Lassa virus, Antibodies, Viral, Antibodies, Neutralizing, Single-Domain Antibodies, Lassa Fever
- Abstract
Lassa virus (LASV) infection is expanding outside its traditionally endemic areas in West Africa, posing a pandemic biothreat. LASV-neutralizing antibodies, moreover, have proven difficult to elicit. To gain insight into LASV neutralization, here we develop a prefusion-stabilized LASV glycoprotein trimer (GPC), pan it against phage libraries comprising single-domain antibodies (nanobodies) from shark and camel, and identify one, D5, which neutralizes LASV. Cryo-EM analyses reveal D5 to recognize a cleavage-dependent site-of-vulnerability at the trimer apex. The recognized site appears specific to GPC intermediates, with protomers lacking full cleavage between GP1 and GP2 subunits. Guinea pig immunizations with the prefusion-stabilized cleavage-intermediate LASV GPC, first as trimer and then as a nanoparticle, induce neutralizing responses, targeting multiple epitopes including that of D5; we identify a neutralizing antibody (GP23) from the immunized guinea pigs. Collectively, our findings define a prefusion-stabilized GPC trimer, reveal an apex-situated site-of-vulnerability, and demonstrate elicitation of LASV-neutralizing responses by a cleavage-intermediate LASV trimer., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
43. Adjuvanted SARS-CoV-2 spike protein vaccination elicits long-lived plasma cells in nonhuman primates.
- Author
-
Prabhakaran M, Matassoli F, Leggat D, Hoover A, Srikanth A, Wu W, Henry AR, Wang J, Lin BC, Teng IT, Schramm CA, Castro M, Serebryannyy L, Jean-Baptiste N, Moore C, Gajjala S, Todd JM, McCarthy E, Narpala S, Francica J, Program VP, Corbett-Helaire KS, Douek DC, Kwong PD, Seder RA, Andrews SF, and McDermott AB
- Subjects
- Animals, Humans, Plasma Cells metabolism, Antibodies, Viral, SARS-CoV-2, Vaccination, Adjuvants, Immunologic, Primates, Antibodies, Neutralizing, Spike Glycoprotein, Coronavirus, COVID-19 prevention & control
- Abstract
Durable humoral immunity is mediated by long-lived plasma cells (LLPCs) that reside in the bone marrow. It remains unclear whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein vaccination is able to elicit and maintain LLPCs. Here, we describe a sensitive method to identify and isolate antigen-specific LLPCs by tethering antibodies secreted by these cells onto the cell surface. Using this method, we found that two doses of adjuvanted SARS-CoV-2 spike protein vaccination are able to induce spike protein-specific LLPC reservoirs enriched for receptor binding domain specificities in the bone marrow of nonhuman primates that are detectable for several months after vaccination. Immunoglobulin gene sequencing confirmed that several of these LLPCs were clones of memory B cells elicited 2 weeks after boost that had undergone further somatic hypermutation. Many of the antibodies secreted by these LLPCs also exhibited improved neutralization and cross-reactivity compared with earlier time points. These findings establish our method as a means to sensitively and reliably detect rare antigen-specific LLPCs and demonstrate that adjuvanted SARS-CoV-2 spike protein vaccination establishes spike protein-specific LLPC reservoirs.
- Published
- 2024
- Full Text
- View/download PDF
44. High frequency of HIV precursor-target-specific B cells in sub-Saharan populations.
- Author
-
Matassoli F, Cagigi A, Shen CH, Henry AR, Johnston TS, Schramm CA, Cottrell CA, Kalyuzhniy O, Spangler A, Eller L, Robb M, Eller M, Naluyima P, Kwong PD, Douek DC, Schief WR, Andrews SF, and McDermott AB
- Subjects
- Humans, Antibodies, Neutralizing, HIV Antibodies, Precursor Cells, B-Lymphoid, HIV Envelope Protein gp120, HIV Infections, HIV-1
- Abstract
HIV gp120 engineered outer domain germline-targeting version 8 (eOD-GT8) was designed specifically to engage naive B cell precursors of VRC01-class antibodies. However, the frequency and affinity of naive B cell precursors able to recognize eOD-GT8 have been evaluated only in U.S. populations. HIV infection is disproportionally concentrated in sub-Saharan Africa, so we seek to characterize naive B cells able to recognize eOD-GT8 in sub-Saharan cohorts. We demonstrate that people from sub-Saharan Africa have a higher or equivalent frequency of naive B cells able to engage eOD-GT8 compared with people from the U.S. Genetically, the higher frequency of eOD-GT8-positive cells is accompanied by a higher level of naive B cells with gene signatures characteristic of the VRC01 class, as well as other CD4bs-directed antibodies. Our study demonstrates that vaccination with eOD-GT8 in sub-Saharan Africa could be successful at expanding and establishing a pool of CD4bs-directed memory B cells from naive precursors., Competing Interests: Declaration of interests The authors declare no competing interests. This work was supported by a cooperative agreement (W81XWH-18-2-0040) between the Henry M. Jackson Foundation (HJF) for the Advancement of Military Medicine, Inc., and the U.S. Department of Defense (DOD). This research was funded, in part, by the National Institute of Allergy and Infectious Diseases. The views expressed are those of the authors and should not be construed to represent the positions of the U.S. Army, the DOD, or the HJF. The investigators have adhered to the policies for protection of human subjects as prescribed in AR 70-25., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
45. Interaction dynamics between innate and adaptive immune cells responding to SARS-CoV-2 vaccination in non-human primates.
- Author
-
Schramm CA, Moon D, Peyton L, Lima NS, Wake C, Boswell KL, Henry AR, Laboune F, Ambrozak D, Darko SW, Teng IT, Foulds KE, Carfi A, Edwards DK, Kwong PD, Koup RA, Seder RA, and Douek DC
- Subjects
- Animals, Humans, COVID-19 Vaccines, Macaca mulatta, SARS-CoV-2, Vaccination, Antibodies, Viral, COVID-19 prevention & control, Blood Group Antigens
- Abstract
As SARS-CoV-2 variants continue evolving, testing updated vaccines in non-human primates remains important for guiding human clinical practice. To date, such studies have focused on antibody titers and antigen-specific B and T cell frequencies. Here, we extend our understanding by integrating innate and adaptive immune responses to mRNA-1273 vaccination in rhesus macaques. We sorted innate immune cells from a pre-vaccine time point, as well as innate immune cells and antigen-specific peripheral B and T cells two weeks after each of two vaccine doses and used single-cell sequencing to assess the transcriptomes and adaptive immune receptors of each cell. We show that a subset of S-specific T cells expresses cytokines critical for activating innate responses, with a concomitant increase in CCR5-expressing intermediate monocytes and a shift of natural killer cells to a more cytotoxic phenotype. The second vaccine dose, administered 4 weeks after the first, elicits an increase in circulating germinal center-like B cells 2 weeks later, which are more clonally expanded and enriched for epitopes in the receptor binding domain. Both doses stimulate inflammatory response genes associated with elevated antibody production. Overall, we provide a comprehensive picture of bidirectional signaling between innate and adaptive components of the immune system and suggest potential mechanisms for the enhanced response to secondary exposure., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2023
- Full Text
- View/download PDF
46. Durable immunity to SARS-CoV-2 in both lower and upper airways achieved with a gorilla adenovirus (GRAd) S-2P vaccine in non-human primates.
- Author
-
Moliva JI, Andrew SF, Flynn BJ, Wagner DA, Foulds KE, Gagne M, Flebbe DR, Lamb E, Provost S, Marquez J, Mychalowych A, Lorag CG, Honeycutt CC, Burnett MR, McCormick L, Henry AR, Godbole S, Davis-Gardner ME, Minai M, Bock KW, Nagata BM, Todd JM, McCarthy E, Dodson A, Kouneski K, Cook A, Pessaint L, Ry AV, Valentin D, Young S, Littman Y, Boon ACM, Suthar MS, Lewis MG, Andersen H, Alves DA, Woodward R, Leuzzi A, Vitelli A, Colloca S, Folgori A, Raggiolli A, Capone S, Nason MC, Douek DC, Roederer M, Seder RA, and Sullivan NJ
- Abstract
SARS-CoV-2 continues to pose a global threat, and current vaccines, while effective against severe illness, fall short in preventing transmission. To address this challenge, there's a need for vaccines that induce mucosal immunity and can rapidly control the virus. In this study, we demonstrate that a single immunization with a novel gorilla adenovirus-based vaccine (GRAd) carrying the pre-fusion stabilized Spike protein (S-2P) in non-human primates provided protective immunity for over one year against the BA.5 variant of SARS-CoV-2. A prime-boost regimen using GRAd followed by adjuvanted S-2P (GRAd+S-2P) accelerated viral clearance in both the lower and upper airways. GRAd delivered via aerosol (GRAd(AE)+S-2P) modestly improved protection compared to its matched intramuscular regimen, but showed dramatically superior boosting by mRNA and, importantly, total virus clearance in the upper airway by day 4 post infection. GrAd vaccination regimens elicited robust and durable systemic and mucosal antibody responses to multiple SARS-CoV-2 variants, but only GRAd(AE)+S-2P generated long-lasting T cell responses in the lung. This research underscores the flexibility of the GRAd vaccine platform to provide durable immunity against SARS-CoV-2 in both the lower and upper airways., Competing Interests: Declaration of interests M.R., N.J.S., and D.C.D. are inventors on U.S. Patent Application No. 63/147,419 entitled “Antibodies Targeting the Spike Protein of Coronaviruses”. L.P., A.V.R., D.V., A.C., A.D., M.G.L., and H.A. are employees of Bioqual, Inc. A.L., A.V., S.Co., A.F., A.R., and S.Ca. are employees of ReiThera Srl. S.Co. and A.F. are shareholders of Keires AG. A.V., S.Co. and A.R. are named inventors of the Patent Application No. 20183515.4 entitled “Gorilla Adenovirus Nucleic Acid- and Amino Acid-Sequences, Vectors Containing Same, and Uses Thereof”. The other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
47. Mucosal Adenoviral-vectored Vaccine Boosting Durably Prevents XBB.1.16 Infection in Nonhuman Primates.
- Author
-
Gagne M, Flynn BJ, Andrew SF, Flebbe DR, Mychalowych A, Lamb E, Davis-Gardner ME, Burnett MR, Serebryannyy LA, Lin BC, Pessaint L, Todd JM, Ziff ZE, Maule E, Carroll R, Naisan M, Jethmalani Y, Case JB, Dmitriev IP, Kashentseva EA, Ying B, Dodson A, Kouneski K, Doria-Rose NA, O'Dell S, Godbole S, Laboune F, Henry AR, Marquez J, Teng IT, Wang L, Zhou Q, Wali B, Ellis M, Zouantchangadou S, Ry AV, Lewis MG, Andersen H, Kwong PD, Curiel DT, Foulds KE, Nason MC, Suthar MS, Roederer M, Diamond MS, Douek DC, and Seder RA
- Abstract
Waning immunity and continued virus evolution have limited the durability of protection from symptomatic infection mediated by intramuscularly (IM)-delivered mRNA vaccines against COVID-19 although protection from severe disease remains high. Mucosal vaccination has been proposed as a strategy to increase protection at the site of SARS-CoV-2 infection by enhancing airway immunity, potentially reducing rates of infection and transmission. Here, we compared protection against XBB.1.16 virus challenge 5 months following IM or mucosal boosting in non-human primates (NHP) that had previously received a two-dose mRNA-1273 primary vaccine regimen. The mucosal boost was composed of a bivalent chimpanzee adenoviral-vectored vaccine encoding for both SARS-CoV-2 WA1 and BA.5 spike proteins (ChAd-SARS-CoV-2-S) and delivered either by an intranasal mist or an inhaled aerosol. An additional group of animals was boosted by the IM route with bivalent WA1/BA.5 spike-matched mRNA (mRNA-1273.222) as a benchmark control. NHP were challenged in the upper and lower airways 18 weeks after boosting with XBB.1.16, a heterologous Omicron lineage strain. Cohorts boosted with ChAd-SARS-CoV-2-S by an aerosolized or intranasal route had low to undetectable virus replication as assessed by levels of subgenomic SARS-CoV-2 RNA in the lungs and nose, respectively. In contrast, animals that received the mRNA-1273.222 boost by the IM route showed minimal protection against virus replication in the upper airway but substantial reduction of virus RNA levels in the lower airway. Immune analysis showed that the mucosal vaccines elicited more durable antibody and T cell responses than the IM vaccine. Protection elicited by the aerosolized vaccine was associated with mucosal IgG and IgA responses, whereas protection elicited by intranasal delivery was mediated primarily by mucosal IgA. Thus, durable immunity and effective protection against a highly transmissible heterologous variant in both the upper and lower airways can be achieved by mucosal delivery of a virus-vectored vaccine. Our study provides a template for the development of mucosal vaccines that limit infection and transmission against respiratory pathogens.
- Published
- 2023
- Full Text
- View/download PDF
48. Comparative Analysis of SARS-CoV-2 Antigenicity across Assays and in Human and Animal Model Sera.
- Author
-
Mühlemann B, Wilks SH, Baracco L, Bekliz M, Carreño JM, Corman VM, Davis-Gardner ME, Dejnirattisai W, Diamond MS, Douek DC, Drosten C, Eckerle I, Edara VV, Ellis M, Fouchier RAM, Frieman M, Godbole S, Haagmans B, Halfmann PJ, Henry AR, Jones TC, Katzelnick LC, Kawaoka Y, Kimpel J, Krammer F, Lai L, Liu C, Lusvarghi S, Meyer B, Mongkolsapaya J, Montefiori DC, Mykytyn A, Netzl A, Pollett S, Rössler A, Screaton GR, Shen X, Sigal A, Simon V, Subramanian R, Supasa P, Suthar M, Türeli S, Wang W, Weiss CD, and Smith DJ
- Abstract
The antigenic evolution of SARS-CoV-2 requires ongoing monitoring to judge the immune escape of newly arising variants. A surveillance system necessitates an understanding of differences in neutralization titers measured in different assays and using human and animal sera. We compared 18 datasets generated using human, hamster, and mouse sera, and six different neutralization assays. Titer magnitude was lowest in human, intermediate in hamster, and highest in mouse sera. Fold change, immunodominance patterns and antigenic maps were similar among sera. Most assays yielded similar results, except for differences in fold change in cytopathic effect assays. Not enough data was available for conclusively judging mouse sera, but hamster sera were a consistent surrogate for human first-infection sera., Competing Interests: VMC: Named on patents regarding SARS-CoV-2 serological testing and monoclonal antibodies. MSD: Consultant for Inbios, Vir Biotechnology, Ocugen, Topspin Therapeutics, Moderna, and Immunome. The Diamond laboratory has received unrelated funding support in sponsored research agreements from Moderna, Vir Biotechnology, Generate Biomedicines, and Emergent BioSolutions. YK: Received unrelated funding support from Daiichi Sankyo Pharmaceutical, Toyama Chemical, Tauns Laboratories, Inc., Shionogi & Co. LTD, Otsuka Pharmaceutical, KM Biologics, Kyoritsu Seiyaku, Shinya Corporation, and Fuji Rebio. IE: Research grant and speakers fees from Moderna. BMe: Research grant from Moderna. GRS: Is on the GSK Vaccines Scientific Advisory Board. Oxford University holds intellectual property related to the Oxford-AstraZeneca vaccine. MS: Serves in an advisory role for Ocugen, Inc. SP: Reports that the Uniformed Services University (USU) Infectious Diseases Clinical Research Program (IDCRP), a US Department of Defense institution, and the Henry M. Jackson Foundation (HJF) were funded under a Cooperative Research and Development Agreement to conduct an unrelated phase III COVID-19 monoclonal antibody immunoprophylaxis trial sponsored by AstraZeneca. The HJF, in support of the USU IDCRP, was funded by the Department of Defense Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense to augment the conduct of an unrelated phase III vaccine trial sponsored by AstraZeneca. Both trials were part of the U.S. Government COVID-19 response. Neither is related to the work presented here.
- Published
- 2023
- Full Text
- View/download PDF
49. Modulation of type I interferon responses potently inhibits SARS-CoV-2 replication and inflammation in rhesus macaques.
- Author
-
Viox EG, Hoang TN, Upadhyay AA, Nchioua R, Hirschenberger M, Strongin Z, Tharp GK, Pino M, Nguyen K, Harper JL, Gagne M, Marciano S, Boddapati AK, Pellegrini KL, Pradhan A, Tisoncik-Go J, Whitmore LS, Karunakaran KA, Roy M, Kirejczyk S, Curran EH, Wallace C, Wood JS, Connor-Stroud F, Voigt EA, Monaco CM, Gordon DE, Kasturi SP, Levit RD, Gale M Jr, Vanderford TH, Silvestri G, Busman-Sahay K, Estes JD, Vaccari M, Douek DC, Sparrer KMJ, Johnson RP, Kirchhoff F, Schreiber G, Bosinger SE, and Paiardini M
- Subjects
- Animals, SARS-CoV-2, Macaca mulatta, Virus Replication, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Inflammation drug therapy, Interferon Type I pharmacology, COVID-19
- Abstract
Type I interferons (IFN-I) are critical mediators of innate control of viral infections but also drive the recruitment of inflammatory cells to sites of infection, a key feature of severe coronavirus disease 2019. Here, IFN-I signaling was modulated in rhesus macaques (RMs) before and during acute SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infection using a mutated IFN-α2 (IFN-modulator; IFNmod), which has previously been shown to reduce the binding and signaling of endogenous IFN-I. IFNmod treatment in uninfected RMs was observed to induce a modest up-regulation of only antiviral IFN-stimulated genes (ISGs); however, in SARS-CoV-2-infected RMs, IFNmod reduced both antiviral and inflammatory ISGs. IFNmod treatment resulted in a potent reduction in SARS-CoV-2 viral loads both in vitro in Calu-3 cells and in vivo in bronchoalveolar lavage (BAL), upper airways, lung, and hilar lymph nodes of RMs. Furthermore, in SARS-CoV-2-infected RMs, IFNmod treatment potently reduced inflammatory cytokines, chemokines, and CD163
+ MRC1- inflammatory macrophages in BAL and expression of Siglec-1 on circulating monocytes. In the lung, IFNmod also reduced pathogenesis and attenuated pathways of inflammasome activation and stress response during acute SARS-CoV-2 infection. Using an intervention targeting both IFN-α and IFN-β pathways, this study shows that, whereas early IFN-I restrains SARS-CoV-2 replication, uncontrolled IFN-I signaling critically contributes to SARS-CoV-2 inflammation and pathogenesis in the moderate disease model of RMs.- Published
- 2023
- Full Text
- View/download PDF
50. HIV-1 neutralizing antibodies elicited in humans by a prefusion-stabilized envelope trimer form a reproducible class targeting fusion peptide.
- Author
-
Wang S, Matassoli F, Zhang B, Liu T, Shen CH, Bylund T, Johnston T, Henry AR, Teng IT, Tripathi P, Becker JE, Changela A, Chaudhary R, Cheng C, Gaudinski M, Gorman J, Harris DR, Lee M, Morano NC, Novik L, O'Dell S, Olia AS, Parchment DK, Rawi R, Roberts-Torres J, Stephens T, Tsybovsky Y, Wang D, Van Wazer DJ, Zhou T, Doria-Rose NA, Koup RA, Shapiro L, Douek DC, McDermott AB, and Kwong PD
- Subjects
- Humans, Antibodies, Neutralizing, env Gene Products, Human Immunodeficiency Virus, HIV Antibodies, Peptides, AIDS Vaccines, HIV Infections, HIV Seropositivity, HIV-1
- Abstract
Elicitation of antibodies that neutralize the tier-2 neutralization-resistant isolates that typify HIV-1 transmission has been a long-sought goal. Success with prefusion-stabilized envelope trimers eliciting autologous neutralizing antibodies has been reported in multiple vaccine-test species, though not in humans. To investigate elicitation of HIV-1 neutralizing antibodies in humans, here, we analyze B cells from a phase I clinical trial of the "DS-SOSIP"-stabilized envelope trimer from strain BG505, identifying two antibodies, N751-2C06.01 and N751-2C09.01 (named for donor-lineage.clone), that neutralize the autologous tier-2 strain, BG505. Though derived from distinct lineages, these antibodies form a reproducible antibody class that targets the HIV-1 fusion peptide. Both antibodies are highly strain specific, which we attribute to their partial recognition of a BG505-specific glycan hole and to their binding requirements for a few BG505-specific residues. Prefusion-stabilized envelope trimers can thus elicit autologous tier-2 neutralizing antibodies in humans, with initially identified neutralizing antibodies recognizing the fusion-peptide site of vulnerability., Competing Interests: Declaration of interests The authors declare no competing interests., (Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.