129 results on '"Oltz EM"'
Search Results
2. Neutralization and Stability of JN.1-derived LB.1, KP.2.3, KP.3 and KP.3.1.1 Subvariants.
- Author
-
Li P, Faraone JN, Hsu CC, Chamblee M, Liu Y, Zheng YM, Xu Y, Carlin C, Horowitz JC, Mallampalli RK, Saif LJ, Oltz EM, Jones D, Li J, Gumina RJ, Bednash JS, Xu K, and Liu SL
- Abstract
During the summer of 2024, COVID-19 cases surged globally, driven by variants derived from JN.1 subvariants of SARS-CoV-2 that feature new mutations, particularly in the N-terminal domain (NTD) of the spike protein. In this study, we report on the neutralizing antibody (nAb) escape, infectivity, fusion, and stability of these subvariants-LB.1, KP.2.3, KP.3, and KP.3.1.1. Our findings demonstrate that all of these subvariants are highly evasive of nAbs elicited by the bivalent mRNA vaccine, the XBB.1.5 monovalent mumps virus-based vaccine, or from infections during the BA.2.86/JN.1 wave. This reduction in nAb titers is primarily driven by a single serine deletion (DelS31) in the NTD of the spike, leading to a distinct antigenic profile compared to the parental JN.1 and other variants. We also found that the DelS31 mutation decreases pseudovirus infectivity in CaLu-3 cells, which correlates with impaired cell-cell fusion. Additionally, the spike protein of DelS31 variants appears more conformationally stable, as indicated by reduced S1 shedding both with and without stimulation by soluble ACE2, and increased resistance to elevated temperatures. Molecular modeling suggests that the DelS31 mutation induces a conformational change that stabilizes the NTD and strengthens the NTD-Receptor-Binding Domain (RBD) interaction, thus favoring the down conformation of RBD and reducing accessibility to both the ACE2 receptor and certain nAbs. Additionally, the DelS31 mutation introduces an N-linked glycan modification at N30, which shields the underlying NTD region from antibody recognition. Our data highlight the critical role of NTD mutations in the spike protein for nAb evasion, stability, and viral infectivity, and suggest consideration of updating COVID-19 vaccines with antigens containing DelS31., Competing Interests: DECLARATION OF INTERESTS The authors do not declare any competing interests.
- Published
- 2024
- Full Text
- View/download PDF
3. Neutralization escape, infectivity, and membrane fusion of JN.1-derived SARS-CoV-2 SLip, FLiRT, and KP.2 variants.
- Author
-
Li P, Faraone JN, Hsu CC, Chamblee M, Zheng YM, Carlin C, Bednash JS, Horowitz JC, Mallampalli RK, Saif LJ, Oltz EM, Jones D, Li J, Gumina RJ, Xu K, and Liu SL
- Subjects
- Animals, Humans, Cricetinae, Antibodies, Viral immunology, Membrane Fusion, Antibodies, Monoclonal immunology, Mutation, Chlorocebus aethiops, Angiotensin-Converting Enzyme 2 metabolism, Epitopes immunology, Vero Cells, Neutralization Tests, COVID-19 Vaccines immunology, SARS-CoV-2 immunology, Antibodies, Neutralizing immunology, COVID-19 immunology, COVID-19 virology, Spike Glycoprotein, Coronavirus immunology, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism
- Abstract
We investigate JN.1-derived subvariants SLip, FLiRT, and KP.2 for neutralization by antibodies in vaccinated individuals, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected patients, or class III monoclonal antibody S309. Compared to JN.1, SLip, KP.2, and especially FLiRT exhibit increased resistance to bivalent-vaccinated and BA.2.86/JN.1-wave convalescent human sera. XBB.1.5 monovalent-vaccinated hamster sera robustly neutralize FLiRT and KP.2 but have reduced efficiency for SLip. All subvariants are resistant to S309 and show decreased infectivity, cell-cell fusion, and spike processing relative to JN.1. Modeling reveals that L455S and F456L in SLip reduce spike binding for ACE2, while R346T in FLiRT and KP.2 strengthens it. These three mutations, alongside D339H, alter key epitopes in spike, likely explaining the reduced sensitivity of these subvariants to neutralization. Our findings highlight the increased neutralization resistance of JN.1 subvariants and suggest that future vaccine formulations should consider the JN.1 spike as an immunogen, although the current XBB.1.5 monovalent vaccine could still offer adequate protection., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Characteristics of JN.1-derived SARS-CoV-2 subvariants SLip, FLiRT, and KP.2 in neutralization escape, infectivity and membrane fusion.
- Author
-
Li P, Faraone JN, Hsu CC, Chamblee M, Zheng YM, Carlin C, Bednash JS, Horowitz JC, Mallampalli RK, Saif LJ, Oltz EM, Jones D, Li J, Gumina RJ, Xu K, and Liu SL
- Abstract
SARS-CoV-2 variants derived from the immune evasive JN.1 are on the rise worldwide. Here, we investigated JN.1-derived subvariants SLip, FLiRT, and KP.2 for their ability to be neutralized by antibodies in bivalent-vaccinated human sera, XBB.1.5 monovalent-vaccinated hamster sera, sera from people infected during the BA.2.86/JN.1 wave, and class III monoclonal antibody (Mab) S309. We found that compared to parental JN.1, SLip and KP.2, and especially FLiRT, exhibit increased resistance to COVID-19 bivalent-vaccinated human sera and BA.2.86/JN.1-wave convalescent sera. Interestingly, antibodies in XBB.1.5 monovalent vaccinated hamster sera robustly neutralized FLiRT and KP.2 but had reduced efficiency for SLip. These JN.1 subvariants were resistant to neutralization by Mab S309. In addition, we investigated aspects of spike protein biology including infectivity, cell-cell fusion and processing, and found that these subvariants, especially SLip, had a decreased infectivity and membrane fusion relative to JN.1, correlating with decreased spike processing. Homology modeling revealed that L455S and F456L mutations in SLip reduced local hydrophobicity in the spike and hence its binding to ACE2. In contrast, the additional R346T mutation in FLiRT and KP.2 strengthened conformational support of the receptor-binding motif, thus counteracting the effects of L455S and F456L. These three mutations, alongside D339H, which is present in all JN.1 sublineages, alter the epitopes targeted by therapeutic Mabs, including class I and class III S309, explaining their reduced sensitivity to neutralization by sera and S309. Together, our findings provide insight into neutralization resistance of newly emerged JN.1 subvariants and suggest that future vaccine formulations should consider JN.1 spike as immunogen, although the current XBB.1.5 monovalent vaccine could still offer adequate protection., Competing Interests: The authors have no competing interests to disclose. S.-L.L. conceived and directed the project. R.J.G led the clinical study/experimental design and implementation. P.L. performed the experiments and data processing and analyses. K.X. performed molecular modeling and data analyses. D.J. led SARS-CoV-2 variant genotyping and DNA sequencing ana- lyses. C.C., J.S.B., J.C.H., R.M., and R.J.G. provided clinical samples and related information. C.C.H, M.C., and J.L. provided hamster serum samples and associated information. P.L., J.N.F. and S.-L.L. wrote the paper. Y.-M.Z, L.J.S., E.M.O. provided insightful discussion and revision of the manuscript. DECLARATION OF INTERESTS The authors do not declare any competing interests.
- Published
- 2024
- Full Text
- View/download PDF
5. Distinct patterns of SARS-CoV-2 BA.2.87.1 and JN.1 variants in immune evasion, antigenicity, and cell-cell fusion.
- Author
-
Li P, Liu Y, Faraone JN, Hsu CC, Chamblee M, Zheng Y-M, Carlin C, Bednash JS, Horowitz JC, Mallampalli RK, Saif LJ, Oltz EM, Jones D, Li J, Gumina RJ, and Liu S-L
- Subjects
- Animals, Humans, Cricetinae, Spike Glycoprotein, Coronavirus immunology, Spike Glycoprotein, Coronavirus genetics, COVID-19 Vaccines immunology, SARS-CoV-2 immunology, SARS-CoV-2 genetics, Antibodies, Neutralizing immunology, Antibodies, Neutralizing blood, Immune Evasion, COVID-19 immunology, COVID-19 virology, Antibodies, Viral blood, Antibodies, Viral immunology, Cell Fusion
- Abstract
The rapid evolution of SARS-CoV-2 variants presents a constant challenge to the global vaccination effort. In this study, we conducted a comprehensive investigation into two newly emerged variants, BA.2.87.1 and JN.1, focusing on their neutralization resistance, infectivity, antigenicity, cell-cell fusion, and spike processing. Neutralizing antibody (nAb) titers were assessed in diverse cohorts, including individuals who received a bivalent mRNA vaccine booster, patients infected during the BA.2.86/JN.1-wave, and hamsters vaccinated with XBB.1.5-monovalent vaccine. We found that BA.2.87.1 shows much less nAb escape from WT-BA.4/5 bivalent mRNA vaccination and JN.1-wave breakthrough infection sera compared to JN.1 and XBB.1.5. Interestingly, BA.2.87.1 is more resistant to neutralization by XBB.1.5-monovalent-vaccinated hamster sera than BA.2.86/JN.1 and XBB.1.5, but efficiently neutralized by a class III monoclonal antibody S309, which largely fails to neutralize BA.2.86/JN.1. Importantly, BA.2.87.1 exhibits higher levels of infectivity, cell-cell fusion activity, and furin cleavage efficiency than BA.2.86/JN.1. Antigenically, we found that BA.2.87.1 is closer to the ancestral BA.2 compared to other recently emerged Omicron subvariants including BA.2.86/JN.1 and XBB.1.5. Altogether, these results highlight immune escape properties as well as biology of new variants and underscore the importance of continuous surveillance and informed decision-making in the development of effective vaccines., Importance: This study investigates the recently emerged SARS-CoV-2 variants, BA.2.87.1 and JN.1, in comparison to earlier variants and the parental D614G. Varied infectivity and cell-cell fusion activity among these variants suggest potential disparities in their ability to infect target cells and possibly pathogenesis. BA.2.87.1 exhibits lower nAb escape from bivalent mRNA vaccinee and BA.2.86/JN.1-infected sera than JN.1 but is relatively resistance to XBB.1.5-vaccinated hamster sera, revealing distinct properties in immune reason and underscoring the significance of continuing surveillance of variants and reformulation of vaccines. Antigenic differences between BA.2.87.1 and other earlier variants yield critical information not only for antibody evasion but also for viral evolution. In conclusion, this study furnishes timely insights into the spike biology and immune escape of the emerging variants BA.2.87.1 and JN.1, thus guiding effective vaccine development and informing public health interventions., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
6. Locus folding mechanisms determine modes of antigen receptor gene assembly.
- Author
-
Allyn BM, Hayer KE, Oyeniran C, Nganga V, Lee K, Mishra B, Sacan A, Oltz EM, and Bassing CH
- Subjects
- Endonucleases, Mutation, Promoter Regions, Genetic genetics, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, Chromatin genetics
- Abstract
The dynamic folding of genomes regulates numerous biological processes, including antigen receptor (AgR) gene assembly. We show that, unlike other AgR loci, homotypic chromatin interactions and bidirectional chromosome looping both contribute to structuring Tcrb for efficient long-range V(D)J recombination. Inactivation of the CTCF binding element (CBE) or promoter at the most 5'Vβ segment (Trbv1) impaired loop extrusion originating locally and extending to DβJβ CBEs at the opposite end of Tcrb. Promoter or CBE mutation nearly eliminated Trbv1 contacts and decreased RAG endonuclease-mediated Trbv1 recombination. Importantly, Trbv1 rearrangement can proceed independent of substrate orientation, ruling out scanning by DβJβ-bound RAG as the sole mechanism of Vβ recombination, distinguishing it from Igh. Our data indicate that CBE-dependent generation of loops cooperates with promoter-mediated activation of chromatin to juxtapose Vβ and DβJβ segments for recombination through diffusion-based synapsis. Thus, the mechanisms that fold a genomic region can influence molecular processes occurring in that space, which may include recombination, repair, and transcriptional programming., (© 2024 Allyn et al.)
- Published
- 2024
- Full Text
- View/download PDF
7. Immune evasion, infectivity, and fusogenicity of SARS-CoV-2 BA.2.86 and FLip variants.
- Author
-
Qu P, Xu K, Faraone JN, Goodarzi N, Zheng YM, Carlin C, Bednash JS, Horowitz JC, Mallampalli RK, Saif LJ, Oltz EM, Jones D, Gumina RJ, and Liu SL
- Subjects
- Humans, Antibodies, Monoclonal, Antibodies, Neutralizing, Antibodies, Viral, COVID-19 immunology, COVID-19 Vaccines, Immune Evasion, SARS-CoV-2 classification, SARS-CoV-2 physiology
- Abstract
Evolution of SARS-CoV-2 requires the reassessment of current vaccine measures. Here, we characterized BA.2.86 and XBB-derived variant FLip by investigating their neutralization alongside D614G, BA.1, BA.2, BA.4/5, XBB.1.5, and EG.5.1 by sera from 3-dose-vaccinated and bivalent-vaccinated healthcare workers, XBB.1.5-wave-infected first responders, and monoclonal antibody (mAb) S309. We assessed the biology of the variant spikes by measuring viral infectivity and membrane fusogenicity. BA.2.86 is less immune evasive compared to FLip and other XBB variants, consistent with antigenic distances. Importantly, distinct from XBB variants, mAb S309 was unable to neutralize BA.2.86, likely due to a D339H mutation based on modeling. BA.2.86 had relatively high fusogenicity and infectivity in CaLu-3 cells but low fusion and infectivity in 293T-ACE2 cells compared to some XBB variants, suggesting a potentially different conformational stability of BA.2.86 spike. Overall, our study underscores the importance of SARS-CoV-2 variant surveillance and the need for updated COVID-19 vaccines., Competing Interests: Declaration of interests The authors do not declare any competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. mRNA vaccines against SARS-CoV-2 induce divergent antigen-specific T-cell responses in patients with lung cancer.
- Author
-
Song NJ, Chakravarthy KB, Jeon H, Bolyard C, Reynolds K, Weller KP, Reisinger S, Wang Y, Li A, Jiang S, Ma Q, Barouch DH, Rubinstein MP, Shields PG, Oltz EM, Chung D, and Li Z
- Subjects
- Humans, Female, mRNA Vaccines, COVID-19 Vaccines therapeutic use, SARS-CoV-2, Leukocytes, Mononuclear, Lung Neoplasms, COVID-19 prevention & control
- Abstract
Background: The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant is highly transmissible and evades pre-established immunity. Messenger RNA (mRNA) vaccination against ancestral strain spike protein can induce intact T-cell immunity against the Omicron variant, but efficacy of booster vaccination in patients with late-stage lung cancer on immune-modulating agents including anti-programmed cell death protein 1(PD-1)/programmed death-ligand 1 (PD-L1) has not yet been elucidated., Methods: We assessed T-cell responses using a modified activation-induced marker assay, coupled with high-dimension flow cytometry analyses. Peripheral blood mononuclear cells (PBMCs) were stimulated with various viral peptides and antigen-specific T-cell responses were evaluated using flow cytometry., Results: Booster vaccines induced CD8
+ T-cell response against the ancestral SARS-CoV-2 strain and Omicron variant in both non-cancer subjects and patients with lung cancer, but only a marginal induction was detected for CD4+ T cells. Importantly, antigen-specific T cells from patients with lung cancer showed distinct subpopulation dynamics with varying degrees of differentiation compared with non-cancer subjects, with evidence of dysfunction. Notably, female-biased T-cell responses were observed., Conclusion: We conclude that patients with lung cancer on immunotherapy show a substantial qualitative deviation from non-cancer subjects in their T-cell response to mRNA vaccines, highlighting the need for heightened protective measures for patients with cancer to minimize the risk of breakthrough infection with the Omicron and other future variants., Competing Interests: Competing interests: No, there are no competing interests., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2024
- Full Text
- View/download PDF
9. The transcription factor Aiolos restrains the activation of intestinal intraepithelial lymphocytes.
- Author
-
Yomogida K, Trsan T, Sudan R, Rodrigues PF, Ulezko Antonova A, Ingle H, Luccia BD, Collins PL, Cella M, Gilfillan S, Baldridge MT, Oltz EM, and Colonna M
- Subjects
- Animals, Mice, CD8 Antigens metabolism, Interleukin-15 metabolism, Intestinal Mucosa metabolism, Mice, Inbred C57BL, Mice, Knockout, Intraepithelial Lymphocytes metabolism, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Intestinal intraepithelial lymphocytes (IELs) exhibit prompt innate-like responses to microenvironmental cues and require strict control of effector functions. Here we showed that Aiolos, an Ikaros zinc-finger family member encoded by Ikzf3, acted as a regulator of IEL activation. Ikzf3
-/- CD8αα+ IELs had elevated expression of NK receptors, cytotoxic enzymes, cytokines and chemokines. Single-cell RNA sequencing of Ikzf3-/- and Ikzf3+/+ IELs showed an amplified effector machinery in Ikzf3-/- CD8αα+ IELs compared to Ikzf3+/+ counterparts. Ikzf3-/- CD8αα+ IELs had increased responsiveness to interleukin-15, which explained a substantial part, but not all, of the observed phenotypes. Aiolos binding sites were close to those for the transcription factors STAT5 and RUNX, which promote interleukin-15 signaling and cytolytic programs, and Ikzf3 deficiency partially increased chromatin accessibility and histone acetylation in these regions. Ikzf3 deficiency in mice enhanced susceptibility to colitis, underscoring the relevance of Aiolos in regulating the effector function in IELs., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
10. Immune evasion and membrane fusion of SARS-CoV-2 XBB subvariants EG.5.1 and XBB.2.3.
- Author
-
Faraone JN, Qu P, Goodarzi N, Zheng YM, Carlin C, Saif LJ, Oltz EM, Xu K, Jones D, Gumina RJ, and Liu SL
- Subjects
- Humans, HEK293 Cells, Immune Evasion, SARS-CoV-2 genetics, Antibodies, Neutralizing, mRNA Vaccines, Antibodies, Viral, Membrane Fusion, COVID-19 prevention & control
- Abstract
Immune evasion by SARS-CoV-2 paired with immune imprinting from monovalent mRNA vaccines has resulted in attenuated neutralizing antibody responses against Omicron subvariants. In this study, we characterized two new XBB variants rising in circulation - EG.5.1 and XBB.2.3, for their neutralization and syncytia formation. We determined the neutralizing antibody titers in sera of individuals that received a bivalent mRNA vaccine booster, BA.4/5-wave infection, or XBB.1.5-wave infection. Bivalent vaccination-induced antibodies neutralized ancestral D614G efficiently, but to a much less extent, two new EG.5.1 and XBB.2.3 variants. In fact, the enhanced neutralization escape of EG.5.1 appeared to be driven by its key defining mutation XBB.1.5-F456L. Notably, infection by BA.4/5 or XBB.1.5 afforded little, if any, neutralization against EG.5.1, XBB.2.3 and previous XBB variants - especially in unvaccinated individuals, with average neutralizing antibody titers near the limit of detection. Additionally, we investigated the infectivity, fusion activity, and processing of variant spikes for EG.5.1 and XBB.2.3 in HEK293T-ACE2 and CaLu-3 cells but found no significant differences compared to earlier XBB variants. Overall, our findings highlight the continued immune evasion of new Omicron subvariants and, more importantly, the need to reformulate mRNA vaccines to include XBB spikes for better protection.
- Published
- 2023
- Full Text
- View/download PDF
11. Continued evasion of neutralizing antibody response by Omicron XBB.1.16.
- Author
-
Faraone JN, Qu P, Zheng YM, Carlin C, Jones D, Panchal AR, Saif LJ, Oltz EM, Gumina RJ, and Liu SL
- Subjects
- Humans, Antibody Formation, Convalescence, Immune Evasion, SARS-CoV-2, Antibodies, Viral, Antibodies, Neutralizing, COVID-19
- Abstract
The evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to challenge the efficacy of vaccination efforts against coronavirus disease 2019 (COVID-19). The Omicron XBB lineage of SARS-CoV-2 has presented dramatic evasion of neutralizing antibodies stimulated by mRNA vaccination and COVID-19 convalescence. XBB.1.16, characterized by two mutations relative to the dominating variant XBB.1.5, i.e., E180V and K478R, has been on the rise globally. In this study, we compare the immune escape of XBB.1.16 with XBB.1.5, alongside ancestral variants D614G, BA.2, and BA.4/5. We demonstrate that XBB.1.16 is strongly immune evasive, with extent comparable to XBB.1.5 in bivalent-vaccinated healthcare worker sera, 3-dose-vaccinated healthcare worker sera, and BA.4/5-wave convalescent sera. Interestingly, the XBB.1.16 spike is less fusogenic than that of XBB.1.5, and this phenotype requires both E180V and K478R mutations to manifest. Overall, our findings emphasize the importance of the continued surveillance of variants and the need for updated mRNA vaccine formulations., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Immune Evasion, Infectivity, and Fusogenicity of SARS-CoV-2 Omicron BA.2.86 and FLip Variants.
- Author
-
Qu P, Xu K, Faraone JN, Goodarzi N, Zheng YM, Carlin C, Bednash JS, Horowitz JC, Mallampalli RK, Saif LJ, Oltz EM, Jones D, Gumina RJ, and Liu SL
- Abstract
Evolution of SARS-CoV-2 requires the reassessment of current vaccine measures. Here, we characterized BA.2.86 and the XBB-lineage variant FLip by investigating their neutralization alongside D614G, BA.1, BA.2, BA.4/5, XBB.1.5, and EG.5.1 by sera from 3-dose vaccinated and bivalent vaccinated healthcare workers, XBB.1.5-wave infected first responders, and monoclonal antibody (mAb) S309. We assessed the biology of the variant Spikes by measuring viral infectivity and membrane fusogenicity. BA.2.86 is less immune evasive compared to FLip and other XBB variants, consistent with antigenic distances. Importantly, distinct from XBB variants, mAb S309 was unable to neutralize BA.2.86, likely due to a D339H mutation based on modeling. BA.2.86 had relatively high fusogenicity and infectivity in CaLu-3 cells but low fusion and infectivity in 293T-ACE2 cells compared to some XBB variants, suggesting a potentially differences conformational stability of BA.2.86 Spike. Overall, our study underscores the importance of SARS-CoV-2 variant surveillance and the need for updated COVID-19 vaccines., Competing Interests: Declaration of interests The authors do not declare any competing interests.
- Published
- 2023
- Full Text
- View/download PDF
13. Enhanced evasion of neutralizing antibody response by Omicron XBB.1.5, CH.1.1, and CA.3.1 variants.
- Author
-
Qu P, Faraone JN, Evans JP, Zheng YM, Carlin C, Anghelina M, Stevens P, Fernandez S, Jones D, Panchal AR, Saif LJ, Oltz EM, Zhang B, Zhou T, Xu K, Gumina RJ, and Liu SL
- Subjects
- Antibody Formation, Mutation genetics, RNA, Messenger genetics, Vaccines, Combined, Antibodies, Viral, Antibodies, Neutralizing, Antibodies, Monoclonal
- Abstract
Omicron subvariants continuingly challenge current vaccination strategies. Here, we demonstrate nearly complete escape of the XBB.1.5, CH.1.1, and CA.3.1 variants from neutralizing antibodies stimulated by three doses of mRNA vaccine or by BA.4/5 wave infection, but neutralization is rescued by a BA.5-containing bivalent booster. CH.1.1 and CA.3.1 show strong immune escape from monoclonal antibody S309. Additionally, XBB.1.5, CH.1.1, and CA.3.1 spike proteins exhibit increased fusogenicity and enhanced processing compared with BA.2. Homology modeling reveals the key roles of G252V and F486P in the neutralization resistance of XBB.1.5, with F486P also enhancing receptor binding. Further, K444T/M and L452R in CH.1.1 and CA.3.1 likely drive escape from class II neutralizing antibodies, whereas R346T and G339H mutations could confer the strong neutralization resistance of these two subvariants to S309-like antibodies. Overall, our results support the need for administration of the bivalent mRNA vaccine and continued surveillance of Omicron subvariants., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. Neutralization escape of Omicron XBB, BR.2, and BA.2.3.20 subvariants.
- Author
-
Faraone JN, Qu P, Evans JP, Zheng YM, Carlin C, Anghelina M, Stevens P, Fernandez S, Jones D, Lozanski G, Panchal A, Saif LJ, Oltz EM, Gumina RJ, and Liu SL
- Subjects
- Humans, HEK293 Cells, Immunization, Secondary, Mutation, RNA, Messenger, Antibodies
- Abstract
New Omicron subvariants continue to emerge throughout the world. In particular, the XBB subvariant, which is a recombinant virus between BA.2.10.1.1 and BA.2.75.3.1.1.1, as well as the BA.2.3.20 and BR.2 subvariants that contain mutations distinct from BA.2 and BA.2.75, are currently increasing in proportion of variants sequenced. Here we show that antibodies induced by 3-dose mRNA booster vaccination as well as BA.1- and BA.4/5-wave infection effectively neutralize BA.2, BR.2, and BA.2.3.20 but have significantly reduced efficiency against XBB. In addition, the BA.2.3.20 subvariant exhibits enhanced infectivity in the lung-derived CaLu-3 cells and in 293T-ACE2 cells. Overall, our results demonstrate that the XBB subvariant is highly neutralization resistant, which highlights the need for continued monitoring of the immune escape and tissue tropism of emerging Omicron subvariants., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
15. Systems Approaches for Studying Immunity.
- Author
-
Vahedi G and Oltz EM
- Subjects
- Systems Biology, Immunity
- Published
- 2023
- Full Text
- View/download PDF
16. Selective suppression of de novo SARS-CoV-2 vaccine antibody responses in patients with cancer on B cell-targeted therapy.
- Author
-
Azar JH, Evans JP, Sikorski MH, Chakravarthy KB, McKenney S, Carmody I, Zeng C, Teodorescu R, Song NJ, Hamon JL, Bucci D, Velegraki M, Bolyard C, Weller KP, Reisinger SA, Bhat SA, Maddocks KJ, Denlinger N, Epperla N, Gumina RJ, Vlasova AN, Oltz EM, Saif LJ, Chung D, Woyach JA, Shields PG, Liu SL, Li Z, and Rubinstein MP
- Subjects
- Humans, COVID-19 Vaccines, Antibody Formation, SARS-CoV-2, Antibodies, Monoclonal, Antibodies, Viral, COVID-19, Neoplasms therapy
- Abstract
We assessed vaccine-induced antibody responses to the SARS-CoV-2 ancestral virus and Omicron variant before and after booster immunization in 57 patients with B cell malignancies. Over one-third of vaccinated patients at the pre-booster time point were seronegative, and these patients were predominantly on active cancer therapies such as anti-CD20 monoclonal antibody. While booster immunization was able to induce detectable antibodies in a small fraction of seronegative patients, the overall booster benefit was disproportionately evident in patients already seropositive and not receiving active therapy. While ancestral virus- and Omicron variant-reactive antibody levels among individual patients were largely concordant, neutralizing antibodies against Omicron tended to be reduced. Interestingly, in all patients, including those unable to generate detectable antibodies against SARS-CoV-2 spike, we observed comparable levels of EBV- and influenza-reactive antibodies, demonstrating that B cell-targeting therapies primarily impair de novo but not preexisting antibody levels. These findings support rationale for vaccination before cancer treatment.
- Published
- 2023
- Full Text
- View/download PDF
17. Enhanced neutralization resistance of SARS-CoV-2 Omicron subvariants BQ.1, BQ.1.1, BA.4.6, BF.7, and BA.2.75.2.
- Author
-
Qu P, Evans JP, Faraone JN, Zheng YM, Carlin C, Anghelina M, Stevens P, Fernandez S, Jones D, Lozanski G, Panchal A, Saif LJ, Oltz EM, Xu K, Gumina RJ, and Liu SL
- Subjects
- Humans, SARS-CoV-2 genetics, Antibodies, Immune Evasion, Mutation, Antibodies, Neutralizing, COVID-19
- Abstract
The continued evolution of SARS-CoV-2 has led to the emergence of several new Omicron subvariants, including BQ.1, BQ.1.1, BA.4.6, BF.7, and BA.2.75.2. Here, we examine the neutralization resistance of these subvariants against sera from 3-dose vaccinated healthcare workers, hospitalized BA.1-wave patients, and BA.4/5-wave patients. We found enhanced neutralization resistance in all new subvariants, especially in the BQ.1 and BQ.1.1 subvariants driven by N460K and K444T mutations, as well as the BA.2.75.2 subvariant driven largely by its F486S mutation. All Omicron subvariants maintained their weakened infectivity in Calu-3 cells, with the F486S mutation driving further diminished titer for the BA.2.75.2 subvariant. Molecular modeling revealed the mechanisms of antibody-mediated immune evasion by R346T, K444T, F486S, and D1199N mutations. Altogether, these findings shed light on the evolution of newly emerging SARS-CoV-2 Omicron subvariants., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. Evasion of neutralizing antibody responses by the SARS-CoV-2 BA.2.75 variant.
- Author
-
Qu P, Evans JP, Zheng YM, Carlin C, Saif LJ, Oltz EM, Xu K, Gumina RJ, and Liu SL
- Subjects
- Humans, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus genetics, Neutralization Tests, Antibodies, Viral, Viral Envelope Proteins, Antibodies, Neutralizing, COVID-19
- Abstract
The newly emerged BA.2.75 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant contains 9 additional mutations in its spike (S) protein compared to the ancestral BA.2 variant. Here, we examine the neutralizing antibody escape of BA.2.75 in mRNA-vaccinated and BA.1-infected individuals, as well as the molecular basis underlying functional changes in S. Notably, BA.2.75 exhibits enhanced neutralization resistance over BA.2 but less than the BA.4/5 variant. The G446S and N460K mutations of BA.2.75 are primarily responsible for its enhanced resistance to neutralizing antibodies. The R493Q mutation, a reversion to the prototype sequence, reduces BA.2.75 neutralization resistance. The impact of these mutations is consistent with their locations in common neutralizing antibody epitopes. Further, BA.2.75 shows enhanced cell-cell fusion over BA.2, driven largely by the N460K mutation, which enhances S processing. Structural modeling reveals enhanced receptor contacts introduced by N460K, suggesting a mechanism of potentiated receptor utilization and syncytia formation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
19. Distinct Neutralizing Antibody Escape of SARS-CoV-2 Omicron Subvariants BQ.1, BQ.1.1, BA.4.6, BF.7 and BA.2.75.2.
- Author
-
Qu P, Evans JP, Faraone J, Zheng YM, Carlin C, Anghelina M, Stevens P, Fernandez S, Jones D, Lozanski G, Panchal A, Saif LJ, Oltz EM, Xu K, Gumina RJ, and Liu SL
- Abstract
Continued evolution of SARS-CoV-2 has led to the emergence of several new Omicron subvariants, including BQ.1, BQ. 1.1, BA.4.6, BF.7 and BA.2.75.2. Here we examine the neutralization resistance of these subvariants, as well as their ancestral BA.4/5, BA.2.75 and D614G variants, against sera from 3-dose vaccinated health care workers, hospitalized BA.1-wave patients, and BA.5-wave patients. We found enhanced neutralization resistance in all new subvariants, especially the BQ.1 and BQ.1.1 subvariants driven by a key N460K mutation, and to a lesser extent, R346T and K444T mutations, as well as the BA.2.75.2 subvariant driven largely by its F486S mutation. The BQ.1 and BQ.1.1 subvariants also exhibited enhanced fusogenicity and S processing dictated by the N460K mutation. Interestingly, the BA.2.75.2 subvariant saw an enhancement by the F486S mutation and a reduction by the D1199N mutation to its fusogenicity and S processing, resulting in minimal overall change. Molecular modelling revealed the mechanisms of receptor-binding and non-receptor binding monoclonal antibody-mediated immune evasion by R346T, K444T, F486S and D1199N mutations. Altogether, these findings shed light on the concerning evolution of newly emerging SARS-CoV-2 Omicron subvariants.
- Published
- 2022
- Full Text
- View/download PDF
20. Durability of Booster mRNA Vaccine against SARS-CoV-2 BA.2.12.1, BA.4, and BA.5 Subvariants.
- Author
-
Qu P, Faraone JN, Evans JP, Zheng YM, Yu L, Ma Q, Carlin C, Lozanski G, Saif LJ, Oltz EM, Gumina RJ, and Liu SL
- Subjects
- Humans, RNA, Messenger, Vaccine Efficacy, Vaccines, Synthetic immunology, Vaccines, Synthetic therapeutic use, mRNA Vaccines immunology, mRNA Vaccines therapeutic use, COVID-19 etiology, COVID-19 prevention & control, COVID-19 Vaccines immunology, COVID-19 Vaccines therapeutic use, Immunization, Secondary, SARS-CoV-2 genetics, SARS-CoV-2 immunology
- Published
- 2022
- Full Text
- View/download PDF
21. Neutralization of SARS-CoV-2 Omicron sub-lineages BA.1, BA.1.1, and BA.2.
- Author
-
Evans JP, Zeng C, Qu P, Faraone J, Zheng YM, Carlin C, Bednash JS, Zhou T, Lozanski G, Mallampalli R, Saif LJ, Oltz EM, Mohler PJ, Xu K, Gumina RJ, and Liu SL
- Subjects
- Antibodies, Neutralizing, Antibodies, Viral, Humans, Neutralization Tests, Spike Glycoprotein, Coronavirus genetics, Vaccines, Synthetic, mRNA Vaccines, COVID-19, SARS-CoV-2 genetics
- Abstract
Recent reports of SARS-CoV-2 Omicron variant sub-lineages, BA.1, BA.1.1, and BA.2, have reignited concern over potential escape from vaccine- and infection-induced immunity. We examine the sensitivity of these sub-lineages and other major variants to neutralizing antibodies from mRNA-vaccinated and boosted individuals, as well as recovered COVID-19 patients, including those infected with Omicron. We find that all Omicron sub-lineages, especially BA.1 and BA.1.1, exhibit substantial immune escape that is largely overcome by mRNA vaccine booster doses. While Omicron BA.1.1 escapes almost completely from neutralization by early-pandemic COVID-19 patient sera and to a lesser extent from sera of Delta-infected patients, BA.1.1 is sensitive to Omicron-infected patient sera. Critically, all Omicron sub-lineages, including BA.2, are comparably neutralized by Omicron patient sera. These results highlight the importance of booster vaccine doses for protection against all Omicron variants and provide insight into the immunity from natural infection against Omicron sub-lineages., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
22. Durability of the Neutralizing Antibody Response to mRNA Booster Vaccination Against SARS-CoV-2 BA.2.12.1 and BA.4/5 Variants.
- Author
-
Qu P, Faraone JN, Evans JP, Zheng YM, Carlin C, Lozanski G, Saif LJ, Oltz EM, Gumina RJ, and Liu SL
- Abstract
The recent emergence of the SARS-CoV-2 BA.4/5 and BA.2.12.1 variants has led to rising COVID-19 case numbers and concerns over the continued efficacy of mRNA booster vaccination. Here we examine the durability of neutralizing antibody (nAb) responses against these SARS-CoV-2 Omicron subvariants in a cohort of health care workers 1-40 weeks after mRNA booster dose administration. Neutralizing antibody titers fell by ~1.5-fold 4-6 months and by ~2.5-fold 7-9 months after booster dose, with average nAb titers falling by 11-15% every 30 days, far more stable than two dose induced immunity. Notably, nAb titers from booster recipients against SARS-CoV-2 BA.1, BA.2.12.1, and BA.4/5 variants were ~4.7-, 7.6-, and 13.4-fold lower than against the ancestral D614G spike. However, the rate of waning of booster dose immunity was comparable across variants. Importantly, individuals reporting prior infection with SARS-CoV-2 exhibited significantly higher nAb titers compared to those without breakthrough infection. Collectively, these results highlight the broad and stable neutralizing antibody response induced by mRNA booster dose administration, implicating a significant role of virus evolution to evade nAb specificity, versus waning humoral immunity, in increasing rates of breakthrough infection., Competing Interests: Declaration of Interests The authors declare no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
23. Neutralization of the SARS-CoV-2 Omicron BA.4/5 and BA.2.12.1 Subvariants.
- Author
-
Qu P, Faraone J, Evans JP, Zou X, Zheng YM, Carlin C, Bednash JS, Lozanski G, Mallampalli RK, Saif LJ, Oltz EM, Mohler PJ, Gumina RJ, and Liu SL
- Subjects
- Antibodies, Neutralizing, Humans, Neutralization Tests, Spike Glycoprotein, Coronavirus immunology, COVID-19 diagnosis, COVID-19 immunology, COVID-19 virology, SARS-CoV-2 immunology
- Published
- 2022
- Full Text
- View/download PDF
24. Neutralization of the SARS-CoV-2 Deltacron and BA.3 Variants.
- Author
-
Evans JP, Qu P, Zeng C, Zheng YM, Carlin C, Bednash JS, Lozanski G, Mallampalli RK, Saif LJ, Oltz EM, Mohler PJ, Gumina RJ, and Liu SL
- Subjects
- Antibodies, Viral, Humans, Neutralization Tests, Spike Glycoprotein, Coronavirus immunology, Antibodies, Neutralizing immunology, COVID-19 immunology, COVID-19 therapy, COVID-19 virology, SARS-CoV-2
- Published
- 2022
- Full Text
- View/download PDF
25. Gene Regulatory Circuits in Innate and Adaptive Immune Cells.
- Author
-
Saini A, Ghoneim HE, Lio CJ, Collins PL, and Oltz EM
- Subjects
- Animals, Gene Expression Regulation, Humans, Promoter Regions, Genetic, Transcription Factors genetics, Chromatin, Gene Regulatory Networks
- Abstract
Cell identity and function largely rely on the programming of transcriptomes during development and differentiation. Signature gene expression programs are orchestrated by regulatory circuits consisting of cis- acting promoters and enhancers, which respond to a plethora of cues via the action of transcription factors. In turn, transcription factors direct epigenetic modifications to revise chromatin landscapes, and drive contacts between distal promoter-enhancer combinations. In immune cells, regulatory circuits for effector genes are especially complex and flexible, utilizing distinct sets of transcription factors and enhancers, depending on the cues each cell type receives during an infection, after sensing cellular damage, or upon encountering a tumor. Here, we review major players in the coordination of gene regulatory programs within innate and adaptive immune cells, as well as integrative omics approaches that can be leveraged to decipher their underlying circuitry.
- Published
- 2022
- Full Text
- View/download PDF
26. Whole-genome profiling of DNA methylation and hydroxymethylation identifies distinct regulatory programs among innate lymphocytes.
- Author
-
Peng V, Xing X, Bando JK, Trsan T, Di Luccia B, Collins PL, Li D, Wang WL, Lee HJ, Oltz EM, Wang T, and Colonna M
- Subjects
- Animals, Chromatin genetics, Epigenesis, Genetic, Killer Cells, Natural, Lymphocytes, Mice, DNA Methylation, Immunity, Innate genetics
- Abstract
Innate lymphocytes encompass a diverse array of phenotypic identities with specialized functions. DNA methylation and hydroxymethylation are essential for epigenetic fidelity and fate commitment. The landscapes of these modifications are unknown in innate lymphocytes. Here, we characterized the whole-genome distribution of methyl-CpG and 5-hydroxymethylcytosine (5hmC) in mouse innate lymphoid cell 3 (ILC3), ILC2 and natural killer (NK) cells. We identified differentially methylated regions (DMRs) and differentially hydroxymethylated regions (DHMRs) between ILC and NK cell subsets and correlated them with transcriptional signatures. We associated lineage-determining transcription factors (LDTFs) with demethylation and demonstrated unique patterns of DNA methylation/hydroxymethylation in relationship to open chromatin regions (OCRs), histone modifications and TF-binding sites. We further identified an association between hydroxymethylation and NK cell superenhancers (SEs). Using mice lacking the DNA hydroxymethylase TET2, we showed the requirement for TET2 in optimal production of hallmark cytokines by ILC3s and interleukin-17A (IL-17A) by inflammatory ILC2s. These findings provide a powerful resource for studying innate lymphocyte epigenetic regulation and decode the regulatory logic governing their identity., (© 2022. Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
27. Neutralizing antibody responses elicited by SARS-CoV-2 mRNA vaccination wane over time and are boosted by breakthrough infection.
- Author
-
Evans JP, Zeng C, Carlin C, Lozanski G, Saif LJ, Oltz EM, Gumina RJ, and Liu SL
- Subjects
- Antibodies, Neutralizing, Antibodies, Viral, BNT162 Vaccine, COVID-19 Vaccines, Humans, RNA, Messenger genetics, Vaccination, Vaccines, Synthetic, mRNA Vaccines, COVID-19 prevention & control, SARS-CoV-2
- Abstract
The waning efficacy of SARS-CoV-2 vaccines, combined with the continued emergence of variants resistant to vaccine-induced immunity, has reignited debate over the need for booster vaccine doses. To address this, we examined the neutralizing antibody response against the spike protein of five major SARS-CoV-2 variants, D614G, Alpha (B.1.1.7), Beta (B.1.351), Delta (B.1.617.2), and Omicron (B.1.1.529), in health care workers (HCWs) vaccinated with SARS-CoV-2 mRNA vaccines. Serum samples were collected before vaccination, 3 weeks after first vaccination, 1 month after second vaccination, and 6 months after second vaccination. Minimal neutralizing antibody titers were detected against Omicron pseudovirus at all four time points, including for most patients who had SARS-CoV-2 breakthrough infections. Neutralizing antibody titers against all other variant spike protein-bearing pseudoviruses declined markedly from 1 to 6 months after the second mRNA vaccine dose, although SARS-CoV-2 infection boosted vaccine responses. In addition, mRNA-1273-vaccinated HCWs exhibited about twofold higher neutralizing antibody titers than BNT162b2-vaccinated HCWs. Together, these results demonstrate possible waning of antibody-mediated protection against SARS-CoV-2 variants that is dependent on prior infection status and the mRNA vaccine received. They also show that the Omicron variant spike protein can almost completely escape from neutralizing antibodies elicited in recipients of only two mRNA vaccine doses.
- Published
- 2022
- Full Text
- View/download PDF
28. SARS-CoV-2 spreads through cell-to-cell transmission.
- Author
-
Zeng C, Evans JP, King T, Zheng YM, Oltz EM, Whelan SPJ, Saif LJ, Peeples ME, and Liu SL
- Subjects
- Angiotensin-Converting Enzyme 2, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral, COVID-19 therapy, Cell Fusion, Chlorocebus aethiops, HEK293 Cells, Humans, Immunization, Passive, Spike Glycoprotein, Coronavirus immunology, Vero Cells, COVID-19 Serotherapy, COVID-19 immunology, COVID-19 transmission, SARS-CoV-2 immunology, Virus Internalization
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly transmissible coronavirus responsible for the global COVID-19 pandemic. Herein, we provide evidence that SARS-CoV-2 spreads through cell-cell contact in cultures, mediated by the spike glycoprotein. SARS-CoV-2 spike is more efficient in facilitating cell-to-cell transmission than is SARS-CoV spike, which reflects, in part, their differential cell-cell fusion activity. Interestingly, treatment of cocultured cells with endosomal entry inhibitors impairs cell-to-cell transmission, implicating endosomal membrane fusion as an underlying mechanism. Compared with cell-free infection, cell-to-cell transmission of SARS-CoV-2 is refractory to inhibition by neutralizing antibody or convalescent sera of COVID-19 patients. While angiotensin-converting enzyme 2 enhances cell-to-cell transmission, we find that it is not absolutely required. Notably, despite differences in cell-free infectivity, the authentic variants of concern (VOCs) B.1.1.7 (alpha) and B.1.351 (beta) have similar cell-to-cell transmission capability. Moreover, B.1.351 is more resistant to neutralization by vaccinee sera in cell-free infection, whereas B.1.1.7 is more resistant to inhibition by vaccinee sera in cell-to-cell transmission. Overall, our study reveals critical features of SARS-CoV-2 spike-mediated cell-to-cell transmission, with important implications for a better understanding of SARS-CoV-2 spread and pathogenesis., Competing Interests: The authors declare no competing interest., (Copyright © 2021 the Author(s). Published by PNAS.)
- Published
- 2022
- Full Text
- View/download PDF
29. Erratum for Zeng et al., "Neutralization of SARS-CoV-2 Variants of Concern Harboring Q677H".
- Author
-
Zeng C, Evans JP, Faraone JN, Qu P, Zheng YM, Saif L, Oltz EM, Lozanski G, Gumina RJ, and Liu SL
- Published
- 2021
- Full Text
- View/download PDF
30. Neutralization and Stability of SARS-CoV-2 Omicron Variant.
- Author
-
Zeng C, Evans JP, Qu P, Faraone J, Zheng YM, Carlin C, Bednash JS, Zhou T, Lozanski G, Mallampalli R, Saif LJ, Oltz EM, Mohler P, Xu K, Gumina RJ, and Liu SL
- Abstract
The SARS-CoV-2 B.1.1.529/Omicron variant was first characterized in South Africa and was swiftly designated a variant of concern
1 . Of great concern is its high number of mutations, including 30-40 mutations in the virus spike (S) protein compared to 7-10 for other variants. Some of these mutations have been shown to enhance escape from vaccine-induced immunity, while others remain uncharacterized. Additionally, reports of increasing frequencies of the Omicron variant may indicate a higher rate of transmission compared to other variants. However, the transmissibility of Omicron and its degree of resistance to vaccine-induced immunity remain unclear. Here we show that Omicron exhibits significant immune evasion compared to other variants, but antibody neutralization is largely restored by mRNA vaccine booster doses. Additionally, the Omicron spike exhibits reduced receptor binding, cell-cell fusion, S1 subunit shedding, but increased cell-to-cell transmission, and homology modeling indicates a more stable closed S structure. These findings suggest dual immune evasion strategies for Omicron, due to altered epitopes and reduced exposure of the S receptor binding domain, coupled with enhanced transmissibility due to enhanced S protein stability. These results highlight the importance of booster vaccine doses for maintaining protection against the Omicron variant, and provide mechanistic insight into the altered functionality of the Omicron spike protein., Competing Interests: Competing Interest Declaration: The authors have no competing interests to disclose- Published
- 2021
- Full Text
- View/download PDF
31. Loss of Neutralizing Antibody Response to mRNA Vaccination against SARS-CoV-2 Variants: Differing Kinetics and Strong Boosting by Breakthrough Infection.
- Author
-
Evans JP, Zeng C, Carlin C, Lozanski G, Saif LJ, Oltz EM, Gumina RJ, and Liu SL
- Abstract
The waning efficacy of SARS-CoV-2 vaccines combined with the continued emergence of variants resistant to vaccine-induced immunity has reignited debate over the need for booster vaccines. To address this, we examined the neutralizing antibody (nAb) response against four major SARS-CoV-2 variants-D614G, Alpha (B.1.1.7), Beta (B.1.351), and Delta (B.1.617.2)-in health care workers (HCWs) at pre-vaccination, post-first and post-second mRNA vaccine dose, and six months post-second mRNA vaccine dose. Neutralizing antibody titers against all variants, especially the Delta variant, declined dramatically from four weeks to six months post-second mRNA vaccine dose. Notably, SARS-CoV-2 infection enhanced vaccine durability, and mRNA-1273 vaccinated HCWs also exhibited ~2-fold higher nAb titers than BNT162b2 vaccinated HCWs. Together these results demonstrate possible waning of protection from infection against SARS-CoV-2 Delta variant based on decreased nAb titers, dependent on COVID-19 status and the mRNA vaccine received., Competing Interests: Competing Interests: The authors declare no competing interest.
- Published
- 2021
- Full Text
- View/download PDF
32. Impaired neutralizing antibody response to COVID-19 mRNA vaccines in cancer patients.
- Author
-
Zeng C, Evans JP, Reisinger S, Woyach J, Liscynesky C, Boghdadly ZE, Rubinstein MP, Chakravarthy K, Saif L, Oltz EM, Gumina RJ, Shields PG, Li Z, and Liu SL
- Abstract
There is currently a critical need to determine the efficacy of SARS-CoV-2 vaccination for immunocompromised patients. In this study, we determined the neutralizing antibody response in 160 cancer patients diagnosed with chronic lymphocytic leukemia (CLL), lung cancer, breast cancer, and various non-Hodgkin's lymphomas (NHL), after they received two doses of mRNA vaccines. Serum from 46 mRNA vaccinated health care workers (HCWs) served as healthy controls. We discovered that (1) cancer patients exhibited reduced neutralizing antibody titer (NT
50 ) compared to HCWs; (2) CLL and NHL patients exhibited the lowest NT50 levels, with 50-60% of them below the detection limit; (3) mean NT50 levels in patients with CLL and NHL was ~2.6 fold lower than those with solid tumors; and (4) cancer patients who received anti-B cell therapy exhibited significantly reduced NT50 levels. Our results demonstrate an urgent need for novel immunization strategies for cancer patients against SARS-CoV-2, particularly those with hematological cancers and those on anti-B cell therapies., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
33. Neutralization of SARS-CoV-2 Variants of Concern Harboring Q677H.
- Author
-
Zeng C, Evans JP, Faraone JN, Qu P, Zheng YM, Saif L, Oltz EM, Lozanski G, Gumina RJ, and Liu SL
- Subjects
- Antibodies, Viral metabolism, HEK293 Cells, Humans, Mutation, Protein Binding, SARS-CoV-2 metabolism, Spike Glycoprotein, Coronavirus metabolism, Antibodies, Neutralizing metabolism, COVID-19 metabolism, SARS-CoV-2 pathogenicity, Spike Glycoprotein, Coronavirus immunology
- Abstract
The sensitivity of SARS-CoV-2 variants of concern (VOCs) to neutralizing antibodies has largely been studied in the context of key receptor binding domain (RBD) mutations, including E484K and N501Y. Little is known about the epistatic effects of combined SARS-CoV-2 spike mutations. We now investigate the neutralization sensitivity of variants containing the non-RBD mutation Q677H, including B.1.525 (Nigerian isolate) and Bluebird (U.S. isolate) variants. The effect on neutralization of Q677H was determined in the context of the RBD mutations and in the background of major VOCs, including B.1.1.7 (United Kingdom, Alpha), B.1.351 (South Africa, Beta), and P1-501Y-V3 (Brazil, Gamma). We demonstrate that the Q677H mutation increases viral infectivity and syncytium formation, as well as enhancing resistance to neutralization for VOCs, including B.1.1.7 and P1-501Y-V3. Our work highlights the importance of epistatic interactions between SARS-CoV-2 spike mutations and the continued need to monitor Q677H-bearing VOCs. IMPORTANCE SARS-CoV-2, the causative agent of COVID-19, is rapidly evolving to be more transmissible and to evade acquired immunity. To date, most investigations of SARS-CoV-2 variants have focused on RBD mutations. However, the impact of non-RBD mutations and their synergy with studied RBD mutations are poorly understood. Here, we examine the role of the non-RBD Q677H mutation arising in many SARS-CoV-2 lineages, including VOCs. We demonstrate that the Q677H mutation enhances viral infectivity and confers neutralizing antibody resistance, particularly in the background of other SARS-CoV-2 VOCs.
- Published
- 2021
- Full Text
- View/download PDF
34. Loss of synergistic transcriptional feedback loops drives diverse B-cell cancers.
- Author
-
Andrews JM, Pyfrom SC, Schmidt JA, Koues OI, Kowalewski RA, Grams NR, Sun JJ, Berman LR, Duncavage EJ, Lee YS, Cashen AF, Oltz EM, and Payton JE
- Subjects
- Adult, Aged, Aged, 80 and over, B-Lymphocytes immunology, Biomarkers, Cell Transformation, Neoplastic metabolism, Chromatin Immunoprecipitation Sequencing, Computational Biology methods, DNA Copy Number Variations, Enhancer Elements, Genetic, Epigenesis, Genetic, Female, Gene Expression Profiling, Humans, Immunophenotyping, Leukemia, B-Cell diagnosis, Leukemia, B-Cell metabolism, Lymphoma, B-Cell diagnosis, Lymphoma, B-Cell metabolism, Male, Middle Aged, Models, Biological, Oncogenes, Signal Transduction, Transcription Factors metabolism, B-Lymphocytes metabolism, Cell Transformation, Neoplastic genetics, Gene Expression Regulation, Neoplastic, Leukemia, B-Cell etiology, Lymphoma, B-Cell etiology, Transcription, Genetic
- Abstract
Background: The most common B-cell cancers, chronic lymphocytic leukemia/lymphoma (CLL), follicular and diffuse large B-cell (FL, DLBCL) lymphomas, have distinct clinical courses, yet overlapping "cell-of-origin". Dynamic changes to the epigenome are essential regulators of B-cell differentiation. Therefore, we reasoned that these distinct cancers may be driven by shared mechanisms of disruption in transcriptional circuitry., Methods: We compared purified malignant B-cells from 52 patients with normal B-cell subsets (germinal center centrocytes and centroblasts, naïve and memory B-cells) from 36 donor tonsils using >325 high-resolution molecular profiling assays for histone modifications, open chromatin (ChIP-, FAIRE-seq), transcriptome (RNA-seq), transcription factor (TF) binding, and genome copy number (microarrays)., Findings: From the resulting data, we identified gains in active chromatin in enhancers/super-enhancers that likely promote unchecked B-cell receptor signaling, including one we validated near the immunoglobulin superfamily receptors FCMR and PIGR. More striking and pervasive was the profound loss of key B-cell identity TFs, tumor suppressors and their super-enhancers, including EBF1, OCT2(POU2F2), and RUNX3. Using a novel approach to identify transcriptional feedback, we showed that these core transcriptional circuitries are self-regulating. Their selective gain and loss form a complex, iterative, and interactive process that likely curbs B-cell maturation and spurs proliferation., Interpretation: Our study is the first to map the transcriptional circuitry of the most common blood cancers. We demonstrate that a critical subset of B-cell TFs and their cognate enhancers form self-regulatory transcriptional feedback loops whose disruption is a shared mechanism underlying these diverse subtypes of B-cell lymphoma., Funding: National Institute of Health, Siteman Cancer Center, Barnes-Jewish Hospital Foundation, Doris Duke Foundation., Competing Interests: Declaration of Competing Interest The authors declare no potential conflicts of interest., (Copyright © 2021 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
35. SARS-CoV-2 Spreads through Cell-to-Cell Transmission.
- Author
-
Zeng C, Evans JP, King T, Zheng YM, Oltz EM, Whelan SPJ, Saif L, Peeples ME, and Liu SL
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly transmissible coronavirus responsible for the global COVID-19 pandemic. Herein we provide evidence that SARS-CoV-2 spreads through cell-cell contact in cultures, mediated by the spike glycoprotein. SARS-CoV-2 spike is more efficient in facilitating cell-to-cell transmission than SARS-CoV spike, which reflects, in part, their differential cell-cell fusion activity. Interestingly, treatment of cocultured cells with endosomal entry inhibitors impairs cell-to-cell transmission, implicating endosomal membrane fusion as an underlying mechanism. Compared with cell-free infection, cell-to-cell transmission of SARS-CoV-2 is refractory to inhibition by neutralizing antibody or convalescent sera of COVID-19 patients. While ACE2 enhances cell-to-cell transmission, we find that it is not absolutely required. Notably, despite differences in cell-free infectivity, the variants of concern (VOC) B.1.1.7 and B.1.351 have similar cell-to-cell transmission capability. Moreover, B.1.351 is more resistant to neutralization by vaccinee sera in cell-free infection, whereas B.1.1.7 is more resistant to inhibition by vaccine sera in cell-to-cell transmission. Overall, our study reveals critical features of SARS-CoV-2 spike-mediated cell-to-cell transmission, with important implications for a better understanding of SARS-CoV-2 spread and pathogenesis.
- Published
- 2021
- Full Text
- View/download PDF
36. Cancer-associated exportin-6 upregulation inhibits the transcriptionally repressive and anticancer effects of nuclear profilin-1.
- Author
-
Zhu C, Kim SJ, Mooradian A, Wang F, Li Z, Holohan S, Collins PL, Wang K, Guo Z, Hoog J, Ma CX, Oltz EM, Held JM, and Shao J
- Subjects
- Animals, Cell Line, Tumor, Female, Heterografts, Humans, Karyopherins genetics, MCF-7 Cells, Mice, Mice, Nude, Neoplasms genetics, Profilins genetics, Survival Analysis, Up-Regulation, Karyopherins metabolism, Neoplasms metabolism, Profilins antagonists & inhibitors, Profilins metabolism
- Abstract
Aberrant expression of nuclear transporters and deregulated subcellular localization of their cargo proteins are emerging as drivers and therapeutic targets of cancer. Here, we present evidence that the nuclear exporter exportin-6 and its cargo profilin-1 constitute a functionally important and frequently deregulated axis in cancer. Exportin-6 upregulation occurs in numerous cancer types and is associated with poor patient survival. Reducing exportin-6 level in breast cancer cells triggers antitumor effects by accumulating nuclear profilin-1. Mechanistically, nuclear profilin-1 interacts with eleven-nineteen-leukemia protein (ENL) within the super elongation complex (SEC) and inhibits the ability of the SEC to drive transcription of numerous pro-cancer genes including MYC. XPO6 and MYC are positively correlated across diverse cancer types including breast cancer. Therapeutically, exportin-6 loss sensitizes breast cancer cells to the bromodomain and extra-terminal (BET) inhibitor JQ1. Thus, exportin-6 upregulation is a previously unrecognized cancer driver event by spatially inhibiting nuclear profilin-1 as a tumor suppressor., Competing Interests: Declaration of interests No authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
37. Stromal Immunology: Frameworks for Development and Response.
- Author
-
Oltz EM and Schwab S
- Subjects
- Animals, Cell Differentiation, Cell Movement, Cellular Microenvironment, Humans, Integrases genetics, Integrases metabolism, Lymphoid Tissue cytology, Neoplasms immunology, Stromal Cells immunology, T-Lymphocytes immunology
- Published
- 2021
- Full Text
- View/download PDF
38. Enhanced epigenetic profiling of classical human monocytes reveals a specific signature of healthy aging in the DNA methylome.
- Author
-
Shchukina I, Bagaitkar J, Shpynov O, Loginicheva E, Porter S, Mogilenko DA, Wolin E, Collins P, Demidov G, Artomov M, Zaitsev K, Sidorov S, Camell C, Bambouskova M, Arthur L, Swain A, Panteleeva A, Dievskii A, Kurbatsky E, Tsurinov P, Chernyatchik R, Dixit VD, Jovanovic M, Stewart SA, Daly MJ, Dmitriev S, Oltz EM, and Artyomov MN
- Subjects
- Male, Humans, Middle Aged, Epigenome, Monocytes, Proteomics, DNA Methylation genetics, Epigenesis, Genetic, Healthy Aging
- Abstract
The impact of healthy aging on molecular programming of immune cells is poorly understood. Here, we report comprehensive characterization of healthy aging in human classical monocytes, with a focus on epigenomic, transcriptomic, and proteomic alterations, as well as the corresponding proteomic and metabolomic data for plasma, using healthy cohorts of 20 young and 20 older males (~27 and ~64 years old on average). For each individual, we performed eRRBS-based DNA methylation profiling, which allowed us to identify a set of age-associated differentially methylated regions (DMRs) - a novel, cell-type specific signature of aging in DNA methylome. Hypermethylation events were associated with H3K27me3 in the CpG islands near promoters of lowly-expressed genes, while hypomethylated DMRs were enriched in H3K4me1 marked regions and associated with age-related increase of expression of the corresponding genes, providing a link between DNA methylation and age-associated transcriptional changes in primary human cells., Competing Interests: COMPETING INTERESTS The authors declare no competing interests
- Published
- 2021
- Full Text
- View/download PDF
39. DNA double-strand breaks induce H2Ax phosphorylation domains in a contact-dependent manner.
- Author
-
Collins PL, Purman C, Porter SI, Nganga V, Saini A, Hayer KE, Gurewitz GL, Sleckman BP, Bednarski JJ, Bassing CH, and Oltz EM
- Subjects
- Animals, Cell Line, Transformed, Chromatin metabolism, Mice, Phosphorylation, DNA Breaks, Double-Stranded, DNA Damage, DNA Repair, Histones metabolism
- Abstract
Efficient repair of DNA double-strand breaks (DSBs) requires a coordinated DNA Damage Response (DDR), which includes phosphorylation of histone H2Ax, forming γH2Ax. This histone modification spreads beyond the DSB into neighboring chromatin, generating a DDR platform that protects against end disassociation and degradation, minimizing chromosomal rearrangements. However, mechanisms that determine the breadth and intensity of γH2Ax domains remain unclear. Here, we show that chromosomal contacts of a DSB site are the primary determinants for γH2Ax landscapes. DSBs that disrupt a topological border permit extension of γH2Ax domains into both adjacent compartments. In contrast, DSBs near a border produce highly asymmetric DDR platforms, with γH2Ax nearly absent from one broken end. Collectively, our findings lend insights into a basic DNA repair mechanism and how the precise location of a DSB may influence genome integrity.
- Published
- 2020
- Full Text
- View/download PDF
40. Barrier-to-Autointegration Factor 1 Protects against a Basal cGAS-STING Response.
- Author
-
Ma H, Qian W, Bambouskova M, Collins PL, Porter SI, Byrum AK, Zhang R, Artyomov M, Oltz EM, Mosammaparast N, Miner JJ, and Diamond MS
- Subjects
- Animals, CRISPR-Cas Systems, Cell Line, DNA-Binding Proteins genetics, Gene Editing, Gene Expression Regulation, Homeostasis immunology, Humans, Immunity, Innate, Interferons immunology, Mice, Microglia immunology, Nuclear Proteins genetics, Signal Transduction, DNA-Binding Proteins immunology, Host-Pathogen Interactions, Membrane Proteins immunology, Nuclear Proteins immunology, Nucleotidyltransferases immunology
- Abstract
Although the pathogen recognition receptor pathways that activate cell-intrinsic antiviral responses are well delineated, less is known about how the host regulates this response to prevent sustained signaling and possible immune-mediated damage. Using a genome-wide CRISPR-Cas9 screening approach to identify host factors that modulate interferon-stimulated gene (ISG) expression, we identified the DNA binding protein Barrier-to-autointegration factor 1 (Banf1), a previously described inhibitor of retrovirus integration, as a modulator of basal cell-intrinsic immunity. Ablation of Banf1 by gene editing resulted in chromatin activation near host defense genes with associated increased expression of ISGs, including Oas2 , Rsad2 (viperin), Ifit1 , and ISG15 The phenotype in Banf1-deficient cells occurred through a cGAS-, STING-, and IRF3-dependent signaling axis, was associated with reduced infection of RNA and DNA viruses, and was reversed in Banf1 complemented cells. Confocal microscopy and biochemical studies revealed that a loss of Banf1 expression resulted in higher level of cytosolic double-stranded DNA at baseline. Our study identifies an undescribed role for Banf1 in regulating the levels of cytoplasmic DNA and cGAS-dependent ISG homeostasis and suggests possible therapeutic directions for promoting or inhibiting cell-intrinsic innate immune responses. IMPORTANCE Although the interferon (IFN) signaling pathway is a key host mechanism to restrict infection of a diverse range of viral pathogens, its unrestrained activity either at baseline or in the context of an immune response can result in host cell damage and injury. Here, we used a genome-wide CRISPR-Cas9 screen and identified the DNA binding protein Barrier-to-autointegration factor 1 (Banf1) as a modulator of basal cell-intrinsic immunity. A loss of Banf1 expression resulted in higher level of cytosolic double-stranded DNA at baseline, which triggered IFN-stimulated gene expression via a cGAS-STING-IRF3 axis that did not require type I IFN or STAT1 signaling. Our experiments define a regulatory network in which Banf1 limits basal inflammation by preventing self DNA accumulation in the cytosol., (Copyright © 2020 Ma et al.)
- Published
- 2020
- Full Text
- View/download PDF
41. Blood natural killer cell deficiency reveals an immunotherapy strategy for atopic dermatitis.
- Author
-
Mack MR, Brestoff JR, Berrien-Elliott MM, Trier AM, Yang TB, McCullen M, Collins PL, Niu H, Bodet ND, Wagner JA, Park E, Xu AZ, Wang F, Chibnall R, Council ML, Heffington C, Kreisel F, Margolis DJ, Sheinbein D, Lovato P, Vivier E, Cella M, Colonna M, Yokoyama WM, Oltz EM, Fehniger TA, and Kim BS
- Subjects
- Animals, Disease Models, Animal, Humans, Immunotherapy, Killer Cells, Natural, Mice, Dermatitis, Atopic therapy, GATA2 Deficiency
- Abstract
Atopic dermatitis (AD) is a widespread, chronic skin disease associated with aberrant allergic inflammation. Current treatments involve either broad or targeted immunosuppression strategies. However, enhancing the immune system to control disease remains untested. We demonstrate that patients with AD harbor a blood natural killer (NK) cell deficiency that both has diagnostic value and improves with therapy. Multidimensional protein and RNA profiling revealed subset-level changes associated with enhanced NK cell death. Murine NK cell deficiency was associated with enhanced type 2 inflammation in the skin, suggesting that NK cells play a critical immunoregulatory role in this context. On the basis of these findings, we used an NK cell-boosting interleukin-15 (IL-15) superagonist and observed marked improvement in AD-like disease in mice. These findings reveal a previously unrecognized application of IL-15 superagonism, currently in development for cancer immunotherapy, as an immunotherapeutic strategy for AD., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
42. Neuroimmunology: To Sense and Protect.
- Author
-
Oltz EM
- Subjects
- Animals, Humans, Neuroimmunomodulation
- Published
- 2020
- Full Text
- View/download PDF
43. Regional Gene Repression by DNA Double-Strand Breaks in G 1 Phase Cells.
- Author
-
Purman CE, Collins PL, Porter SI, Saini A, Gupta H, Sleckman BP, and Oltz EM
- Subjects
- Animals, Cell Cycle genetics, Cell Line, DNA genetics, DNA Breaks, Double-Stranded, DNA Damage physiology, DNA End-Joining Repair genetics, DNA Repair physiology, DNA-Binding Proteins metabolism, G1 Phase physiology, Humans, Mice, Regulatory Elements, Transcriptional genetics, Regulatory Elements, Transcriptional physiology, Silencer Elements, Transcriptional physiology, DNA Repair genetics, G1 Phase genetics, Silencer Elements, Transcriptional genetics
- Abstract
DNA damage responses (DDR) to double-strand breaks (DSBs) alter cellular transcription programs at the genome-wide level. Through processes that are less well understood, DSBs also alter transcriptional responses locally, which may be important for efficient DSB repair. Here, we developed an approach to elucidate the cis -acting responses to DSBs in G
1 phase cells. We found that DSBs within a gene body silence its expression, as well as the transcription of local undamaged genes at a distance defined by the spread of γ-H2AX from the DSB. Importantly, DSBs not only repress ongoing transcription but also block the inducible expression of regional genes. DSB-mediated transcriptional repression depends on DDR signaling but does not require the generation of inaccessible chromatin. Our findings demonstrate that in G1 phase cells, DDR signaling establishes a robust and extensive region of transcriptional repression spreading from DSB sites and introduce an approach to study the mechanistic impact of targeted DNA breaks in nearly any chromatin environment., (Copyright © 2019 American Society for Microbiology.)- Published
- 2019
- Full Text
- View/download PDF
44. Circadian rhythm-dependent and circadian rhythm-independent impacts of the molecular clock on type 3 innate lymphoid cells.
- Author
-
Wang Q, Robinette ML, Billon C, Collins PL, Bando JK, Fachi JL, Sécca C, Porter SI, Saini A, Gilfillan S, Solt LA, Musiek ES, Oltz EM, Burris TP, and Colonna M
- Subjects
- Animals, Mice, Mice, Congenic, Mice, Inbred C57BL, Mice, Knockout, Nuclear Receptor Subfamily 1, Group D, Member 1 deficiency, Nuclear Receptor Subfamily 1, Group D, Member 1 immunology, Circadian Rhythm immunology, Immunity, Innate immunology, Intestines immunology, Lymphocytes immunology, Nuclear Receptor Subfamily 1, Group F, Member 3 immunology
- Abstract
Many gut functions are attuned to circadian rhythm. Intestinal group 3 innate lymphoid cells (ILC3s) include NKp46
+ and NKp46- subsets, which are RORγt dependent and provide mucosal defense through secretion of interleukin-22 (IL-22) and IL-17. Because ILC3s highly express some key circadian clock genes, we investigated whether ILC3s are also attuned to circadian rhythm. We noted circadian oscillations in the expression of clock and cytokine genes, such as REV-ERBα, IL-22, and IL-17, whereas acute disruption of the circadian rhythm affected cytokine secretion by ILC3s. Because of prominent and rhythmic expression of REV-ERBα in ILC3s, we also investigated the impact of constitutive deletion of REV-ERBα, which has been previously shown to inhibit the expression of a RORγt repressor, NFIL3, while also directly antagonizing DNA binding of RORγt. Development of the NKp46+ ILC3 subset was markedly impaired, with reduced cell numbers, RORγt expression, and IL-22 production in REV-ERBα-deficient mice. The NKp46- ILC3 subsets developed normally, potentially due to compensatory expression of other clock genes, but IL-17 secretion paradoxically increased, probably because RORγt was not antagonized by REV-ERBα. We conclude that ILC3s are attuned to circadian rhythm, but clock regulator REV-ERBα also has circadian-independent impacts on ILC3 development and functions due to its roles in the regulation of RORγt., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
45. Publisher Correction: Subsets of ILC3-ILC1-like cells generate a diversity spectrum of innate lymphoid cells in human mucosal tissues.
- Author
-
Cella M, Gamini R, Sécca C, Collins PL, Zhao S, Peng V, Robinette ML, Schettini J, Zaitsev K, Gordon W, Bando JK, Yomogida K, Cortez V, Fronick C, Fulton R, Lin LL, Gilfillan S, Flavell RA, Shan L, Artyomov MN, Bowman M, Oltz EM, Jelinsky SA, and Colonna M
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
46. Subsets of ILC3-ILC1-like cells generate a diversity spectrum of innate lymphoid cells in human mucosal tissues.
- Author
-
Cella M, Gamini R, Sécca C, Collins PL, Zhao S, Peng V, Robinette ML, Schettini J, Zaitsev K, Gordon W, Bando JK, Yomogida K, Cortez V, Fronick C, Fulton R, Lin LL, Gilfillan S, Flavell RA, Shan L, Artyomov MN, Bowman M, Oltz EM, Jelinsky SA, and Colonna M
- Subjects
- Animals, Cell Differentiation immunology, Cells, Cultured, Child, Child, Preschool, Humans, Ikaros Transcription Factor metabolism, Intestinal Mucosa cytology, Lymphocytes classification, Lymphocytes cytology, Mice, T-Box Domain Proteins metabolism, Interleukin-22, Immunity, Innate immunology, Interferon-gamma metabolism, Interleukins metabolism, Intestinal Mucosa immunology, Lymphocytes immunology, Palatine Tonsil immunology
- Abstract
Innate lymphoid cells (ILCs) are tissue-resident lymphocytes categorized on the basis of their core regulatory programs and the expression of signature cytokines. Human ILC3s that produce the cytokine interleukin-22 convert into ILC1-like cells that produce interferon-γ in vitro, but whether this conversion occurs in vivo remains unclear. In the present study we found that ILC3s and ILC1s in human tonsils represented the ends of a spectrum that included additional discrete subsets. RNA velocity analysis identified an intermediate ILC3-ILC1 cluster, which had strong directionality toward ILC1s. In humanized mice, the acquisition of ILC1 features by ILC3s showed tissue dependency. Chromatin studies indicated that the transcription factors Aiolos and T-bet cooperated to repress regulatory elements active in ILC3s. A transitional ILC3-ILC1 population was also detected in the human intestine. We conclude that ILC3s undergo conversion into ILC1-like cells in human tissues in vivo, and that tissue factors and Aiolos were required for this process.
- Published
- 2019
- Full Text
- View/download PDF
47. Immunity to Influenza: Closing in on a Moving Target.
- Author
-
Oltz EM
- Subjects
- Antibodies, Neutralizing metabolism, Antibodies, Viral metabolism, Antigenic Variation, Cross Reactions, Humans, Immune Evasion, Immunologic Memory, Immunity, Influenza Vaccines immunology, Influenza, Human immunology, Orthomyxoviridae physiology, T-Lymphocytes immunology
- Published
- 2019
- Full Text
- View/download PDF
48. Gene Regulatory Programs Conferring Phenotypic Identities to Human NK Cells.
- Author
-
Collins PL, Cella M, Porter SI, Li S, Gurewitz GL, Hong HS, Johnson RP, Oltz EM, and Colonna M
- Subjects
- Cytokines immunology, Cytokines metabolism, Gene Expression Regulation genetics, Gene Expression Regulation immunology, Humans, Phenotype, Killer Cells, Natural classification, Killer Cells, Natural immunology, Killer Cells, Natural physiology
- Abstract
Natural killer (NK) cells develop from common progenitors but diverge into distinct subsets, which differ in cytokine production, cytotoxicity, homing, and memory traits. Given their promise in adoptive cell therapies for cancer, a deeper understanding of regulatory modules controlling clinically beneficial NK phenotypes is of high priority. We report integrated "-omics" analysis of human NK subsets, which revealed super-enhancers associated with gene cohorts that may coordinate NK functions and localization. A transcription factor-based regulatory scheme also emerged, which is evolutionarily conserved and shared by innate and adaptive lymphocytes. For both NK and T lineages, a TCF1-LEF1-MYC axis dominated the regulatory landscape of long-lived, proliferative subsets that traffic to lymph nodes. In contrast, effector populations circulating between blood and peripheral tissues shared a PRDM1-dominant landscape. This resource defines transcriptional modules, regulated by feedback loops, which may be leveraged to enhance phenotypes for NK cell-based therapies., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
49. A B-Cell-Specific Enhancer Orchestrates Nuclear Architecture to Generate a Diverse Antigen Receptor Repertoire.
- Author
-
Barajas-Mora EM, Kleiman E, Xu J, Carrico NC, Lu H, Oltz EM, Murre C, and Feeney AJ
- Subjects
- Animals, Cell Line, Tumor, Cell Nucleus genetics, Cell Nucleus metabolism, Cell Nucleus Shape, Chromatin Assembly and Disassembly, Genotype, HEK293 Cells, Humans, Immunoglobulin kappa-Chains chemistry, Immunoglobulin kappa-Chains genetics, Immunoglobulin kappa-Chains metabolism, Mice, Inbred C57BL, Mice, Transgenic, Phenotype, Precursor Cells, B-Lymphoid metabolism, Protein Conformation, Receptors, Antigen, B-Cell chemistry, Receptors, Antigen, B-Cell genetics, Receptors, Antigen, B-Cell metabolism, Structure-Activity Relationship, Antibody Diversity, Cell Nucleus immunology, Enhancer Elements, Genetic, Gene Rearrangement, B-Lymphocyte, Immunoglobulin kappa-Chains immunology, Precursor Cells, B-Lymphoid immunology, Receptors, Antigen, B-Cell immunology
- Abstract
The genome is organized into topologically associated domains (TADs) that enclose smaller subTADs. Here, we identify and characterize an enhancer that is located in the middle of the V gene region of the immunoglobulin kappa light chain (Igκ) locus that becomes active preceding the stage at which this locus undergoes V(D)J recombination. This enhancer is a hub of long-range chromatin interactions connecting subTADs in the V gene region with the recombination center at the J genes. Deletion of this element results in a highly altered long-range chromatin interaction pattern across the locus and, importantly, affects individual V gene utilization locus-wide. These results indicate the existence of an enhancer-dependent framework in the Igκ locus and further suggest that the composition of the diverse antibody repertoire is regulated in a subTAD-specific manner. This enhancer thus plays a structural role in orchestrating the proper folding of the Igκ locus in preparation for V(D)J recombination., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
50. A New JI : The Home for All Immunology.
- Author
-
Oltz EM
- Subjects
- Humans, Allergy and Immunology, Periodicals as Topic, Publishing
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.