202 results on '"Betts MR"'
Search Results
2. OA05-02. Analysis of DNA compared to Ad5 vaccination, as single and mixed modalities, demonstrates robust induction of cellular immune responses in macaques
- Author
-
Sardesai NY, Betts MR, Shiver J, Casimiro D, Finnefrock A, Khan AS, Satishchandran A, Wu L, Hirao LA, and Weiner DB
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
3. P03-11. DNA vaccination with IL-12 lowers viral replication following SIVmac251 challenge
- Author
-
Boyer JD, Sardesai NY, Betts MR, Miller N, Watkins D, O'Connor D, Wiseman R, Lank S, Karl J, Dai A, Khan AS, Wu L, Hirao LA, and Weiner DB
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
4. P19-46. Co-delivery of mucosal chemokine plasmids in a systemically delivered DNA vaccine elicits systemic and mucosal immune responses in mice and macaques
- Author
-
Betts MR, Mestecky J, Moldoveanu Z, Sardesai N, Khan AS, Carnathan D, Yan J, Sylvester A, Pahar B, Kutzler MA, Kraynyak KA, Marx P, and Weiner DB
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
5. P01-05. Rapid perforin upregulation dominates the HIV-specific CD8 T cell response during acute HIV-infection
- Author
-
Weinhold KJ, Ostrowski MA, Guidonis D, Frank I, Makedonas G, and Betts MR
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
6. P16-09. Adenovirus 5 vector HIV vaccination does not affect mucosal homing markers on Ad5-specific CD4+ T-cells in humans
- Author
-
Ertl HC, Casimiro DR, Robertson MN, Ratcliffe S, Cox K, Dubey S, Carnathan D, Hutnick N, and Betts MR
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
7. S03-06 OA. Rapid perforin upregulation by CD8 T cells in elite controllers as a correlate of immune-mediated control of HIV replication
- Author
-
Kaul R, Lederman MM, Goepfert PA, Teixeira-Johnson L, Sieg SF, Rodriguez B, Shin LY, Sheth P, Demers K, Pereyra F, Hersperger AR, Makedonas G, Walker BD, and Betts MR
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
8. Progress Note 2024: Curing HIV; Not in My Lifetime or Just Around the Corner?
- Author
-
Harper, J, Betts, MR, Lichterfeld, M, Müller-Trutwin, M, Margolis, D, Bar, KJ, Li, JZ, McCune, JM, Lewin, S, Kulpa, D, Diallo, DD, Lederman, MM, Paiardini, M, Harper, J, Betts, MR, Lichterfeld, M, Müller-Trutwin, M, Margolis, D, Bar, KJ, Li, JZ, McCune, JM, Lewin, S, Kulpa, D, Diallo, DD, Lederman, MM, and Paiardini, M
- Abstract
Once a death sentence, HIV is now considered a manageable chronic disease due to the development of antiretroviral therapy (ART) regimens with minimal toxicity and a high barrier for genetic resistance. While highly effective in arresting AIDS progression and rendering the virus untransmissible in people living with HIV (PLWH) with undetectable viremia (U=U) [1, 2]), ART alone is incapable of eradicating the "reservoir" of resting, latently infected CD4+ T cells from which virus recrudesces upon treatment cessation. As of 2022 estimates, there are 39 million PLWH, of whom 86% are aware of their status and 76% are receiving ART [3]. As of 2017, ART-treated PLWH exhibit near normalized life expectancies without adjustment for socioeconomic differences [4]. Furthermore, there is a global deceleration in the rate of new infections [3] driven by expanded access to pre-exposure prophylaxis (PrEP), HIV testing in vulnerable populations, and by ART treatment [5]. Therefore, despite outstanding issues pertaining to cost and access in developing countries, there is strong enthusiasm that aggressive testing, treatment, and effective viral suppression may be able to halt the ongoing HIV epidemic (ie, UNAIDS' 95-95-95 targets) [6-8]; especially as evidenced by recent encouraging observations in Sydney [9]. Despite these promising efforts to limit further viral transmission, for PLWH, a "cure" remains elusive; whether it be to completely eradicate the viral reservoir (ie, cure) or to induce long-term viral remission in the absence of ART (ie, control; Figure 1). In a previous salon hosted by Pathogens and Immunity in 2016 [10], some researchers were optimistic that a cure was a feasible, scalable goal, albeit with no clear consensus on the best route. So, how are these cure strategies panning out? In this commentary, 8 years later, we will provide a brief overview on recent advances and failures towards identifying determinants of viral persistence and developing a scalable cure
- Published
- 2023
9. Erratum to: Progress Note 2024: Curing HIV; Not in My Lifetime or Just Around the Corner?
- Author
-
Harper, J, Betts, MR, Lichterfeld, M, Müller-Trutwin, M, Margolis, D, Bar, KJ, Li, JZ, McCune, JM, Lewin, SR, Kulpa, D, Ávila-Ríos, S, Diallo, DD, Lederman, MM, Paiardini, M, Harper, J, Betts, MR, Lichterfeld, M, Müller-Trutwin, M, Margolis, D, Bar, KJ, Li, JZ, McCune, JM, Lewin, SR, Kulpa, D, Ávila-Ríos, S, Diallo, DD, Lederman, MM, and Paiardini, M
- Abstract
[This corrects the article DOI: 10.20411/pai.v8i2.665.].
- Published
- 2023
10. Single-cell multi-omics analysis of human pancreatic islets reveals novel cellular states in type 1 diabetes
- Author
-
Fasolino M, Schwartz GW, Patil AR, Mongia A, Golson ML, Wang YJ, Morgan A, Liu C, Schug J, Liu J, Wu M, Traum D, Kondo A, May CL, Goldman N, Wang W, Feldman M, Moore JH, Japp AS, Betts MR, HPAP Consortium, Faryabi RB, Naji A, Kaestner KH, and Vahedi G
- Subjects
General Economics, Econometrics and Finance - Published
- 2022
11. Sialyl-Lewis(X) Glycoantigen Is Enriched on Cells with Persistent HIV Transcription during Therapy
- Author
-
Colomb, F, Giron, LB, Kuri-Cervantes, L, Adeniji, OS, Ma, T, Dweep, H, Battivelli, E, Verdin, E, Palmer, CS, Tateno, H, Kossenkov, A, Roan, NR, Betts, MR, Abdel-Mohsen, M, Colomb, F, Giron, LB, Kuri-Cervantes, L, Adeniji, OS, Ma, T, Dweep, H, Battivelli, E, Verdin, E, Palmer, CS, Tateno, H, Kossenkov, A, Roan, NR, Betts, MR, and Abdel-Mohsen, M
- Abstract
A comprehensive understanding of the phenotype of persistent HIV-infected cells, transcriptionally active and/or transcriptionally inactive, is imperative for developing a cure. The relevance of cell-surface glycosylation to HIV persistence has never been explored. We characterize the relationship between cell-surface glycomic signatures and persistent HIV transcription in vivo. We find that the cell surface of CD4+ T cells actively transcribing HIV, despite suppressive therapy, harbors high levels of fucosylated carbohydrate ligands, including the cell extravasation mediator Sialyl-LewisX (SLeX), compared with HIV-infected transcriptionally inactive cells. These high levels of SLeX are induced by HIV transcription in vitro and are maintained after therapy in vivo. Cells with high-SLeX are enriched with markers associated with HIV susceptibility, signaling pathways that drive HIV transcription, and pathways involved in leukocyte extravasation. We describe a glycomic feature of HIV-infected transcriptionally active cells that not only differentiates them from their transcriptionally inactive counterparts but also may affect their trafficking abilities.
- Published
- 2020
12. P19-46. Co-delivery of mucosal chemokine plasmids in a systemically delivered DNA vaccine elicits systemic and mucosal immune responses in mice and macaques
- Author
-
Kraynyak, KA, primary, Kutzler, MA, additional, Pahar, B, additional, Sylvester, A, additional, Yan, J, additional, Carnathan, D, additional, Khan, AS, additional, Sardesai, N, additional, Moldoveanu, Z, additional, Mestecky, J, additional, Betts, MR, additional, Marx, P, additional, and Weiner, DB, additional
- Published
- 2009
- Full Text
- View/download PDF
13. P03-11. DNA vaccination with IL-12 lowers viral replication following SIVmac251 challenge
- Author
-
Hirao, LA, primary, Wu, L, additional, Khan, AS, additional, Dai, A, additional, Karl, J, additional, Lank, S, additional, Wiseman, R, additional, O'Connor, D, additional, Watkins, D, additional, Miller, N, additional, Betts, MR, additional, Sardesai, NY, additional, Boyer, JD, additional, and Weiner, DB, additional
- Published
- 2009
- Full Text
- View/download PDF
14. P01-05. Rapid perforin upregulation dominates the HIV-specific CD8 T cell response during acute HIV-infection
- Author
-
Makedonas, G, primary, Frank, I, additional, Guidonis, D, additional, Ostrowski, MA, additional, Weinhold, KJ, additional, and Betts, MR, additional
- Published
- 2009
- Full Text
- View/download PDF
15. P16-09. Adenovirus 5 vector HIV vaccination does not affect mucosal homing markers on Ad5-specific CD4+ T-cells in humans
- Author
-
Hutnick, N, primary, Carnathan, D, additional, Dubey, S, additional, Cox, K, additional, Ratcliffe, S, additional, Robertson, MN, additional, Casimiro, DR, additional, Ertl, HC, additional, and Betts, MR, additional
- Published
- 2009
- Full Text
- View/download PDF
16. S03-06 OA. Rapid perforin upregulation by CD8 T cells in elite controllers as a correlate of immune-mediated control of HIV replication
- Author
-
Hersperger, AR, primary, Pereyra, F, additional, Demers, K, additional, Sheth, P, additional, Shin, LY, additional, Rodriguez, B, additional, Sieg, SF, additional, Teixeira-Johnson, L, additional, Goepfert, PA, additional, Lederman, MM, additional, Kaul, R, additional, Makedonas, G, additional, Walker, BD, additional, and Betts, MR, additional
- Published
- 2009
- Full Text
- View/download PDF
17. OA05-02. Analysis of DNA compared to Ad5 vaccination, as single and mixed modalities, demonstrates robust induction of cellular immune responses in macaques
- Author
-
Hirao, LA, primary, Wu, L, additional, Satishchandran, A, additional, Khan, AS, additional, Finnefrock, A, additional, Casimiro, D, additional, Shiver, J, additional, Betts, MR, additional, Sardesai, NY, additional, and Weiner, DB, additional
- Published
- 2009
- Full Text
- View/download PDF
18. No Evidence for Persistent Enteroviral B Infection of Pancreatic Islets in Patients With Type 1 Diabetes and Prediabetes From RNA Sequencing Data.
- Author
-
Manduchi E, Descamps HC, Schug J, Da T, Lahori D, El-Mekkoussi H, Betts MR, and Kaestner KH
- Subjects
- Humans, Male, Female, Adult, Diabetes Mellitus, Type 1 virology, Diabetes Mellitus, Type 1 genetics, Islets of Langerhans virology, Enterovirus Infections virology, Enterovirus Infections genetics, Prediabetic State virology, Prediabetic State genetics, Enterovirus B, Human genetics, Sequence Analysis, RNA methods
- Abstract
Persistent enterovirus B infection has been proposed as an important contributor to the etiology of type 1 diabetes. We leveraged extensive bulk RNA-sequencing (RNA-seq) data from α-, β-, and exocrine cells, as well as islet single-cell RNA-seq data from the Human Pancreas Analysis Program (HPAP), to evaluate the presence of enterovirus B sequences in the pancreas of patients with type 1 diabetes and prediabetes (no diabetes but positive for autoantibodies). We examined all available HPAP data for either assay type, including donors without diabetes and with type 1 and type 2 diabetes. To assess the presence of viral reads, we analyzed all reads not mapping to the human genome with the taxonomic classification system Kraken2 and its full viral database augmented to encompass representatives for all 28 enterovirus B serotypes for which a complete genome is available. As a secondary approach, we input the same sequence reads into the STAR aligner using these 28 enterovirus B genomes as the reference. No enterovirus B sequences were detected by either approach in any of the 243 bulk RNA libraries or in any of the 79 single-cell RNA libraries. While we cannot rule out the possibility of a very-low-grade persistent enterovirus B infection in the donors analyzed, our data do not support the notion of chronic viral infection by these viruses as a major driver of type 1 diabetes., (© 2024 by the American Diabetes Association.)
- Published
- 2024
- Full Text
- View/download PDF
19. Immune perturbations in human pancreas lymphatic tissues prior to and after type 1 diabetes onset.
- Author
-
Golden GJ, Wu VH, Hamilton JT, Amses KR, Shapiro MR, Japp AS, Liu C, Pampena MB, Kuri-Cervantes L, Knox JJ, Gardner JS, Atkinson MA, Brusko TM, Prak ETL, Kaestner KH, Naji A, and Betts MR
- Abstract
Autoimmune destruction of pancreatic β cells results in type 1 diabetes (T1D), with pancreatic immune infiltrate representing a key feature in this process. Studies of human T1D immunobiology have predominantly focused on circulating immune cells in the blood, while mouse models suggest diabetogenic lymphocytes primarily reside in pancreas-draining lymph nodes (pLN). A comprehensive study of immune cells in human T1D was conducted using pancreas draining lymphatic tissues, including pLN and mesenteric lymph nodes, and the spleen from non-diabetic control, β cell autoantibody positive non-diabetic (AAb+), and T1D organ donors using complementary approaches of high parameter flow cytometry and CITEseq. Immune perturbations suggestive of a proinflammatory environment were specific for T1D pLN and AAb+ pLN. In addition, certain immune populations correlated with high T1D genetic risk independent of disease state. These datasets form an extensive resource for profiling human lymphatic tissue immune cells in the context of autoimmunity and T1D., Competing Interests: Ethics Declarations The authors declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
20. Distinct SIV-specific CD8 + T cells in the lymph node exhibit simultaneous effector and stem-like profiles and are associated with limited SIV persistence.
- Author
-
Strongin Z, Raymond Marchand L, Deleage C, Pampena MB, Cardenas MA, Beusch CM, Hoang TN, Urban EA, Gourves M, Nguyen K, Tharp GK, Lapp S, Rahmberg AR, Harper J, Del Rio Estrada PM, Gonzalez-Navarro M, Torres-Ruiz F, Luna-Villalobos YA, Avila-Rios S, Reyes-Teran G, Sekaly R, Silvestri G, Kulpa DA, Saez-Cirion A, Brenchley JM, Bosinger SE, Gordon DE, Betts MR, Kissick HT, and Paiardini M
- Subjects
- Animals, Humans, Macaca mulatta, HIV Infections immunology, HIV Infections virology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Simian Immunodeficiency Virus immunology, CD8-Positive T-Lymphocytes immunology, Simian Acquired Immunodeficiency Syndrome immunology, Simian Acquired Immunodeficiency Syndrome virology, Lymph Nodes immunology
- Abstract
Human immunodeficiency virus (HIV) cure efforts are increasingly focused on harnessing CD8
+ T cell functions, which requires a deeper understanding of CD8+ T cells promoting HIV control. Here we identifiy an antigen-responsive TOXhi TCF1+ CD39+ CD8+ T cell population with high expression of inhibitory receptors and low expression of canonical cytolytic molecules. Transcriptional analysis of simian immunodeficiency virus (SIV)-specific CD8+ T cells and proteomic analysis of purified CD8+ T cell subsets identified TOXhi TCF1+ CD39+ CD8+ T cells as intermediate effectors that retained stem-like features with a lineage relationship with terminal effector T cells. TOXhi TCF1+ CD39+ CD8+ T cells were found at higher frequency than TCF1- CD39+ CD8+ T cells in follicular microenvironments and were preferentially located in proximity of SIV-RNA+ cells. Their frequency was associated with reduced plasma viremia and lower SIV reservoir size. Highly similar TOXhi TCF1+ CD39+ CD8+ T cells were detected in lymph nodes from antiretroviral therapy-naive and antiretroviral therapy-suppressed people living with HIV, suggesting this population of CD8+ T cells contributes to limiting SIV and HIV persistence., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
21. Erratum to: Progress Note 2024: Curing HIV; Not in My Lifetime or Just Around the Corner?
- Author
-
Harper J, Betts MR, Lichterfeld M, Müller-Trutwin M, Margolis D, Bar KJ, Li JZ, McCune JM, Lewin SR, Kulpa D, Ávila-Ríos S, Diallo DD, Lederman MM, and Paiardini M
- Abstract
[This corrects the article DOI: 10.20411/pai.v8i2.665.]., Competing Interests: JH and MP have active collaborations with Merck & Co., Inc., and routinely receive antiretroviral compounds for nonhuman primate studies from ViiV Healthcare and Gilead Sciences, but the authors declare no financial stake. MML has received competitive grant funding from Gilead., (Copyright © 2024 Pathogens and Immunity.)
- Published
- 2024
- Full Text
- View/download PDF
22. Progress Note 2024: Curing HIV; Not in My Lifetime or Just Around the Corner?
- Author
-
Harper J, Betts MR, Lichterfeld M, Müller-Trutwin M, Margolis D, Bar KJ, Li JZ, McCune JM, Lewin SR, Kulpa D, Diallo DD, Lederman MM, and Paiardini M
- Abstract
Once a death sentence, HIV is now considered a manageable chronic disease due to the development of antiretroviral therapy (ART) regimens with minimal toxicity and a high barrier for genetic resistance. While highly effective in arresting AIDS progression and rendering the virus untransmissible in people living with HIV (PLWH) with undetectable viremia (U=U) [1, 2]), ART alone is incapable of eradicating the "reservoir" of resting, latently infected CD4
+ T cells from which virus recrudesces upon treatment cessation. As of 2022 estimates, there are 39 million PLWH, of whom 86% are aware of their status and 76% are receiving ART [3]. As of 2017, ART-treated PLWH exhibit near normalized life expectancies without adjustment for socioeconomic differences [4]. Furthermore, there is a global deceleration in the rate of new infections [3] driven by expanded access to pre-exposure prophylaxis (PrEP), HIV testing in vulnerable populations, and by ART treatment [5]. Therefore, despite outstanding issues pertaining to cost and access in developing countries, there is strong enthusiasm that aggressive testing, treatment, and effective viral suppression may be able to halt the ongoing HIV epidemic (ie, UNAIDS' 95-95-95 targets) [6-8]; especially as evidenced by recent encouraging observations in Sydney [9]. Despite these promising efforts to limit further viral transmission, for PLWH, a "cure" remains elusive; whether it be to completely eradicate the viral reservoir (ie, cure) or to induce long-term viral remission in the absence of ART (ie, control; Figure 1). In a previous salon hosted by Pathogens and Immunity in 2016 [10], some researchers were optimistic that a cure was a feasible, scalable goal, albeit with no clear consensus on the best route. So, how are these cure strategies panning out? In this commentary, 8 years later, we will provide a brief overview on recent advances and failures towards identifying determinants of viral persistence and developing a scalable cure for HIV. Based on these observations, and as in the earlier salon, we have asked several prominent HIV cure researchers for their perspectives., Competing Interests: JH and MP have active collaborations with Merck & Co., Inc., and routinely receive antiretroviral compounds for nonhuman primate studies from ViiV Healthcare and Gilead Sciences, but the authors declare no financial stake. MML has received competitive grant funding from Gilead., (Copyright © 2024 Pathogens and Immunity.)- Published
- 2024
- Full Text
- View/download PDF
23. Preferential selection of viral escape mutants by CD8+ T cell 'sieving' of SIV reactivation from latency.
- Author
-
Docken SS, McCormick K, Pampena MB, Samer S, Lindemuth E, Pinkevych M, Viox EG, Wu Y, Schlub TE, Cromer D, Keele BF, Paiardini M, Betts MR, Bar KJ, and Davenport MP
- Subjects
- Animals, Macaca mulatta, Virus Replication physiology, CD8-Positive T-Lymphocytes, Epitopes, Viral Load, Anti-Retroviral Agents therapeutic use, Anti-Retroviral Agents pharmacology, Simian Acquired Immunodeficiency Syndrome, Simian Immunodeficiency Virus, HIV Infections
- Abstract
HIV rapidly rebounds after interruption of antiretroviral therapy (ART). HIV-specific CD8+ T cells may act to prevent early events in viral reactivation. However, the presence of viral immune escape mutations may limit the effect of CD8+ T cells on viral rebound. Here, we studied the impact of CD8 immune pressure on post-treatment rebound of barcoded SIVmac293M in 14 Mamu-A*01 positive rhesus macaques that initiated ART on day 14, and subsequently underwent two analytic treatment interruptions (ATIs). Rebound following the first ATI (seven months after ART initiation) was dominated by virus that retained the wild-type sequence at the Mamu-A*01 restricted Tat-SL8 epitope. By the end of the two-month treatment interruption, the replicating virus was predominantly escaped at the Tat-SL8 epitope. Animals reinitiated ART for 3 months prior to a second treatment interruption. Time-to-rebound and viral reactivation rate were significantly slower during the second treatment interruption compared to the first. Tat-SL8 escape mutants dominated early rebound during the second treatment interruption, despite the dominance of wild-type virus in the proviral reservoir. Furthermore, the escape mutations detected early in the second treatment interruption were well predicted by those replicating at the end of the first, indicating that escape mutant virus in the second interruption originated from the latent reservoir as opposed to evolving de novo post rebound. SL8-specific CD8+ T cell levels in blood prior to the second interruption were marginally, but significantly, higher (median 0.73% vs 0.60%, p = 0.016). CD8+ T cell depletion approximately 95 days after the second treatment interruption led to the reappearance of wild-type virus. This work suggests that CD8+ T cells can actively suppress the rebound of wild-type virus, leading to the dominance of escape mutant virus after treatment interruption., Competing Interests: Michael R. Betts is a paid consultant of Interius Biotherapeutics. The authors have no other competing interests to declare., (Copyright: This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.)
- Published
- 2023
- Full Text
- View/download PDF
24. Siglec-7 glyco-immune binding mAbs or NK cell engager biologics induce potent antitumor immunity against ovarian cancers.
- Author
-
Bordoloi D, Kulkarni AJ, Adeniji OS, Pampena MB, Bhojnagarwala PS, Zhao S, Ionescu C, Perales-Puchalt A, Parzych EM, Zhu X, Ali AR, Cassel J, Zhang R, Betts MR, Abdel-Mohsen M, and Weiner DB
- Subjects
- Female, Humans, Mice, Animals, Killer Cells, Natural, Antigens, CD metabolism, Sialic Acid Binding Immunoglobulin-like Lectins metabolism, Biological Products metabolism, Ovarian Neoplasms therapy, Ovarian Neoplasms metabolism
- Abstract
Ovarian cancer (OC) is a lethal gynecologic malignancy, with modest responses to CPI. Engagement of additional immune arms, such as NK cells, may be of value. We focused on Siglec-7 as a surface antigen for engaging this population. Human antibodies against Siglec-7 were developed and characterized. Coculture of OC cells with PBMCs/NKs and Siglec-7 binding antibodies showed NK-mediated killing of OC lines. Anti-Siglec-7 mAb (DB7.2) enhanced survival in OC-challenged mice. In addition, the combination of DB7.2 and anti-PD-1 demonstrated further improved OC killing in vitro. To use Siglec-7 engagement as an OC-specific strategy, we engineered an NK cell engager (NKCE) to simultaneously engage NK cells through Siglec-7, and OC targets through FSHR. The NKCE demonstrated robust in vitro killing of FSHR
+ OC, controlled tumors, and improved survival in OC-challenged mice. These studies support additional investigation of the Siglec-7 targeting approaches as important tools for OC and other recalcitrant cancers.- Published
- 2023
- Full Text
- View/download PDF
25. Timing of initiation of anti-retroviral therapy predicts post-treatment control of SIV replication.
- Author
-
Pinkevych M, Docken SS, Okoye AA, Fennessey CM, Del Prete GQ, Pino M, Harper JL, Betts MR, Paiardini M, Keele BF, and Davenport MP
- Subjects
- Animals, CD8-Positive T-Lymphocytes, RNA, Viral, Viral Load, Anti-Retroviral Agents pharmacology, Anti-Retroviral Agents therapeutic use, HIV Infections drug therapy
- Abstract
One approach to 'functional cure' of HIV infection is to induce durable control of HIV replication after the interruption of antiretroviral therapy (ART). However, the major factors that determine the viral 'setpoint' level after treatment interruption are not well understood. Here we combine data on ART interruption following SIV infection for 124 total animals from 10 independent studies across 3 institutional cohorts to understand the dynamics and predictors of post-treatment viral control. We find that the timing of treatment initiation is an important determinant of both the peak and early setpoint viral levels after treatment interruption. During the first 3 weeks of infection, every day of delay in treatment initiation is associated with a 0.22 log10 copies/ml decrease in post-rebound peak and setpoint viral levels. However, delay in initiation of ART beyond 3 weeks of infection is associated with higher post-rebound setpoint viral levels. For animals treated beyond 3 weeks post-infection, viral load at ART initiation was the primary predictor of post-rebound setpoint viral levels. Potential alternative predictors of post-rebound setpoint viral loads including cell-associated DNA or RNA, time from treatment interruption to rebound, and pre-interruption CD8+ T cell responses were also examined in the studies where these data were available. This analysis suggests that optimal timing of treatment initiation may be an important determinant of post-treatment control of HIV., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: MRB is a paid consultant of Interius Biotherapeutics. All other authors have declared that no competing interests exist., (Copyright: This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.)
- Published
- 2023
- Full Text
- View/download PDF
26. Author Correction: Human circulating and tissue-resident memory CD8 + T cells.
- Author
-
Buggert M, Price DA, Mackay LK, and Betts MR
- Published
- 2023
- Full Text
- View/download PDF
27. Human circulating and tissue-resident memory CD8 + T cells.
- Author
-
Buggert M, Price DA, Mackay LK, and Betts MR
- Subjects
- Humans, Lymphocyte Activation, Memory T Cells, Immunologic Memory, CD8-Positive T-Lymphocytes, COVID-19
- Abstract
Our current knowledge of human memory CD8
+ T cells is derived largely from studies of the intravascular space. However, emerging data are starting to challenge some of the dogmas based on this work, suggesting that a conceptual revision may be necessary. In this review, we provide a brief history of the field and summarize the biology of circulating and tissue-resident memory CD8+ T cells, which are ultimately responsible for effective immune surveillance. We also incorporate recent findings into a biologically integrated model of human memory CD8+ T cell differentiation. Finally, we address how future innovative human studies could improve our understanding of anatomically localized CD8+ T cells to inform the development of more effective immunotherapies and vaccines, the need for which has been emphasized by the global struggle to contain severe acute respiratory syndrome coronavirus 2., (© 2023. Springer Nature America, Inc.)- Published
- 2023
- Full Text
- View/download PDF
28. Lymph-Node-Based CD3 + CD20 + Cells Emerge from Membrane Exchange between T Follicular Helper Cells and B Cells and Increase Their Frequency following Simian Immunodeficiency Virus Infection.
- Author
-
Samer S, Chowdhury A, Wiche Salinas TR, Estrada PMDR, Reuter M, Tharp G, Bosinger S, Cervasi B, Auger J, Gill K, Ablanedo-Terrazas Y, Reyes-Teran G, Estes JD, Betts MR, Silvestri G, and Paiardini M
- Subjects
- Animals, Humans, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes virology, HIV Infections immunology, HIV Infections virology, Lymph Nodes cytology, Macaca mulatta, B-Lymphocytes immunology, B-Lymphocytes virology, CD40 Ligand genetics, Gene Expression immunology, DNA, Viral metabolism, Lymphoid Tissue cytology, Lymphoid Tissue immunology, Lymphoid Tissue virology, Simian Acquired Immunodeficiency Syndrome immunology, Simian Acquired Immunodeficiency Syndrome virology, Simian Immunodeficiency Virus genetics, T Follicular Helper Cells immunology, T Follicular Helper Cells virology
- Abstract
CD4
+ T follicular helper (TFH ) cells are key targets for human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) replication and contribute to the virus reservoir under antiretroviral therapy (ART). Here, we describe a novel CD3+ CD20+ double-positive (DP) lymphocyte subset, resident in secondary lymphoid organs of humans and rhesus macaques (RMs), that appear predominantly after membrane exchange between TFH and B cells. DP lymphocytes are enriched in cells displaying a TFH phenotype (CD4+ PD1hi CXCR5hi ), function (interleukin 21 positive [IL-21+ ]), and gene expression profile. Importantly, expression of CD40L upon brief in vitro mitogen stimulation identifies, by specific gene-expression signatures, DP cells of TFH -cell origin versus those of B-cell origin. Analysis of 56 RMs showed that DP cells (i) significantly increase following SIV infection, (ii) are reduced after 12 months of ART in comparison to pre-ART levels, and (iii) expand to a significantly higher frequency following ART interruption. Quantification of total SIV-gag DNA on sorted DP cells from chronically infected RMs showed that these cells are susceptible to SIV infection. These data reinforce earlier observations that CD20+ T cells are infected and expanded by HIV infection, while suggesting that these cells phenotypically overlap activated CD4+ TFH cells that acquire CD20 expression via trogocytosis and can be targeted as part of therapeutic strategies aimed at HIV remission. IMPORTANCE The HIV reservoir is largely composed of latently infected memory CD4+ T cells that persist during antiretroviral therapy and constitute a major barrier toward HIV eradication. In particular, CD4+ T follicular helper cells have been demonstrated as key targets for viral replication and persistence under ART. In lymph nodes from HIV-infected humans and SIV-infected rhesus macaques, we show that CD3+ CD20+ lymphocytes emerge after membrane exchange between T cells and B cells and are enriched in phenotypic, functional, and gene expression profiles found in T follicular helper cells. Furthermore, in SIV-infected rhesus macaques, these cells expand following experimental infection and after interruption of ART and harbor SIV DNA at levels similar to those found in CD4+ T cells; thus, CD3+ CD20+ lymphocytes are susceptible to SIV infection and can contribute to SIV persistence., Competing Interests: The authors declare no conflict of interest.- Published
- 2023
- Full Text
- View/download PDF
29. Multivariate indicators of disease severity in COVID-19.
- Author
-
Bean J, Kuri-Cervantes L, Pennella M, Betts MR, Meyer NJ, and Hassan WM
- Subjects
- Humans, SARS-CoV-2, Neutrophils, Patient Acuity, HLA-DR Antigens, Severity of Illness Index, COVID-19
- Abstract
The novel coronavirus pandemic continues to cause significant morbidity and mortality around the world. Diverse clinical presentations prompted numerous attempts to predict disease severity to improve care and patient outcomes. Equally important is understanding the mechanisms underlying such divergent disease outcomes. Multivariate modeling was used here to define the most distinctive features that separate COVID-19 from healthy controls and severe from moderate disease. Using discriminant analysis and binary logistic regression models we could distinguish between severe disease, moderate disease, and control with rates of correct classifications ranging from 71 to 100%. The distinction of severe and moderate disease was most reliant on the depletion of natural killer cells and activated class-switched memory B cells, increased frequency of neutrophils, and decreased expression of the activation marker HLA-DR on monocytes in patients with severe disease. An increased frequency of activated class-switched memory B cells and activated neutrophils was seen in moderate compared to severe disease and control. Our results suggest that natural killer cells, activated class-switched memory B cells, and activated neutrophils are important for protection against severe disease. We show that binary logistic regression was superior to discriminant analysis by attaining higher rates of correct classification based on immune profiles. We discuss the utility of these multivariate techniques in biomedical sciences, contrast their mathematical basis and limitations, and propose strategies to overcome such limitations., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
30. Differential CD4+ T-Cell Cytokine and Cytotoxic Responses Between Reactivation and Latent Phases of Herpes Zoster Infection.
- Author
-
Jin W, Fang M, Sayin I, Smith C, Hunter JL, Richardson B, Golden JB, Haley C, Schmader KE, Betts MR, Tyring SK, Cameron CM, Cameron MJ, and Canaday DH
- Abstract
Background: CD4+ T cells are a critical component of effective immune responses to varicella zoster virus (VZV), but their functional properties during the reactivation acute vs latent phase of infection remain poorly defined., Methods: Here we assessed the functional and transcriptomic properties of peripheral blood CD4+ T cells in persons with acute herpes zoster (HZ) compared to those with a prior history of HZ infection using multicolor flow cytometry and RNA sequencing., Results: We found significant differences between the polyfunctionality of VZV-specific total memory, effector memory, and central memory CD4+ T cells in acute vs prior HZ. VZV-specific CD4+ memory T-cell responses in acute HZ reactivation had higher frequencies of IFN-γ and IL-2 producing cells compared to those with prior HZ. In addition, cytotoxic markers were higher in VZV-specific CD4+ T cells than non-VZV-specific cells. Transcriptomic analysis of ex vivo total memory CD4+ T cells from these individuals showed differential regulation of T-cell survival and differentiation pathways, including TCR, cytotoxic T lymphocytes (CTL), T helper, inflammation, and MTOR signaling pathways. These gene signatures correlated with the frequency of IFN-γ and IL-2 producing cells responding to VZV., Conclusions: In summary, VZV-specific CD4+ T cells from acute HZ individuals had unique functional and transcriptomic features, and VZV-specific CD4+ T cells as a group had a higher expression of cytotoxic molecules including Perforin, Granzyme-B, and CD107a., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2023 Pathogens and Immunity.)
- Published
- 2023
- Full Text
- View/download PDF
31. Profound phenotypic and epigenetic heterogeneity of the HIV-1-infected CD4 + T cell reservoir.
- Author
-
Wu VH, Nordin JML, Nguyen S, Joy J, Mampe F, Del Rio Estrada PM, Torres-Ruiz F, González-Navarro M, Luna-Villalobos YA, Ávila-Ríos S, Reyes-Terán G, Tebas P, Montaner LJ, Bar KJ, Vella LA, and Betts MR
- Subjects
- Humans, CD4-Positive T-Lymphocytes, Virus Latency genetics, Epigenesis, Genetic, Viral Load, Anti-Retroviral Agents therapeutic use, HIV-1 physiology, HIV Infections drug therapy, HIV Infections genetics
- Abstract
Understanding the complexity of the long-lived HIV reservoir during antiretroviral therapy (ART) remains a considerable impediment in research towards a cure for HIV. To address this, we developed a single-cell strategy to precisely define the unperturbed peripheral blood HIV-infected memory CD4
+ T cell reservoir from ART-treated people living with HIV (ART-PLWH) via the presence of integrated accessible proviral DNA in concert with epigenetic and cell surface protein profiling. We identified profound reservoir heterogeneity within and between ART-PLWH, characterized by new and known surface markers within total and individual memory CD4+ T cell subsets. We further uncovered new epigenetic profiles and transcription factor motifs enriched in HIV-infected cells that suggest infected cells with accessible provirus, irrespective of reservoir distribution, are poised for reactivation during ART treatment. Together, our findings reveal the extensive inter- and intrapersonal cellular heterogeneity of the HIV reservoir, and establish an initial multiomic atlas to develop targeted reservoir elimination strategies., (© 2022. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
32. Validating a Proteomic Signature of Severe COVID-19.
- Author
-
Cosgriff CV, Miano TA, Mathew D, Huang AC, Giannini HM, Kuri-Cervantes L, Pampena MB, Ittner CAG, Weisman AR, Agyekum RS, Dunn TG, Oniyide O, Turner AP, D'Andrea K, Adamski S, Greenplate AR, Anderson BJ, Harhay MO, Jones TK, Reilly JP, Mangalmurti NS, Shashaty MGS, Betts MR, Wherry EJ, and Meyer NJ
- Abstract
COVID-19 is a heterogenous disease. Biomarker-based approaches may identify patients at risk for severe disease, who may be more likely to benefit from specific therapies. Our objective was to identify and validate a plasma protein signature for severe COVID-19., Design: Prospective observational cohort study., Setting: Two hospitals in the United States., Patients: One hundred sixty-seven hospitalized adults with COVID-19., Intervention: None., Measurements and Main Results: We measured 713 plasma proteins in 167 hospitalized patients with COVID-19 using a high-throughput platform. We classified patients as nonsevere versus severe COVID-19, defined as the need for high-flow nasal cannula, mechanical ventilation, extracorporeal membrane oxygenation, or death, at study entry and in 7-day intervals thereafter. We compared proteins measured at baseline between these two groups by logistic regression adjusting for age, sex, symptom duration, and comorbidities. We used lead proteins from dysregulated pathways as inputs for elastic net logistic regression to identify a parsimonious signature of severe disease and validated this signature in an external COVID-19 dataset. We tested whether the association between corticosteroid use and mortality varied by protein signature. One hundred ninety-four proteins were associated with severe COVID-19 at the time of hospital admission. Pathway analysis identified multiple pathways associated with inflammatory response and tissue repair programs. Elastic net logistic regression yielded a 14-protein signature that discriminated 90-day mortality in an external cohort with an area under the receiver-operator characteristic curve of 0.92 (95% CI, 0.88-0.95). Classifying patients based on the predicted risk from the signature identified a heterogeneous response to treatment with corticosteroids ( p = 0.006)., Conclusions: Inpatients with COVID-19 express heterogeneous patterns of plasma proteins. We propose a 14-protein signature of disease severity that may have value in developing precision medicine approaches for COVID-19 pneumonia., Competing Interests: Dr. Wherry has consulting agreements with and/or is on the scientific advisory board for Merck, Roche, Pieris, Elstar, and Surface Oncology. He is a founder of Surface Oncology and Arsenal Biosciences. EJW has a patent licensing agreement on the PD-1 pathway with Roche/Genetech. He is an inventor on U.S. patent number 10,270,446 submitted by Emory University that covers the use of programmed death ligand 1 blockade to treat infections and cancer. Dr. Meyer receives funding to her institution from Quantum Leap Healthcare Consortium, Athersys, Inc, Biomarck Inc, and the Marcus Foundation for work unrelated to this article. The remaining authors have disclosed that they do not have any potential conflicts of interest., (Copyright © 2022 The Authors. Published by Wolters Kluwer Health, Inc. on behalf of the Society of Critical Care Medicine.)
- Published
- 2022
- Full Text
- View/download PDF
33. The immune synapses reveal aberrant functions of CD8 T cells during chronic HIV infection.
- Author
-
Anikeeva N, Steblyanko M, Kuri-Cervantes L, Buggert M, Betts MR, and Sykulev Y
- Subjects
- Humans, CD8-Positive T-Lymphocytes, Lymphocyte Count, Cell Differentiation, Synapses, HIV Infections
- Abstract
Chronic HIV infection causes persistent low-grade inflammation that induces premature aging of the immune system including senescence of memory and effector CD8 T cells. To uncover the reasons of gradually diminished potency of CD8 T cells from people living with HIV, here we expose the T cells to planar lipid bilayers containing ligands for T-cell receptor and a T-cell integrins and analyze the cellular morphology, dynamics of synaptic interface formation and patterns of the cellular degranulation. We find a large fraction of phenotypically naive T cells from chronically infected people are capable to form mature synapse with focused degranulation, a signature of a differentiated T cells. Further, differentiation of aberrant naive T cells may lead to the development of anomalous effector T cells undermining their capacity to control HIV and other pathogens that could be contained otherwise., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
34. Limited impact of fingolimod treatment during the initial weeks of ART in SIV-infected rhesus macaques.
- Author
-
Pino M, Pagliuzza A, Pampena MB, Deleage C, Viox EG, Nguyen K, Shim I, Zhang A, Harper JL, Samer S, King CT, Cervasi B, Gill KP, Ehnert S, Jean SM, Freeman ML, Lifson JD, Kulpa D, Betts MR, Chomont N, Lederman MM, and Paiardini M
- Subjects
- Animals, Anti-Retroviral Agents, CD4-Positive T-Lymphocytes, Fingolimod Hydrochloride, Macaca mulatta, Viral Load, HIV Infections, Simian Acquired Immunodeficiency Syndrome, Simian Immunodeficiency Virus
- Abstract
Antiretroviral therapy (ART) is not curative due to the persistence of a reservoir of HIV-infected cells, particularly in tissues such as lymph nodes, with the potential to cause viral rebound after treatment cessation. In this study, fingolimod (FTY720), a lysophospholipid sphingosine-1-phosphate receptor modulator is administered to SIV-infected rhesus macaques at initiation of ART to block the egress from lymphoid tissues of natural killer and T-cells, thereby promoting proximity between cytolytic cells and infected CD4+ T-cells. When compared with the ART-only controls, FTY720 treatment during the initial weeks of ART induces a profound lymphopenia and increases frequencies of CD8+ T-cells expressing perforin in lymph nodes, but not their killing capacity; FTY720 also increases frequencies of cytolytic NK cells in lymph nodes. This increase of cytolytic cells, however, does not limit measures of viral persistence during ART, including intact proviral genomes. After ART interruption, a subset of animals that initially receives FTY720 displays a modest delay in viral rebound, with reduced plasma viremia and frequencies of infected T follicular helper cells. Further research is needed to optimize the potential utility of FTY720 when coupled with strategies that boost the antiviral function of T-cells in lymphoid tissues., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
35. Cytomegalovirus Latent Infection is Associated with an Increased Risk of COVID-19-Related Hospitalization.
- Author
-
Alanio C, Verma A, Mathew D, Gouma S, Liang G, Dunn T, Oldridge DA, Weaver J, Kuri-Cervantes L, Pampena MB, Betts MR, Collman RG, Bushman FD, Meyer NJ, Hensley SE, Rader D, and Wherry EJ
- Subjects
- Cytomegalovirus, Hospitalization, Humans, SARS-CoV-2, COVID-19, Cytomegalovirus Infections, Latent Infection
- Abstract
Some risk factors for severe coronavirus disease 2019 (COVID-19) have been identified, including age, race, and obesity. However, 20%-50% of severe cases occur in the absence of these factors. Cytomegalovirus (CMV) is a herpesvirus that infects about 50% of all individuals worldwide and is among the most significant nongenetic determinants of immune system. We hypothesized that latent CMV infection might influence the severity of COVID-19. Our analyses demonstrate that CMV seropositivity is associated with more than twice the risk of hospitalization due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Immune profiling of blood and CMV DNA quantitative polymerase chain reaction in a subset of patients for whom respiratory tract samples were available revealed altered T-cell activation profiles in absence of extensive CMV replication in the upper respiratory tract. These data suggest a potential role for CMV-driven immune perturbations in affecting the outcome of SARS-CoV-2 infection and may have implications for the discrepancies in COVID-19 severity between different human populations., (© The Author(s) 2022. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
36. Innate lymphoid cells and COVID-19 severity in SARS-CoV-2 infection.
- Author
-
Silverstein NJ, Wang Y, Manickas-Hill Z, Carbone C, Dauphin A, Boribong BP, Loiselle M, Davis J, Leonard MM, Kuri-Cervantes L, Meyer NJ, Betts MR, Li JZ, Walker BD, Yu XG, Yonker LM, and Luban J
- Subjects
- Amphiregulin, Child, Female, Humans, Immunity, Innate, Inflammation, Male, SARS-CoV-2, Systemic Inflammatory Response Syndrome, T-Lymphocyte Subsets, COVID-19 complications, Lymphopenia
- Abstract
Background: Risk of severe COVID-19 increases with age, is greater in males, and is associated with lymphopenia, but not with higher burden of SARS-CoV-2. It is unknown whether effects of age and sex on abundance of specific lymphoid subsets explain these correlations., Methods: Multiple regression was used to determine the relationship between abundance of specific blood lymphoid cell types, age, sex, requirement for hospitalization, duration of hospitalization, and elevation of blood markers of systemic inflammation, in adults hospitalized for severe COVID-19 (n = 40), treated for COVID-19 as outpatients (n = 51), and in uninfected controls (n = 86), as well as in children with COVID-19 (n = 19), recovering from COVID-19 (n = 14), MIS-C (n = 11), recovering from MIS-C (n = 7), and pediatric controls (n = 17)., Results: This observational study found that the abundance of innate lymphoid cells (ILCs) decreases more than 7-fold over the human lifespan - T cell subsets decrease less than 2-fold - and is lower in males than in females. After accounting for effects of age and sex, ILCs, but not T cells, were lower in adults hospitalized with COVID-19, independent of lymphopenia. Among SARS-CoV-2-infected adults, the abundance of ILCs, but not of T cells, correlated inversely with odds and duration of hospitalization, and with severity of inflammation. ILCs were also uniquely decreased in pediatric COVID-19 and the numbers of these cells did not recover during follow-up. In contrast, children with MIS-C had depletion of both ILCs and T cells, and both cell types increased during follow-up. In both pediatric COVID-19 and MIS-C, ILC abundance correlated inversely with inflammation. Blood ILC mRNA and phenotype tracked closely with ILCs from lung. Importantly, blood ILCs produced amphiregulin, a protein implicated in disease tolerance and tissue homeostasis. Among controls, the percentage of ILCs that produced amphiregulin was higher in females than in males, and people hospitalized with COVID-19 had a lower percentage of ILCs that produced amphiregulin than did controls., Conclusions: These results suggest that, by promoting disease tolerance, homeostatic ILCs decrease morbidity and mortality associated with SARS-CoV-2 infection, and that lower ILC abundance contributes to increased COVID-19 severity with age and in males., Funding: This work was supported in part by the Massachusetts Consortium for Pathogen Readiness and NIH grants R37AI147868, R01AI148784, F30HD100110, 5K08HL143183., Competing Interests: NS, YW, ZM, CC, AD, BB, ML, JD, ML, LK, NM, MB, JL, BW, XY, LY, JL No competing interests declared, (© 2022, Silverstein et al.)
- Published
- 2022
- Full Text
- View/download PDF
37. Prednisolone reduces the interferon response to AAV in cynomolgus macaques and may increase liver gene expression.
- Author
-
Wang L, Warzecha CC, Kistner A, Chichester JA, Bell P, Buza EL, He Z, Pampena MB, Couthouis J, Sethi S, McKeever K, Betts MR, Kakkis E, Wilson JM, Wadsworth S, and Sullivan BA
- Abstract
Ornithine transcarbamylase deficiency is a rare X-linked genetic urea cycle disorder leading to episodes of acute hyperammonemia, adverse cognitive and neurological effects, hospitalizations, and in some cases death. DTX301, a non-replicating, recombinant self-complimentary adeno-associated virus vector serotype 8 (scAAV8)-encoding human ornithine transcarbamylase, is a promising gene therapy for ornithine transcarbamylase deficiency; however, the impact of sex and prophylactic immunosuppression on ornithine transcarbamylase gene therapy outcomes is not well characterized. This study sought to describe the impact of sex and immunosuppression in adult, sexually mature female and male cynomolgus macaques through day 140 after DTX301 administration. Four study groups (n = 3/group) were included: male non-immunosuppressed; male immunosuppressed; female non-immunosuppressed; and female immunosuppressed. DTX301 was well tolerated with and without immunosuppression; no notable differences were observed between female and male groups across outcome measures. Prednisolone-treated animals exhibited a trend toward greater vector genome and transgene expression, although the differences were not statistically significant. The hepatic interferon gene signature was significantly decreased in prednisolone-treated animals, and a significant inverse relationship was observed between interferon gene signature levels and hepatic vector DNA and transgene RNA. These observations were not sustained upon immunosuppression withdrawal. Further studies may determine whether the observed effect can be prolonged., Competing Interests: A.K., J.C., K.M., S.W., E.K., and B.A.S. are employees of Ultragenyx Pharmaceutical Inc. J.M.W. is a paid advisor to and holds equity in Scout Bio and Passage Bio; he also has sponsored research agreements with Amicus Therapeutics, Biogen, Elaaj Bio, FA212, Janssen, Passage Bio, and Scout Bio, which are licensees of Penn technology. He also has a sponsored research agreement with G2 Bio. J.M.W. holds equity in the G2-Bio-associated asset companies. J.M.W. and L.W. are inventors on patents that have been licensed to various biopharmaceutical companies and for which they may receive payments., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
38. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern.
- Author
-
Goel RR, Painter MM, Apostolidis SA, Mathew D, Meng W, Rosenfeld AM, Lundgreen KA, Reynaldi A, Khoury DS, Pattekar A, Gouma S, Kuri-Cervantes L, Hicks P, Dysinger S, Hicks A, Sharma H, Herring S, Korte S, Baxter AE, Oldridge DA, Giles JR, Weirick ME, McAllister CM, Awofolaju M, Tanenbaum N, Drapeau EM, Dougherty J, Long S, D'Andrea K, Hamilton JT, McLaughlin M, Williams JC, Adamski S, Kuthuru O, Frank I, Betts MR, Vella LA, Grifoni A, Weiskopf D, Sette A, Hensley SE, Davenport MP, Bates P, Luning Prak ET, Greenplate AR, and Wherry EJ
- Subjects
- Humans, COVID-19 Vaccines immunology, Immunologic Memory, SARS-CoV-2 genetics, SARS-CoV-2 immunology, mRNA Vaccines immunology
- Abstract
The durability of immune memory after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) messenger RNA (mRNA) vaccination remains unclear. In this study, we longitudinally profiled vaccine responses in SARS-CoV-2–naïve and –recovered individuals for 6 months after vaccination. Antibodies declined from peak levels but remained detectable in most subjects at 6 months. By contrast, mRNA vaccines generated functional memory B cells that increased from 3 to 6 months postvaccination, with the majority of these cells cross-binding the Alpha, Beta, and Delta variants. mRNA vaccination further induced antigen-specific CD4
+ and CD8+ T cells, and early CD4+ T cell responses correlated with long-term humoral immunity. Recall responses to vaccination in individuals with preexisting immunity primarily increased antibody levels without substantially altering antibody decay rates. Together, these findings demonstrate robust cellular immune memory to SARS-CoV-2 and its variants for at least 6 months after mRNA vaccination.- Published
- 2021
- Full Text
- View/download PDF
39. Siglec-9 defines and restrains a natural killer subpopulation highly cytotoxic to HIV-infected cells.
- Author
-
Adeniji OS, Kuri-Cervantes L, Yu C, Xu Z, Ho M, Chew GM, Shikuma C, Tomescu C, George AF, Roan NR, Ndhlovu LC, Liu Q, Muthumani K, Weiner DB, Betts MR, Xiao H, and Abdel-Mohsen M
- Subjects
- Antigens, CD genetics, HIV Infections immunology, HIV Infections metabolism, HIV Infections virology, Humans, Killer Cells, Natural metabolism, Sialic Acid Binding Immunoglobulin-like Lectins genetics, Viremia immunology, Viremia metabolism, Viremia virology, Antigens, CD metabolism, CD56 Antigen immunology, HIV physiology, HIV Infections pathology, Killer Cells, Natural immunology, Sialic Acid Binding Immunoglobulin-like Lectins metabolism, Viral Load, Viremia pathology
- Abstract
Siglec-9 is an MHC-independent inhibitory receptor expressed on a subset of natural killer (NK) cells. Siglec-9 restrains NK cytotoxicity by binding to sialoglycans (sialic acid-containing glycans) on target cells. Despite the importance of Siglec-9 interactions in tumor immune evasion, their role as an immune evasion mechanism during HIV infection has not been investigated. Using in vivo phenotypic analyses, we found that Siglec-9+ CD56dim NK cells, during HIV infection, exhibit an activated phenotype with higher expression of activating receptors and markers (NKp30, CD38, CD16, DNAM-1, perforin) and lower expression of the inhibitory receptor NKG2A, compared to Siglec-9- CD56dim NK cells. We also found that levels of Siglec-9+ CD56dim NK cells inversely correlate with viral load during viremic infection and CD4+ T cell-associated HIV DNA during suppressed infection. Using in vitro cytotoxicity assays, we confirmed that Siglec-9+ NK cells exhibit higher cytotoxicity towards HIV-infected cells compared to Siglec-9- NK cells. These data are consistent with the notion that Siglec-9+ NK cells are highly cytotoxic against HIV-infected cells. However, blocking Siglec-9 enhanced NK cells' ability to lyse HIV-infected cells, consistent with the known inhibitory function of the Siglec-9 molecule. Together, these data support a model in which the Siglec-9+ CD56dim NK subpopulation is highly cytotoxic against HIV-infected cells even whilst being restrained by the inhibitory effects of Siglec-9. To harness the cytotoxic capacity of the Siglec-9+ NK subpopulation, which is dampened by Siglec-9, we developed a proof-of-concept approach to selectively disrupt Siglec/sialoglycan interactions between NK and HIV-infected cells. We achieved this goal by conjugating Sialidase to several HIV broadly neutralizing antibodies. These conjugates selectively desialylated HIV-infected cells and enhanced NK cells' capacity to kill them. In summary, we identified a novel, glycan-based interaction that may contribute to HIV-infected cells' ability to evade NK immunosurveillance and developed an approach to break this interaction., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
40. Cellular and humoral immune responses following SARS-CoV-2 mRNA vaccination in patients with multiple sclerosis on anti-CD20 therapy.
- Author
-
Apostolidis SA, Kakara M, Painter MM, Goel RR, Mathew D, Lenzi K, Rezk A, Patterson KR, Espinoza DA, Kadri JC, Markowitz DM, E Markowitz C, Mexhitaj I, Jacobs D, Babb A, Betts MR, Prak ETL, Weiskopf D, Grifoni A, Lundgreen KA, Gouma S, Sette A, Bates P, Hensley SE, Greenplate AR, Wherry EJ, Li R, and Bar-Or A
- Subjects
- Animals, Antibodies, Monoclonal therapeutic use, Antibodies, Viral analysis, Antibodies, Viral blood, Antigens, CD20 immunology, COVID-19 prevention & control, Case-Control Studies, Chlorocebus aethiops, HEK293 Cells, Humans, Immunity, Cellular, Immunity, Humoral drug effects, Immunity, Humoral physiology, Immunotherapy methods, Longitudinal Studies, Multiple Sclerosis blood, RNA, Messenger immunology, RNA, Viral immunology, Rituximab pharmacology, Rituximab therapeutic use, SARS-CoV-2 genetics, Vaccination, Vero Cells, COVID-19 Vaccines therapeutic use, Multiple Sclerosis immunology, Multiple Sclerosis therapy, SARS-CoV-2 immunology
- Abstract
SARS-CoV-2 messenger RNA vaccination in healthy individuals generates immune protection against COVID-19. However, little is known about SARS-CoV-2 mRNA vaccine-induced responses in immunosuppressed patients. We investigated induction of antigen-specific antibody, B cell and T cell responses longitudinally in patients with multiple sclerosis (MS) on anti-CD20 antibody monotherapy (n = 20) compared with healthy controls (n = 10) after BNT162b2 or mRNA-1273 mRNA vaccination. Treatment with anti-CD20 monoclonal antibody (aCD20) significantly reduced spike-specific and receptor-binding domain (RBD)-specific antibody and memory B cell responses in most patients, an effect ameliorated with longer duration from last aCD20 treatment and extent of B cell reconstitution. By contrast, all patients with MS treated with aCD20 generated antigen-specific CD4 and CD8 T cell responses after vaccination. Treatment with aCD20 skewed responses, compromising circulating follicular helper T (T
FH ) cell responses and augmenting CD8 T cell induction, while preserving type 1 helper T (TH 1) cell priming. Patients with MS treated with aCD20 lacking anti-RBD IgG had the most severe defect in circulating TFH responses and more robust CD8 T cell responses. These data define the nature of the SARS-CoV-2 vaccine-induced immune landscape in aCD20-treated patients and provide insights into coordinated mRNA vaccine-induced immune responses in humans. Our findings have implications for clinical decision-making and public health policy for immunosuppressed patients including those treated with aCD20., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
41. Innate lymphoid cells and disease tolerance in SARS-CoV-2 infection.
- Author
-
Silverstein NJ, Wang Y, Manickas-Hill Z, Carbone C, Dauphin A, Boribong BP, Loiselle M, Davis J, Leonard MM, Kuri-Cervantes L, Meyer NJ, Betts MR, Li JZ, Walker B, Yu XG, Yonker LM, and Luban J
- Abstract
Risk of severe COVID-19 increases with age, is greater in males, and is associated with lymphopenia, but not with higher burden of SARS-CoV-2. It is unknown whether effects of age and sex on abundance of specific lymphoid subsets explain these correlations. This study found that the abundance of innate lymphoid cells (ILCs) decreases more than 7-fold over the human lifespan - T cell subsets decrease less than 2-fold - and is lower in males than in females. After accounting for effects of age and sex, ILCs, but not T cells, were lower in adults hospitalized with COVID-19, independent of lymphopenia. Among SARS-CoV-2-infected adults, the abundance of ILCs, but not of T cells, correlated inversely with odds and duration of hospitalization, and with severity of inflammation. ILCs were also uniquely decreased in pediatric COVID-19 and the numbers of these cells did not recover during follow-up. In contrast, children with MIS-C had depletion of both ILCs and T cells, and both cell types increased during follow-up. In both pediatric COVID-19 and MIS-C, ILC abundance correlated inversely with inflammation. Blood ILC mRNA and phenotype tracked closely with ILCs from lung. Importantly, blood ILCs produced amphiregulin, a protein implicated in disease tolerance and tissue homeostasis, and the percentage of amphiregulin-producing ILCs was higher in females than in males. These results suggest that, by promoting disease tolerance, homeostatic ILCs decrease morbidity and mortality associated with SARS-CoV-2 infection, and that lower ILC abundance accounts for increased COVID-19 severity with age and in males.
- Published
- 2021
- Full Text
- View/download PDF
42. Rapid induction of antigen-specific CD4 + T cells is associated with coordinated humoral and cellular immunity to SARS-CoV-2 mRNA vaccination.
- Author
-
Painter MM, Mathew D, Goel RR, Apostolidis SA, Pattekar A, Kuthuru O, Baxter AE, Herati RS, Oldridge DA, Gouma S, Hicks P, Dysinger S, Lundgreen KA, Kuri-Cervantes L, Adamski S, Hicks A, Korte S, Giles JR, Weirick ME, McAllister CM, Dougherty J, Long S, D'Andrea K, Hamilton JT, Betts MR, Bates P, Hensley SE, Grifoni A, Weiskopf D, Sette A, Greenplate AR, and Wherry EJ
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Adult, Antibodies, Neutralizing blood, Antibodies, Viral blood, Antigens, CD metabolism, Antigens, Differentiation, T-Lymphocyte metabolism, BNT162 Vaccine, Female, Humans, Immunity, Cellular, Immunity, Humoral, Immunization, Secondary, Immunologic Memory, Lectins, C-Type metabolism, Lymphocyte Activation, Male, Middle Aged, Peptides immunology, Spike Glycoprotein, Coronavirus immunology, Vaccination, Young Adult, CD8-Positive T-Lymphocytes immunology, COVID-19 immunology, COVID-19 Vaccines immunology, SARS-CoV-2 physiology, Th1 Cells immunology
- Abstract
SARS-CoV-2 mRNA vaccines have shown remarkable clinical efficacy, but questions remain about the nature and kinetics of T cell priming. We performed longitudinal antigen-specific T cell analyses on healthy SARS-CoV-2-naive and recovered individuals prior to and following mRNA prime and boost vaccination. Vaccination induced rapid antigen-specific CD4
+ T cell responses in naive subjects after the first dose, whereas CD8+ T cell responses developed gradually and were variable in magnitude. Vaccine-induced Th1 and Tfh cell responses following the first dose correlated with post-boost CD8+ T cells and neutralizing antibodies, respectively. Integrated analysis revealed coordinated immune responses with distinct trajectories in SARS-CoV-2-naive and recovered individuals. Last, whereas booster vaccination improved T cell responses in SARS-CoV-2-naive subjects, the second dose had little effect in SARS-CoV-2-recovered individuals. These findings highlight the role of rapidly primed CD4+ T cells in coordinating responses to the second vaccine dose in SARS-CoV-2-naive individuals., Competing Interests: Declaration of interests S.E.H. has received consultancy fees from Sanofi Pasteur, Lumen, Novavax, and Merk for work unrelated to this study. E.J.W. is a consultant or an adviser for Merck, Elstar, Janssen, Related Sciences, Synthekine, and Surface Oncology. E.J.W. is a founder of Surface Oncology and Arsenal Biosciences. E.J.W. is an inventor on a patent (U.S. Patent No. 10,370,446) submitted by Emory University that covers the use of PD-1 blockade to treat infections and cancer. A.S. is a consultant for Gritstone, Flow Pharma, CellCarta, Arcturus, Oxfordimmunotech, and Avalia. La Jolla Institute for Immunology has filed for patent protection for various aspects of T cell epitope and vaccine design work., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
43. An AAV-based, room-temperature-stable, single-dose COVID-19 vaccine provides durable immunogenicity and protection in non-human primates.
- Author
-
Zabaleta N, Dai W, Bhatt U, Hérate C, Maisonnasse P, Chichester JA, Sanmiguel J, Estelien R, Michalson KT, Diop C, Maciorowski D, Dereuddre-Bosquet N, Cavarelli M, Gallouët AS, Naninck T, Kahlaoui N, Lemaitre J, Qi W, Hudspeth E, Cucalon A, Dyer CD, Pampena MB, Knox JJ, LaRocque RC, Charles RC, Li D, Kim M, Sheridan A, Storm N, Johnson RI, Feldman J, Hauser BM, Contreras V, Marlin R, Tsong Fang RH, Chapon C, van der Werf S, Zinn E, Ryan A, Kobayashi DT, Chauhan R, McGlynn M, Ryan ET, Schmidt AG, Price B, Honko A, Griffiths A, Yaghmour S, Hodge R, Betts MR, Freeman MW, Wilson JM, Le Grand R, and Vandenberghe LH
- Subjects
- Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 Vaccines administration & dosage, COVID-19 Vaccines genetics, Dependovirus genetics, Dependovirus metabolism, Female, Humans, Immunogenicity, Vaccine immunology, Immunologic Memory immunology, Macaca fascicularis, Macaca mulatta, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, T-Lymphocytes immunology, Transgenes genetics, Vaccination methods, Viral Load immunology, Antibodies, Neutralizing blood, Antibodies, Viral blood, COVID-19 prevention & control, COVID-19 Vaccines immunology, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
The SARS-CoV-2 pandemic has affected more than 185 million people worldwide resulting in over 4 million deaths. To contain the pandemic, there is a continued need for safe vaccines that provide durable protection at low and scalable doses and can be deployed easily. Here, AAVCOVID-1, an adeno-associated viral (AAV), spike-gene-based vaccine candidate demonstrates potent immunogenicity in mouse and non-human primates following a single injection and confers complete protection from SARS-CoV-2 challenge in macaques. Peak neutralizing antibody titers are sustained at 1 year and complemented by functional memory T cell responses. The AAVCOVID vector has no relevant pre-existing immunity in humans and does not elicit cross-reactivity to common AAVs used in gene therapy. Vector genome persistence and expression wanes following injection. The single low-dose requirement, high-yield manufacturability, and 1-month stability for storage at room temperature may make this technology well suited to support effective immunization campaigns for emerging pathogens on a global scale., Competing Interests: Declaration of interests J.M.W. is a paid advisor to and holds equity in Scout Bio and Passage Bio; he holds equity in Surmount Bio; he also has sponsored research agreements with Amicus Therapeutics, Biogen, Elaaj Bio, Janssen, Moderna, Passage Bio, Regeneron, Scout Bio, Surmount Bio, and Ultragenyx, which are licensees of Penn technology. J.M.W. had a sponsored research agreement with Albamunity that funded this work. He also has a sponsored research agreement with G2 Bio. L.H.V. and J.M.W. are inventors on patents that have been licensed to various biopharmaceutical companies and for which they may receive payments including vector and vaccine technologies herein described. L.H.V., W.D., U.B., N.Z. are named inventors on two patent applications relevant to AAVCOVID. W.Q., E.H., S.Y., and R.H. are employees of Novartis. M.W.F. is a paid consultant to 5AM Ventures and to Mitobridge/Astellas. L.H.V. is a paid advisor to Novartis, Akouos, and Affinia Therapeutics and serves on the Board of Directors of Affinia, Addgene, and Odylia Therapeutics. L.H.V. holds equity in Akouos and Affinia and receives sponsored research funding from Albamunity Inc. to which he is an unpaid consultant. M.R.B. receives consulting fees from Interius Biotherapeutics. R.C.L. is a subcontractor with the CDC Foundation and receives royalties from UpToDate., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
44. mRNA Vaccination Induces Durable Immune Memory to SARS-CoV-2 with Continued Evolution to Variants of Concern.
- Author
-
Goel RR, Painter MM, Apostolidis SA, Mathew D, Meng W, Rosenfeld AM, Lundgreen KA, Reynaldi A, Khoury DS, Pattekar A, Gouma S, Kuri-Cervantes L, Hicks P, Dysinger S, Hicks A, Sharma H, Herring S, Korte S, Baxter AE, Oldridge DA, Giles JR, Weirick ME, McAllister CM, Awofolaju M, Tanenbaum N, Drapeau EM, Dougherty J, Long S, D'Andrea K, Hamilton JT, McLaughlin M, Williams JC, Adamski S, Kuthuru O, Frank I, Betts MR, Vella LA, Grifoni A, Weiskopf D, Sette A, Hensley SE, Davenport MP, Bates P, Luning Prak ET, Greenplate AR, and Wherry EJ
- Abstract
SARS-CoV-2 mRNA vaccines have shown remarkable efficacy, especially in preventing severe illness and hospitalization. However, the emergence of several variants of concern and reports of declining antibody levels have raised uncertainty about the durability of immune memory following vaccination. In this study, we longitudinally profiled both antibody and cellular immune responses in SARS-CoV-2 naïve and recovered individuals from pre-vaccine baseline to 6 months post-mRNA vaccination. Antibody and neutralizing titers decayed from peak levels but remained detectable in all subjects at 6 months post-vaccination. Functional memory B cell responses, including those specific for the receptor binding domain (RBD) of the Alpha (B.1.1.7), Beta (B.1.351), and Delta (B.1.617.2) variants, were also efficiently generated by mRNA vaccination and continued to increase in frequency between 3 and 6 months post-vaccination. Notably, most memory B cells induced by mRNA vaccines were capable of cross-binding variants of concern, and B cell receptor sequencing revealed significantly more hypermutation in these RBD variant-binding clones compared to clones that exclusively bound wild-type RBD. Moreover, the percent of variant cross-binding memory B cells was higher in vaccinees than individuals who recovered from mild COVID-19. mRNA vaccination also generated antigen-specific CD8+ T cells and durable memory CD4+ T cells in most individuals, with early CD4+ T cell responses correlating with humoral immunity at later timepoints. These findings demonstrate robust, multi-component humoral and cellular immune memory to SARS-CoV-2 and current variants of concern for at least 6 months after mRNA vaccination. Finally, we observed that boosting of pre-existing immunity with mRNA vaccination in SARS-CoV-2 recovered individuals primarily increased antibody responses in the short-term without significantly altering antibody decay rates or long-term B and T cell memory. Together, this study provides insights into the generation and evolution of vaccine-induced immunity to SARS-CoV-2, including variants of concern, and has implications for future booster strategies.
- Published
- 2021
- Full Text
- View/download PDF
45. Erythrocytes identify complement activation in patients with COVID-19.
- Author
-
Lam LKM, Reilly JP, Rux AH, Murphy SJ, Kuri-Cervantes L, Weisman AR, Ittner CAG, Pampena MB, Betts MR, Wherry EJ, Song WC, Lambris JD, Meyer NJ, Cines DB, and Mangalmurti NS
- Subjects
- COVID-19 immunology, COVID-19 virology, Complement C3b metabolism, Complement C4b metabolism, Erythrocytes metabolism, Erythrocytes virology, Female, Humans, Male, Middle Aged, Peptide Fragments metabolism, Respiratory Insufficiency immunology, Respiratory Insufficiency metabolism, Respiratory Insufficiency virology, SARS-CoV-2 isolation & purification, Sepsis immunology, Sepsis metabolism, Sepsis virology, COVID-19 complications, Complement Activation immunology, Complement C3b immunology, Complement C4b immunology, Erythrocytes immunology, Peptide Fragments immunology, Respiratory Insufficiency diagnosis, Sepsis diagnosis
- Abstract
COVID-19, the disease caused by the SARS-CoV-2 virus, can progress to multisystem organ failure and viral sepsis characterized by respiratory failure, arrhythmias, thromboembolic complications, and shock with high mortality. Autopsy and preclinical evidence implicate aberrant complement activation in endothelial injury and organ failure. Erythrocytes express complement receptors and are capable of binding immune complexes; therefore, we investigated complement activation in patients with COVID-19 using erythrocytes as a tool to diagnose complement activation. We discovered enhanced C3b and C4d deposition on erythrocytes in COVID-19 sepsis patients and non-COVID sepsis patients compared with healthy controls, supporting the role of complement in sepsis-associated organ injury. Our data suggest that erythrocytes may contribute to a precision medicine approach to sepsis and have diagnostic value in monitoring complement dysregulation in COVID-19-sepsis and non-COVID sepsis and identifying patients who may benefit from complement targeted therapies.
- Published
- 2021
- Full Text
- View/download PDF
46. Role of nuclear localization in the regulation and function of T-bet and Eomes in exhausted CD8 T cells.
- Author
-
McLane LM, Ngiow SF, Chen Z, Attanasio J, Manne S, Ruthel G, Wu JE, Staupe RP, Xu W, Amaravadi RK, Xu X, Karakousis GC, Mitchell TC, Schuchter LM, Huang AC, Freedman BD, Betts MR, and Wherry EJ
- Subjects
- Animals, Cell Differentiation, Humans, Mice, Signal Transduction, CD8-Positive T-Lymphocytes metabolism, T-Box Domain Proteins metabolism
- Abstract
The transcription factors T-bet and Eomesodermin (Eomes) regulate CD8 T cell exhaustion through undefined mechanisms. Here, we show that the subcellular localization of T-bet and Eomes dictate their regulatory activity in exhausted T cells (T
EX s). TEX s had a higher ratio of nuclear Eomes:T-bet than memory T cells (TMEM s) during chronic lymphocytic choriomeningitis virus (LCMV) infection in preclinical cancer models and in human tumors. Biochemically, T-bet and Eomes compete for the same DNA sequences, including the Pdcd1 T-box. High nuclear T-bet strongly represses Pdcd1 transcription in TMEM , whereas low nuclear T-bet in TEX leads to a dominant effect of Eomes that acts as a weaker repressor of Pdcd1. Blocking PD-1 signaling in TEX s increases nuclear T-bet, restoring stronger repression of Pdcd1, and driving T-bet-associated gene expression programs of chemotaxis, homing, and activation. These data identify a mechanism whereby the T-bet-Eomes axis regulates exhaustion through their nuclear localization, providing insights into how these transcription factors regulate TEX biology., Competing Interests: Declaration of interests E.J.W. has consulting agreements with and/or is on the scientific advisory board for Merck, Elstar, Janssen, Related Sciences, Synthekine, and Surface Oncology. E.J.W. is a founder of Surface Oncology and Arsenal Biosciences. E.J.W. has a patent licensing agreement on the PD-1 pathway with Roche/Genentech., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
47. Distinct antibody and memory B cell responses in SARS-CoV-2 naïve and recovered individuals following mRNA vaccination.
- Author
-
Goel RR, Apostolidis SA, Painter MM, Mathew D, Pattekar A, Kuthuru O, Gouma S, Hicks P, Meng W, Rosenfeld AM, Dysinger S, Lundgreen KA, Kuri-Cervantes L, Adamski S, Hicks A, Korte S, Oldridge DA, Baxter AE, Giles JR, Weirick ME, McAllister CM, Dougherty J, Long S, D'Andrea K, Hamilton JT, Betts MR, Luning Prak ET, Bates P, Hensley SE, Greenplate AR, and Wherry EJ
- Subjects
- Adult, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Female, Humans, Male, Middle Aged, RNA, Messenger, Spike Glycoprotein, Coronavirus immunology, Vaccination, Young Adult, mRNA Vaccines, Antibodies, Viral immunology, B-Lymphocytes immunology, COVID-19 immunology, COVID-19 Vaccines, SARS-CoV-2 immunology, Vaccines, Synthetic
- Abstract
Novel mRNA vaccines for SARS-CoV-2 have been authorized for emergency use. Despite their efficacy in clinical trials, data on mRNA vaccine-induced immune responses are mostly limited to serological analyses. Here, we interrogated antibody and antigen-specific memory B cells over time in 33 SARS-CoV-2 naïve and 11 SARS-CoV-2 recovered subjects. SARS-CoV-2 naïve individuals required both vaccine doses for optimal increases in antibodies, particularly for neutralizing titers against the B.1.351 variant. Memory B cells specific for full-length spike protein and the spike receptor binding domain (RBD) were also efficiently primed by mRNA vaccination and detectable in all SARS-CoV-2 naive subjects after the second vaccine dose, though the memory B cell response declined slightly with age. In SARS-CoV-2 recovered individuals, antibody and memory B cell responses were significantly boosted after the first vaccine dose; however, there was no increase in circulating antibodies, neutralizing titers, or antigen-specific memory B cells after the second dose. This robust boosting after the first vaccine dose strongly correlated with levels of pre-existing memory B cells in recovered individuals, identifying a key role for memory B cells in mounting recall responses to SARS-CoV-2 antigens. Together, our data demonstrated robust serological and cellular priming by mRNA vaccines and revealed distinct responses based on prior SARS-CoV-2 exposure, whereby COVID-19 recovered subjects may only require a single vaccine dose to achieve peak antibody and memory B cell responses. These findings also highlight the utility of defining cellular responses in addition to serologies and may inform SARS-CoV-2 vaccine distribution in a resource-limited setting., (Copyright © 2021, American Association for the Advancement of Science.)
- Published
- 2021
- Full Text
- View/download PDF
48. Seasonal human coronavirus antibodies are boosted upon SARS-CoV-2 infection but not associated with protection.
- Author
-
Anderson EM, Goodwin EC, Verma A, Arevalo CP, Bolton MJ, Weirick ME, Gouma S, McAllister CM, Christensen SR, Weaver J, Hicks P, Manzoni TB, Oniyide O, Ramage H, Mathew D, Baxter AE, Oldridge DA, Greenplate AR, Wu JE, Alanio C, D'Andrea K, Kuthuru O, Dougherty J, Pattekar A, Kim J, Han N, Apostolidis SA, Huang AC, Vella LA, Kuri-Cervantes L, Pampena MB, Betts MR, Wherry EJ, Meyer NJ, Cherry S, Bates P, Rader DJ, and Hensley SE
- Subjects
- Adolescent, Adult, Animals, COVID-19 Serological Testing, Child, Child, Preschool, Chlorocebus aethiops, Cross Protection, Cross Reactions, Disease Susceptibility, HEK293 Cells, Humans, Infant, Infant, Newborn, Vero Cells, Alphacoronavirus immunology, Antibodies, Viral blood, Antibodies, Viral immunology, Betacoronavirus immunology, COVID-19 immunology
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread within the human population. Although SARS-CoV-2 is a novel coronavirus, most humans had been previously exposed to other antigenically distinct common seasonal human coronaviruses (hCoVs) before the coronavirus disease 2019 (COVID-19) pandemic. Here, we quantified levels of SARS-CoV-2-reactive antibodies and hCoV-reactive antibodies in serum samples collected from 431 humans before the COVID-19 pandemic. We then quantified pre-pandemic antibody levels in serum from a separate cohort of 251 individuals who became PCR-confirmed infected with SARS-CoV-2. Finally, we longitudinally measured hCoV and SARS-CoV-2 antibodies in the serum of hospitalized COVID-19 patients. Our studies indicate that most individuals possessed hCoV-reactive antibodies before the COVID-19 pandemic. We determined that ∼20% of these individuals possessed non-neutralizing antibodies that cross-reacted with SARS-CoV-2 spike and nucleocapsid proteins. These antibodies were not associated with protection against SARS-CoV-2 infections or hospitalizations, but they were boosted upon SARS-CoV-2 infection., Competing Interests: Declaration of interests A.C.H. is a consultant for Immunai. E.J.W. has consulting agreements with and/or is on the scientific advisory board for Merck, Elstar, Janssen, Related Sciences, Synthekine, and Surface Oncology. E.J.W. is a founder of Surface Oncology and Arsenal Biosciences. E.J.W. is an inventor on a patent (U.S. patent number 10,370,446) submitted by Emory University that covers the use of PD-1 blockade to treat infections and cancer. D.J.R. is on scientific advisory boards for Alnylam, Metrea, Novartis, Pfizer, and Verve; is a consultant for and receives research support from Regeneron for work unrelated to this report; and is a founder of Vascular Strategies and Staten Biotechnologies. S.E.H. has received consultancy fee from Sanofi Pasteur, Lumen, Novavax, and Merck for work unrelated to this report., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
49. Longitudinal Analysis Reveals Distinct Antibody and Memory B Cell Responses in SARS-CoV2 Naïve and Recovered Individuals Following mRNA Vaccination.
- Author
-
Goel RR, Apostolidis SA, Painter MM, Mathew D, Pattekar A, Kuthuru O, Gouma S, Kuri-Cervantes L, Meng W, Adamski S, Baxter AE, Giles JR, Weirick ME, McAllister CM, Hicks A, Korte S, Dougherty J, Long S, D'Andrea K, Hamilton JT, Prak ETL, Betts MR, Bates P, Hensley SE, Greenplate AR, and Wherry EJ
- Abstract
Novel mRNA vaccines for SARS-CoV2 have been authorized for emergency use and are currently being administered to millions of individuals worldwide. Despite their efficacy in clinical trials, there is limited data on vaccine-induced immune responses in individuals with a prior SARS-CoV2 infection compared to SARS-CoV2 naïve subjects. Moreover, how mRNA vaccines impact the development of antibodies as well as memory B cells in COVID-19 experienced versus COVID-19 naïve subjects remains poorly understood. In this study, we evaluated antibody responses and antigen-specific memory B cell responses over time in 33 SARS-CoV2 naïve and 11 SARS-CoV2 recovered subjects. mRNA vaccination induced significant antibody and memory B cell responses against full-length SARS-CoV2 spike protein and the spike receptor binding domain (RBD). SARS-CoV2 naïve individuals benefitted from both doses of mRNA vaccine with additional increases in antibodies and memory B cells following booster immunization. In contrast, SARS-CoV2 recovered individuals had a significant immune response after the first dose with no increase in circulating antibodies or antigen-specific memory B cells after the second dose. Moreover, the magnitude of the memory B cell response induced by vaccination was lower in older individuals, revealing an age-dependence to mRNA vaccine-induced B cell memory. Side effects also tended to associate with post-boost antibody levels, but not with post-boost memory B cells, suggesting that side effect severity may be a surrogate of short-term antibody responses. The frequency of pre-vaccine antigen-specific memory B cells in SARS-CoV2 recovered individuals strongly correlated with post-vaccine antibody levels, supporting a key role for memory B cells in humoral recall responses to SARS-CoV2. This observation may have relevance for future booster vaccines and for responses to viral variants that partially escape pre-existing antibodies and require new humoral responses to be generated from memory B cells. Finally, post-boost antibody levels were not correlated with post-boost memory responses in SARS-CoV2 naïve individuals, indicating that short-term antibody levels and memory B cells are complementary immunological endpoints that should be examined in tandem when evaluating vaccine response. Together, our data provide evidence of both serological response and immunological memory following mRNA vaccination that is distinct based on prior SARS-CoV2 exposure. These findings may inform vaccine distribution in a resource-limited setting., Competing Interests: COMPETING INTERESTS EJW is consulting or is an advisor for Merck, Elstar, Janssen, Related Sciences, Synthekine and Surface Oncology. EJW is a founder of Surface Oncology and Arsenal Biosciences. EJW is an inventor on a patent (US Patent number 10,370,446) submitted by Emory University that covers the use of PD-1 blockade to treat infections and cancer.
- Published
- 2021
- Full Text
- View/download PDF
50. Deep immune profiling of MIS-C demonstrates marked but transient immune activation compared to adult and pediatric COVID-19.
- Author
-
Vella LA, Giles JR, Baxter AE, Oldridge DA, Diorio C, Kuri-Cervantes L, Alanio C, Pampena MB, Wu JE, Chen Z, Huang YJ, Anderson EM, Gouma S, McNerney KO, Chase J, Burudpakdee C, Lee JH, Apostolidis SA, Huang AC, Mathew D, Kuthuru O, Goodwin EC, Weirick ME, Bolton MJ, Arevalo CP, Ramos A, Jasen CJ, Conrey PE, Sayed S, Giannini HM, D'Andrea K, Meyer NJ, Behrens EM, Bassiri H, Hensley SE, Henrickson SE, Teachey DT, Betts MR, and Wherry EJ
- Subjects
- Adolescent, Adult, Aging immunology, Child, Child, Preschool, Female, Flow Cytometry, Humans, Leukopenia immunology, Male, Young Adult, COVID-19 immunology, Lymphocyte Activation, Systemic Inflammatory Response Syndrome immunology, T-Lymphocytes immunology
- Abstract
Pediatric COVID-19 following SARS-CoV-2 infection is associated with fewer hospitalizations and often milder disease than in adults. A subset of children, however, present with Multisystem Inflammatory Syndrome in Children (MIS-C) that can lead to vascular complications and shock, but rarely death. The immune features of MIS-C compared to pediatric COVID-19 or adult disease remain poorly understood. We analyzed peripheral blood immune responses in hospitalized SARS-CoV-2 infected pediatric patients (pediatric COVID-19) and patients with MIS-C. MIS-C patients had patterns of T cell-biased lymphopenia and T cell activation similar to severely ill adults, and all patients with MIS-C had SARS-CoV-2 spike-specific antibodies at admission. A distinct feature of MIS-C patients was robust activation of vascular patrolling CX3CR1+ CD8+ T cells that correlated with the use of vasoactive medication. Finally, whereas pediatric COVID-19 patients with acute respiratory distress syndrome (ARDS) had sustained immune activation, MIS-C patients displayed clinical improvement over time, concomitant with decreasing immune activation. Thus, non-MIS-C versus MIS-C SARS-CoV-2 associated illnesses are characterized by divergent immune signatures that are temporally distinct from one another and implicate CD8+ T cells in the clinical presentation and trajectory of MIS-C., (Copyright © 2021, American Association for the Advancement of Science.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.