43 results on '"Kenneth B. Hoehn"'
Search Results
2. Tracking B cell responses to the SARS-CoV-2 mRNA-1273 vaccine
- Author
-
Felipe Lopes de Assis, Kenneth B. Hoehn, Xiaozhen Zhang, Lela Kardava, Connor D. Smith, Omar El Merhebi, Clarisa M. Buckner, Krittin Trihemasava, Wei Wang, Catherine A. Seamon, Vicky Chen, Paul Schaughency, Foo Cheung, Andrew J. Martins, Chi-I Chiang, Yuxing Li, John S. Tsang, Tae-Wook Chun, Steven H. Kleinstein, and Susan Moir
- Subjects
CP: Immunology ,Biology (General) ,QH301-705.5 - Abstract
Summary: Protective immunity following vaccination is sustained by long-lived antibody-secreting cells and resting memory B cells (MBCs). Responses to two-dose SARS-CoV-2 mRNA-1273 vaccination are evaluated longitudinally by multimodal single-cell analysis in three infection-naïve individuals. Integrated surface protein, transcriptomics, and B cell receptor (BCR) repertoire analysis of sorted plasmablasts and spike+ (S-2P+) and S-2P− B cells reveal clonal expansion and accumulating mutations among S-2P+ cells. These cells are enriched in a cluster of immunoglobulin G-expressing MBCs and evolve along a bifurcated trajectory rooted in CXCR3+ MBCs. One branch leads to CD11c+ atypical MBCs while the other develops from CD71+ activated precursors to resting MBCs, the dominant population at month 6. Among 12 evolving S-2P+ clones, several are populated with plasmablasts at early timepoints as well as CD71+ activated and resting MBCs at later timepoints, and display intra- and/or inter-cohort BCR convergence. These relationships suggest a coordinated and predictable evolution of SARS-CoV-2 vaccine-generated MBCs.
- Published
- 2023
- Full Text
- View/download PDF
3. Single-cell multi-omics reveals dyssynchrony of the innate and adaptive immune system in progressive COVID-19
- Author
-
Avraham Unterman, Tomokazu S. Sumida, Nima Nouri, Xiting Yan, Amy Y. Zhao, Victor Gasque, Jonas C. Schupp, Hiromitsu Asashima, Yunqing Liu, Carlos Cosme, Wenxuan Deng, Ming Chen, Micha Sam Brickman Raredon, Kenneth B. Hoehn, Guilin Wang, Zuoheng Wang, Giuseppe DeIuliis, Neal G. Ravindra, Ningshan Li, Christopher Castaldi, Patrick Wong, John Fournier, Santos Bermejo, Lokesh Sharma, Arnau Casanovas-Massana, Chantal B. F. Vogels, Anne L. Wyllie, Nathan D. Grubaugh, Anthony Melillo, Hailong Meng, Yan Stein, Maksym Minasyan, Subhasis Mohanty, William E. Ruff, Inessa Cohen, Khadir Raddassi, The Yale IMPACT Research Team, Laura E. Niklason, Albert I. Ko, Ruth R. Montgomery, Shelli F. Farhadian, Akiko Iwasaki, Albert C. Shaw, David van Dijk, Hongyu Zhao, Steven H. Kleinstein, David A. Hafler, Naftali Kaminski, and Charles S. Dela Cruz
- Subjects
Science - Abstract
SARS-CoV-2 infection can lead to progressive pathology in patients with COVID-19, but information for this disease progression is sparse. Here the authors use multi-omics approach to profile the immune responses of patients, assessing immune repertoire and effects of tocilizumab treatments, to find a dyssynchrony between innate and adaptive immunity in progressive COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
4. Phylogenetic analysis of migration, differentiation, and class switching in B cells
- Author
-
Kenneth B. Hoehn, Oliver G. Pybus, and Steven H. Kleinstein
- Subjects
Biology (General) ,QH301-705.5 - Abstract
B cells undergo rapid mutation and selection for antibody binding affinity when producing antibodies capable of neutralizing pathogens. This evolutionary process can be intermixed with migration between tissues, differentiation between cellular subsets, and switching between functional isotypes. B cell receptor (BCR) sequence data has the potential to elucidate important information about these processes. However, there is currently no robust, generalizable framework for making such inferences from BCR sequence data. To address this, we develop three parsimony-based summary statistics to characterize migration, differentiation, and isotype switching along B cell phylogenetic trees. We use simulations to demonstrate the effectiveness of this approach. We then use this framework to infer patterns of cellular differentiation and isotype switching from high throughput BCR sequence datasets obtained from patients in a study of HIV infection and a study of food allergy. These methods are implemented in the R package dowser, available at https://dowser.readthedocs.io. Author summary B cells produce high affinity antibodies through an evolutionary process of mutation and selection during adaptive immune responses. Migration between tissues, differentiation to cellular subtypes, and switching between different antibody isotypes can be important factors in shaping the role B cells play in response to infection, autoimmune disease, and allergies. B cell receptor (BCR) sequence data has the potential to elucidate important information about these processes. However, there is currently no robust, generalizable framework for making such inferences from BCR sequence data. Here, we develop three parsimony-based summary statistics to characterize migration, differentiation, and isotype switching along B cell phylogenetic trees. We confirm the effectiveness of our approach using simulations and further apply our method to data from patients with HIV and allergy. Our methods are released in the R package dowser: https://dowser.readthedocs.io.
- Published
- 2022
5. B Cell Mobilization, Dissemination, Fine Tuning of Local Antigen Specificity and Isotype Selection in Asthma
- Author
-
Line Ohm-Laursen, Hailong Meng, Kenneth B. Hoehn, Nima Nouri, Yue Jiang, Chris Clouser, Timothy G. Johnstone, Ron Hause, Balraj S. Sandhar, Nadine E. G. Upton, Elfy B. Chevretton, Raj Lakhani, Chris J. Corrigan, Steven H. Kleinstein, and Hannah J. Gould
- Subjects
asthma ,immunoglobulin ,allergy ,adaptive immunity ,IgD ,innate immunity ,Immunologic diseases. Allergy ,RC581-607 - Abstract
In order to better understand how the immune system interacts with environmental triggers to produce organ-specific disease, we here address the hypothesis that B and plasma cells are free to migrate through the mucosal surfaces of the upper and lower respiratory tracts, and that their total antibody repertoire is modified in a common respiratory tract disease, in this case atopic asthma. Using Adaptive Immune Receptor Repertoire sequencing (AIRR-seq) we have catalogued the antibody repertoires of B cell clones retrieved near contemporaneously from multiple sites in the upper and lower respiratory tract mucosa of adult volunteers with atopic asthma and non-atopic controls and traced their migration. We show that the lower and upper respiratory tracts are immunologically connected, with trafficking of B cells directionally biased from the upper to the lower respiratory tract and points of selection when migrating from the nasal mucosa and into the bronchial mucosa. The repertoires are characterized by both IgD-only B cells and others undergoing class switch recombination, with restriction of the antibody repertoire distinct in asthmatics compared with controls. We conclude that B cells and plasma cells migrate freely throughout the respiratory tract and exhibit distinct antibody repertoires in health and disease.
- Published
- 2021
- Full Text
- View/download PDF
6. Single-cell immunophenotyping of the skin lesion erythema migrans identifies IgM memory B cells
- Author
-
Ruoyi Jiang, Hailong Meng, Khadir Raddassi, Ira Fleming, Kenneth B. Hoehn, Kenneth R. Dardick, Alexia A. Belperron, Ruth R. Montgomery, Alex K. Shalek, David A. Hafler, Steven H. Kleinstein, and Linda K. Bockenstedt
- Subjects
Immunology ,Infectious disease ,Medicine - Abstract
The skin lesion erythema migrans (EM) is an initial sign of the Ixodes tick–transmitted Borreliella spirochetal infection known as Lyme disease. T cells and innate immune cells have previously been shown to predominate the EM lesion and promote the reaction. Despite the established importance of B cells and antibodies in preventing infection, the role of B cells in the skin immune response to Borreliella is unknown. Here, we used single-cell RNA-Seq in conjunction with B cell receptor (BCR) sequencing to immunophenotype EM lesions and their associated B cells and BCR repertoires. We found that B cells were more abundant in EM in comparison with autologous uninvolved skin; many were clonally expanded and had circulating relatives. EM-associated B cells upregulated the expression of MHC class II genes and exhibited preferential IgM isotype usage. A subset also exhibited low levels of somatic hypermutation despite a gene expression profile consistent with memory B cells. Our study demonstrates that single-cell gene expression with paired BCR sequencing can be used to interrogate the sparse B cell populations in human skin and reveals that B cells in the skin infection site in early Lyme disease expressed a phenotype consistent with local antigen presentation and antibody production.
- Published
- 2021
- Full Text
- View/download PDF
7. Age-associated B cells are heterogeneous and dynamic drivers of autoimmunity in mice
- Author
-
Kevin M. Nickerson, Shuchi Smita, Kenneth B. Hoehn, Anthony D. Marinov, Kayla B. Thomas, Justin T. Kos, Yi Yang, Sheldon I. Bastacky, Corey T. Watson, Steven H. Kleinstein, and Mark J. Shlomchik
- Subjects
Immunology ,Immunology and Allergy - Abstract
Age-associated B cells (ABCs) are formed under inflammatory conditions and are considered a type of memory B cell (MBC) expressing the transcription factor T-bet. In SLE, ABC frequency is correlated with disease, and they are thought to be the source of autoantibody-secreting cells. However, in inflammatory conditions, whether autoreactive B cells can become resting MBCs is uncertain. Further, the phenotypic identity of ABCs and their relationship to other B cell subsets, such as plasmablasts, is unclear. Whether ABCs directly promote disease is untested. Here we report, in the MRL/lpr SLE model, unexpected heterogeneity among ABC-like cells for expression of the integrins CD11b and CD11c, T-bet, and memory or plasmablast markers. Transfer and labeling studies demonstrated that ABCs are dynamic, rapidly turning over. scRNA-seq identified B cell clones present in multiple subsets, revealing that ABCs can be plasmablast precursors or undergo cycles of reactivation. Deletion of CD11c-expressing B cells revealed a direct role for ABC-like B cells in lupus pathogenesis.
- Published
- 2023
8. The memory of pathogenic IgE is contained within CD23 (+) IgG1 (+) memory B cells poised to switch to IgE in food allergy
- Author
-
Miyo Ota, Kenneth B. Hoehn, Takayuki Ota, Carlos J. Aranda, Sara Friedman, Weslley F. Braga, Alefiyah Malbari, Steven H. Kleinstein, Scott H. Sicherer, and Maria A. Curotto de Lafaille
- Subjects
Article - Abstract
Food allergy is caused by allergen-specific IgE antibodies but little is known about the B cell memory of persistent IgE responses. Here we describe in human pediatric peanut allergy CD23+IgG1+memory B cells arising in type 2 responses that contain peanut specific clones and generate IgE cells on activation. These ‘type2-marked’ IgG1+memory B cells differentially express IL-4/IL-13 regulated genesFCER2/CD23, IL4R, and germlineIGHEand carry highly mutated B cell receptors (BCRs). Further, high affinity memory B cells specific for the main peanut allergen Ara h 2 mapped to the population of ‘type2-marked’ IgG1+memory B cells and included convergent BCRs across different individuals. Our findings indicate that CD23+IgG1+memory B cells transcribing germlineIGHEare a unique memory population containing precursors of pathogenic IgE.One-Sentence SummaryWe describe a unique population of IgG+memory B cells poised to switch to IgE that contains high affinity allergen-specific clones in peanut allergy.
- Published
- 2023
9. BTG1 mutation yields supercompetitive B cells primed for malignant transformation
- Author
-
Coraline Mlynarczyk, Matt Teater, Juhee Pae, Christopher R. Chin, Ling Wang, Theinmozhi Arulraj, Darko Barisic, Antonin Papin, Kenneth B. Hoehn, Ekaterina Kots, Jonatan Ersching, Arnab Bandyopadhyay, Ersilia Barin, Hui Xian Poh, Chiara M. Evans, Amy Chadburn, Zhengming Chen, Hao Shen, Hannah M. Isles, Benedikt Pelzer, Ioanna Tsialta, Ashley S. Doane, Huimin Geng, Muhammad Hassan Rehman, Jonah Melnick, Wyatt Morgan, Diu T. T. Nguyen, Olivier Elemento, Michael G. Kharas, Samie R. Jaffrey, David W. Scott, George Khelashvili, Michael Meyer-Hermann, Gabriel D. Victora, and Ari Melnick
- Subjects
Multidisciplinary - Abstract
Multicellular life requires altruistic cooperation between cells. The adaptive immune system is a notable exception, wherein germinal center B cells compete vigorously for limiting positive selection signals. Studying primary human lymphomas and developing new mouse models, we found that mutations affecting BTG1 disrupt a critical immune gatekeeper mechanism that strictly limits B cell fitness during antibody affinity maturation. This mechanism converted germinal center B cells into supercompetitors that rapidly outstrip their normal counterparts. This effect was conferred by a small shift in MYC protein induction kinetics but resulted in aggressive invasive lymphomas, which in humans are linked to dire clinical outcomes. Our findings reveal a delicate evolutionary trade-off between natural selection of B cells to provide immunity and potentially dangerous features that recall the more competitive nature of unicellular organisms.
- Published
- 2023
10. Adaptive immune responses to SARS-CoV-2 persist in the pharyngeal lymphoid tissue of children
- Author
-
Qin Xu, Pedro Milanez-Almeida, Andrew J. Martins, Andrea J. Radtke, Kenneth B. Hoehn, Cihan Oguz, Jinguo Chen, Can Liu, Juanjie Tang, Gabrielle Grubbs, Sydney Stein, Sabrina Ramelli, Juraj Kabat, Hengameh Behzadpour, Maria Karkanitsa, Jacquelyn Spathies, Heather Kalish, Lela Kardava, Martha Kirby, Foo Cheung, Silvia Preite, Patrick C. Duncker, Moses M. Kitakule, Nahir Romero, Diego Preciado, Lyuba Gitman, Galina Koroleva, Grace Smith, Arthur Shaffer, Ian T. McBain, Peter J. McGuire, Stefania Pittaluga, Ronald N. Germain, Richard Apps, Daniella M. Schwartz, Kaitlyn Sadtler, Susan Moir, Daniel S. Chertow, Steven H. Kleinstein, Surender Khurana, John S. Tsang, Pamela Mudd, Pamela L. Schwartzberg, and Kalpana Manthiram
- Subjects
SARS-CoV-2 ,Immunology ,Palatine Tonsil ,Immunology and Allergy ,Humans ,COVID-19 ,Adaptive Immunity ,Child ,Antibodies, Viral ,Pandemics - Abstract
Most studies of adaptive immunity to SARS-CoV-2 infection focus on peripheral blood, which may not fully reflect immune responses at the site of infection. Using samples from 110 children undergoing tonsillectomy and adenoidectomy during the COVID-19 pandemic, we identified 24 samples with evidence of previous SARS-CoV-2 infection, including neutralizing antibodies in serum and SARS-CoV-2-specific germinal center and memory B cells in the tonsils and adenoids. Single-cell B cell receptor (BCR) sequencing indicated virus-specific BCRs were class-switched and somatically hypermutated, with overlapping clones in the two tissues. Expanded T cell clonotypes were found in tonsils, adenoids and blood post-COVID-19, some with CDR3 sequences identical to previously reported SARS-CoV-2-reactive T cell receptors (TCRs). Pharyngeal tissues from COVID-19-convalescent children showed persistent expansion of germinal center and antiviral lymphocyte populations associated with interferon (IFN)-γ-type responses, particularly in the adenoids, and viral RNA in both tissues. Our results provide evidence for persistent tissue-specific immunity to SARS-CoV-2 in the upper respiratory tract of children after infection.
- Published
- 2022
11. PD-1highCXCR5–CD4+ peripheral helper T cells promote CXCR3+ plasmablasts in human acute viral infection
- Author
-
Hiromitsu Asashima, Subhasis Mohanty, Michela Comi, William E. Ruff, Kenneth B. Hoehn, Patrick Wong, Jon Klein, Carolina Lucas, Inessa Cohen, Sarah Coffey, Nikhil Lele, Leissa Greta, Khadir Raddassi, Omkar Chaudhary, Avraham Unterman, Brinda Emu, Steven H. Kleinstein, Ruth R. Montgomery, Akiko Iwasaki, Charles S. Dela Cruz, Naftali Kaminski, Albert C. Shaw, David A. Hafler, and Tomokazu S. Sumida
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
12. PD-1
- Author
-
Hiromitsu, Asashima, Subhasis, Mohanty, Michela, Comi, William E, Ruff, Kenneth B, Hoehn, Patrick, Wong, Jon, Klein, Carolina, Lucas, Inessa, Cohen, Sarah, Coffey, Nikhil, Lele, Leissa, Greta, Khadir, Raddassi, Omkar, Chaudhary, Avraham, Unterman, Brinda, Emu, Steven H, Kleinstein, Ruth R, Montgomery, Akiko, Iwasaki, Charles S, Dela Cruz, Naftali, Kaminski, Albert C, Shaw, David A, Hafler, and Tomokazu S, Sumida
- Abstract
T cell-B cell interaction is the key immune response to protect the host from severe viral infection. However, how T cells support B cells to exert protective humoral immunity in humans is not well understood. Here, we use COVID-19 as a model of acute viral infections and analyze CD4
- Published
- 2021
13. Human B cell lineages associated with germinal centers following influenza vaccination are measurably evolving
- Author
-
Frederick I. Miller, Ali H. Ellebedy, Ruoyi Jiang, Jackson S. Turner, Steven H. Kleinstein, Oliver G. Pybus, and Kenneth B. Hoehn
- Subjects
QH301-705.5 ,Science ,temporal evolution ,Somatic hypermutation ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Evolution, Molecular ,Affinity maturation ,03 medical and health sciences ,Immunology and Inflammation ,0302 clinical medicine ,Immunity ,Influenza, Human ,medicine ,Humans ,Biology (General) ,Phylogeny ,B cell ,030304 developmental biology ,B-Lymphocytes ,0303 health sciences ,General Immunology and Microbiology ,biology ,B cell receptor ,General Neuroscience ,Vaccination ,Germinal center ,General Medicine ,measurable evolution ,Germinal Center ,Virology ,3. Good health ,somatic hypermutation ,phylogenetics ,medicine.anatomical_structure ,Influenza Vaccines ,biology.protein ,Medicine ,Antibody ,Research Article ,Computational and Systems Biology ,Human ,030215 immunology - Abstract
The poor efficacy of seasonal influenza virus vaccines is often attributed to pre-existing immunity interfering with the persistence and maturation of vaccine-induced B cell responses. We previously showed that a subset of vaccine-induced B cell lineages are recruited into germinal centers (GCs) following vaccination, suggesting that affinity maturation of these lineages against vaccine antigens can occur. However, it remains to be determined whether seasonal influenza vaccination stimulates additional evolution of vaccine-specific lineages, and previous work has found no significant increase in somatic hypermutation among influenza-binding lineages sampled from the blood following seasonal vaccination in humans. Here, we investigate this issue using a phylogenetic test of measurable immunoglobulin sequence evolution. We first validate this test through simulations and survey measurable evolution across multiple conditions. We find significant heterogeneity in measurable B cell evolution across conditions, with enrichment in primary response conditions such as HIV infection and early childhood development. We then show that measurable evolution following influenza vaccination is highly compartmentalized: while lineages in the blood are rarely measurably evolving following influenza vaccination, lineages containing GC B cells are frequently measurably evolving. Many of these lineages appear to derive from memory B cells. We conclude from these findings that seasonal influenza virus vaccination can stimulate additional evolution of responding B cell lineages, and imply that the poor efficacy of seasonal influenza vaccination is not due to a complete inhibition of vaccine-specific B cell evolution., eLife digest When the immune system encounters a disease-causing pathogen, it releases antibodies that can bind to specific regions of the bacterium or virus and help to clear the infection. These proteins are generated by B cells which, upon detecting the pathogen, can begin to mutate and alter the structure of the antibody they produce: the better the antibody is at binding to the pathogen, the more likely the B cell is to survive. This process of evolution produces B cells that make more effective antibodies. After the infection, some of these cells become ‘memory B cells’ which can be stimulated in to action when the pathogen invades again. Many vaccines also depend on this process to trigger the production of memory B cells that can fight off a specific disease-causing agent. However, it is unclear to what extent memory B cells that already exist are able to continue to evolve and modify their antibodies. This is particularly important for the flu vaccine, as the virus that causes influenza rapidly mutates. To provide high levels of protection, the memory B cells formed following the vaccine may therefore need to evolve to make different antibodies that recognize mutated forms of the virus. It is thought that the low effectiveness of the flu vaccine is partially because the response it triggers does not stimulate additional evolution of memory B cells. To test this theory, Hoehn et al. developed a computational method that can detect the evolution of B cells over time. The tool was applied to samples collected from the blood and lymph nodes (organ where immune cells reside) of people who recently received the flu vaccine. The results were then compared to B cells taken from people after different infections, vaccinations, and other conditions. Hoehn et al. found the degree to which B cells evolve varies significantly between conditions. For example, B cells produced during chronic HIV infections frequently evolved over time, while such evolution was rarely observed during the autoimmune disease myasthenia gravis. The analysis also showed that memory B cells produced by the flu vaccine were able to evolve if recruited to the lymph nodes, but this was rarely detected in B cells in the blood. These findings suggest the low efficacy of the flu vaccine is not due to a complete lack of B cell evolution, but likely due to other factors. For instance, it is possible the evolutionary process it stimulates is not as robust as in other conditions, or is less likely to produce long-lived B cells that release antibodies. More research is needed to explore these ideas and could lead to the development of more effective flu vaccines.
- Published
- 2021
14. Author response: Human B cell lineages associated with germinal centers following influenza vaccination are measurably evolving
- Author
-
Kenneth B Hoehn, Jackson S Turner, Frederick I Miller, Ruoyi Jiang, Oliver G Pybus, Ali H Ellebedy, and Steven H Kleinstein
- Published
- 2021
15. Elevated N-linked glycosylation of IgG variable regions in myasthenia gravis disease subtypes
- Author
-
Sara E. Vazquez, Neil L. Kelleher, Minh C. Pham, Michael R. Wilson, Miriam L. Fichtner, Kenneth B. Hoehn, Caleigh Mandel-Brehm, Valerie J. Winton, Steven H. Kleinstein, Ruoyi Jiang, Joseph L. DeRisi, Kevin C. O’Connor, and Richard Nowak
- Subjects
Adult ,Male ,Glycosylation ,Immunology ,Immunoglobulin Variable Region ,Somatic hypermutation ,Receptors, Antigen, B-Cell ,Immune receptor ,Biology ,medicine.disease_cause ,Autoimmune Disease ,Article ,Autoimmunity ,chemistry.chemical_compound ,Young Adult ,Rare Diseases ,N-linked glycosylation ,Clinical Research ,Receptors ,Myasthenia Gravis ,medicine ,Immunology and Allergy ,Humans ,Aged ,Autoantibodies ,Autoimmune disease ,B-Lymphocytes ,B-Cell ,Neurosciences ,Autoantibody ,Middle Aged ,medicine.disease ,Molecular biology ,Myasthenia gravis ,Phenotype ,chemistry ,Antigen ,Immunoglobulin G ,Female - Abstract
Elevated N-linked glycosylation of immunoglobulin G variable regions (IgG-V(N-Glyc)) is an emerging molecular phenotype associated with autoimmune disorders. To test the broader specificity of elevated IgG-V(N-Glyc), we studied patients with distinct subtypes of myasthenia gravis (MG), a B cell-mediated autoimmune disease. Our experimental design focused on examining the B cell repertoire and total IgG. It specifically included adaptive immune receptor repertoire sequencing to quantify and characterize N-linked glycosylation sites in the circulating B cell receptor repertoire, proteomics to examine glycosylation patterns of the total circulating IgG, and an exploration of human-derived recombinant autoantibodies, which were studied with mass spectrometry and antigen binding assays to respectively confirm occupation of glycosylation sites and determine whether they alter binding. We found that the frequency of IgG-V(N-Glyc) motifs was increased in the total B cell receptor repertoire of MG patients when compared to healthy donors. The elevated frequency was attributed to both biased V gene segment usage and somatic hypermutation. IgG-V(N-Glyc) could be observed in the total circulating IgG in a subset of MG patients. Autoantigen binding, by four patient-derived MG autoantigen-specific monoclonal antibodies with experimentally confirmed presence of IgG-V(N-Glyc), was not altered by the glycosylation. Our findings extend prior work on patterns of immunoglobulin variable region N-linked glycosylation in autoimmunity to MG subtypes.
- Published
- 2021
16. The Physiological Landscape and Specificity of Antibody Repertoires
- Author
-
Lucia Csepregi, Kenneth B. Hoehn, Bruno E. Correia, Simon Friedensohn, Sai T. Reddy, Cédric R. Weber, Arkadij Kummer, Fabian Sesterhenn, Joseph M. Taft, and Daniel Neumeier
- Subjects
0303 health sciences ,biology ,Repertoire ,Spleen ,Germline ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Lymphatic system ,medicine.anatomical_structure ,Antibody Repertoire ,Antigen ,030220 oncology & carcinogenesis ,Humoral immunity ,Immunology ,biology.protein ,medicine ,Antibody ,030304 developmental biology - Abstract
Diverse antibody repertoires spanning multiple lymphoid organs (e.g., bone marrow, spleen, lymph nodes) form the foundation of protective humoral immunity. Changes in their composition across lymphoid organs are a consequence of B-cell selection and migration events leading to a highly dynamic and unique physiological landscape of antibody repertoires upon antigenic challenge (e.g., vaccination). However, to what extent B cells encoding identical or similar antibody sequences (clones) are distributed across multiple lymphoid organs and how this is shaped by the strength of a humoral response, remains largely unexplored. Here, we performed an in-depth systems analysis of antibody repertoires across multiple distinct lymphoid organs of immunized mice, and discovered that organ-specific antibody repertoire features (e.g., germline V-gene usage and clonal expansion profiles) equilibrated upon a strong humoral response (multiple immunizations and high serum titers). This resulted in a surprisingly high degree of repertoire consolidation, characterized by highly connected and overlapping B-cell clones across multiple lymphoid organs. Finally, we revealed distinct physiological axes indicating clonal migrations and showed that antibody repertoire consolidation directly correlated with antigen-specificity. Our study uncovered how a strong humoral response resulted in a more uniform but redundant physiological landscape of antibody repertoires, indicating that increases in antibody serum titers were a result of synergistic contributions from antigen-specific B-cell clones distributed across multiple lymphoid organs. Our findings provide valuable insights for the assessment and design of vaccine strategies.
- Published
- 2021
17. Abstract A24: BTG1 mutations confer a fitness advantage and promote aggressive B cell lymphoma development by lowering the threshold for MYC induction
- Author
-
Coraline Mlynarczyk, Matt Teater, Juhee Pae, Ling Wang, Jonatan Ersching, Antonin Papin, Darko Barisic, Ersilia Barin, Kenneth B. Hoehn, Zhengming Chen, Diu T. T. Nguyen, Chiara Evans, Ashley S. Doane, Michael G. Kharas, David W. Scott, Gabriel Victora, and Ari Melnick
- Subjects
General Medicine - Abstract
BTG1 somatic mutations are exclusively found in mature B cell malignancies, ~12% diffuse large B cell lymphoma (DLBCL) and are particularly enriched in the MCD/cluster 5 subtype of ABC-DLBCL, characterized by extranodal dissemination and poor clinical outcome. However, the mechanism of action and clinical relevance of BTG1 mutations remain unknown. We find that BTG1 mutations score among the top mutations with DLBCL driver potential, using a rigorous genomic and epigenomic covariates analysis. Most notably, BTG1 mutant patients presented with inferior clinical outcome (p=0.0011) in ABC-DLBCL cases from 5 cohorts and BTG1 mutation was independently associated with lower overall survival in a multivariable Cox regression analysis (p=0.0190) DLBCL originates from mature B cells having experienced the germinal center (GC) reaction. We therefore generated a conditional knockin mouse model to express the most frequent Btg1 Q36H in B cells. Btg1 Q36H GC B cells showed a dramatic fitness advantage in in vivo competitive assays. This effect was specific to the GC compartment and was dependent on T cells. RNAseq showed that Btg1Q36H GC B cells were markedly enriched for genes normally transiently induced upon positive selection by T cells, including MYC targets. The same signatures were enriched in BTG1 mutant DLBCL patients and isogenic BTG1Q36H vs BTG1WT human DLBCL cell lines. We observed a higher proportion of MYC-expressing cells in Btg1Q36H GCs without an increase in maximal MYC levels per cell, also confirmed in human DLBCL lines and primary human tonsillar B cells, suggesting a lowered threshold for MYC induction in mutant cells. Mechanistically, ~800 transcripts associated with BTG1WT, but not BTG1Q36H. These belong to the same gene sets that characterize positively selected GC B cell, including MYC itself. BTG1Q36H therefore showed reduced association with the MYC mRNA. We found that this associated with facilitated MYC protein synthesis. Polysome profiling also showed higher translation capacity in BTG1Q36H DLBCL cells. Crossing our Btg1 Q36H mice to the VavP-Bcl2 model showed that VavP-Bcl2+Btg1Q36H mice displayed shorter survival, earlier onset of lymphoma and dysplastic B cell infiltration into non lymphoid organs. Immunoglobulin genes sequencing showed that VavP-Bcl2+Btg1Q36H lymphoma B cells were highly clonal, extensively mutated and selected. Importantly, the lack of BTG1 deletions suggested that BTG1 missense mutations do not cause a full loss-of-function of the protein. Indeed, we observed that shRNA-mediated knockdown of BTG1 resulted in apoptosis. Collectively, BTG1 mutations contribute to the formation of aggressive lymphomas through an entirely novel mechanism, by lowering the threshold to MYC induction in response to T cell selection signals, conferring dramatic fitness. This effect appears to correspond to a partial loss of function disrupting a novel GC context-specific check point, whereby BTG1 normally attenuates spurious MYC translation to tightly restrict fitness potential. Citation Format: Coraline Mlynarczyk, Matt Teater, Juhee Pae, Ling Wang, Jonatan Ersching, Antonin Papin, Darko Barisic, Ersilia Barin, Kenneth B. Hoehn, Zhengming Chen, Diu T. T. Nguyen, Chiara Evans, Ashley S. Doane, Michael G. Kharas, David W. Scott, Gabriel Victora, Ari Melnick. BTG1 mutations confer a fitness advantage and promote aggressive B cell lymphoma development by lowering the threshold for MYC induction [abstract]. In: Proceedings of the Third AACR International Meeting: Advances in Malignant Lymphoma: Maximizing the Basic-Translational Interface for Clinical Application; 2022 Jun 23-26; Boston, MA. Philadelphia (PA): AACR; Blood Cancer Discov 2022;3(5_Suppl):Abstract nr A24.
- Published
- 2022
18. Phylogenetic analysis of migration, differentiation, and class switching in B cells
- Author
-
Kenneth B. Hoehn, Steven H. Kleinstein, and Oliver G. Pybus
- Subjects
Cellular differentiation ,B-cell receptor ,Receptors, Antigen, B-Cell ,HIV Infections ,Computational biology ,medicine.disease_cause ,Cellular and Molecular Neuroscience ,Genetics ,medicine ,Humans ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,B cell ,Phylogeny ,Mutation ,B-Lymphocytes ,Ecology ,biology ,Phylogenetic tree ,breakpoint cluster region ,Cell Differentiation ,Immunoglobulin Class Switching ,medicine.anatomical_structure ,Computational Theory and Mathematics ,Immunoglobulin class switching ,Modeling and Simulation ,biology.protein ,Antibody - Abstract
B cells undergo rapid mutation and selection for antibody binding affinity when producing antibodies capable of neutralizing pathogens. This evolutionary process can be intermixed with migration between tissues, differentiation between cellular subsets, and switching between functional isotypes. B cell receptor (BCR) sequence data has the potential to elucidate important information about these processes. However, there is currently no robust, generalizable framework for making such inferences from BCR sequence data. To address this, we develop three parsimony-based summary statistics to characterize migration, differentiation, and isotype switching along B cell phylogenetic trees. We use simulations to demonstrate the effectiveness of this approach. We then use this framework to infer patterns of cellular differentiation and isotype switching from high throughput BCR sequence datasets obtained from patients in a study of HIV infection and a study of food allergy. These methods are implemented in the R packagedowser, available athttps://bitbucket.org/kleinstein/dowser.Author summaryB cells produce high affinity antibodies through an evolutionary process of mutation and selection during adaptive immune responses. Migration between tissues, differentiation to cellular subtypes, and switching between different antibody isotypes can be important factors in shaping the role B cells play in response to infection, autoimmune disease, and allergies. B cell receptor (BCR) sequence data has the potential to elucidate important information about these processes. However, there is currently no robust, generalizable framework for making such inferences from BCR sequence data. Here, we develop three parsimony-based summary statistics to characterize migration, differentiation, and isotype switching along B cell phylogenetic trees. Using simulations, we confirm the effectiveness of our approach, as well as identify some caveats. We further use these summary statistics to investigate patterns of cellular differentiation in three HIV patients, and patterns of isotype switching in an individual with food allergies. Our methods are released in the R packagedowser:https://bitbucket.org/kleinstein/dowser.
- Published
- 2021
19. Repertoire-wide phylogenetic models of B cell molecular evolution reveal evolutionary signatures of aging and vaccination
- Author
-
Gerton Lunter, Steven H. Kleinstein, Jason A. Vander Heiden, Julian Q. Zhou, Oliver G. Pybus, and Kenneth B. Hoehn
- Subjects
0301 basic medicine ,Aging ,Evolution ,Somatic hypermutation ,Biology ,Affinity maturation ,Evolution, Molecular ,03 medical and health sciences ,Negative selection ,0302 clinical medicine ,Molecular evolution ,Phylogenetics ,antibody ,Humans ,Selection (genetic algorithm) ,Phylogeny ,030304 developmental biology ,0303 health sciences ,B-Lymphocytes ,Multidisciplinary ,Natural selection ,B cell repertoire ,Phylogenetic tree ,Repertoire ,Vaccination ,Biological Sciences ,BCR ,somatic hypermutation ,phylogenetics ,030104 developmental biology ,PNAS Plus ,Evolutionary biology ,Mutation ,030217 neurology & neurosurgery ,030215 immunology - Abstract
Significance High-affinity antibodies that protect us from infection are produced by B cells through an evolutionary process of mutation and selection during adaptive immune responses. B cell repertoire sequencing combined with phylogenetic methods has provided unprecedented potential to study B cells as an evolving population. However, phylogenetic models operate on individual lineages rather than the thousands of lineages often found in B cell repertoires. Here, we develop an evolutionary framework that incorporates B cell-specific features and combines information across lineages to characterize mutation and selection dynamics of entire repertoires. We use this technique to demonstrate evidence of age-associated changes in somatic hypermutation targeting and uncover a general trend within our datasets toward negative selection over the course of affinity maturation., In order to produce effective antibodies, B cells undergo rapid somatic hypermutation (SHM) and selection for binding affinity to antigen via a process called affinity maturation. The similarities between this process and evolution by natural selection have led many groups to use phylogenetic methods to characterize the development of immunological memory, vaccination, and other processes that depend on affinity maturation. However, these applications are limited by the fact that most phylogenetic models are designed to be applied to individual lineages comprising genetically diverse sequences, while B cell repertoires often consist of hundreds to thousands of separate low-diversity lineages. Further, several features of affinity maturation violate important assumptions in standard phylogenetic models. Here, we introduce a hierarchical phylogenetic framework that integrates information from all lineages in a repertoire to more precisely estimate model parameters while simultaneously incorporating the unique features of SHM. We demonstrate the power of this repertoire-wide approach by characterizing previously undescribed phenomena in affinity maturation. First, we find evidence consistent with age-related changes in SHM hot-spot targeting. Second, we identify a consistent relationship between increased tree length and signs of increased negative selection, apparent in the repertoires of recently vaccinated subjects and those without any known recent infections or vaccinations. This suggests that B cell lineages shift toward negative selection over time as a general feature of affinity maturation. Our study provides a framework for undertaking repertoire-wide phylogenetic testing of SHM hypotheses and provides a means of characterizing dynamics of mutation and selection during affinity maturation.
- Published
- 2019
20. Immune dysregulation and autoreactivity correlate with disease severity in SARS-CoV-2-associated multisystem inflammatory syndrome in children
- Author
-
Andrew J. Rice, Alamzeb Khan, Ningshan Li, Charles S. Dela Cruz, Xiting Yan, Kenneth B. Hoehn, Hiromitsu Asashima, Victoria Habet, Tomokazu Sumida, Jason Bishai, Merrick Lopez, Carrie L. Lucas, Jason Catanzaro, Brian Sellers, John S. Tsang, Pamela A. Guerrerio, David van Dijk, Michela Comi, Richard W. Pierce, Anjali Ramaswamy, Harsha K. Chandnani, Zuoheng Wang, Aagam Shah, Avraham Unterman, Yunqing Liu, David A. Hafler, Steven H. Kleinstein, Nina N. Brodsky, William W. Lau, Naftali Kaminski, Neha Bansal, and Neal G. Ravindra
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Myeloid ,Adolescent ,Plasma Cells ,Immunology ,Receptors, Antigen, T-Cell ,Inflammation ,MIS-C ,plasmablasts ,CD8-Positive T-Lymphocytes ,Biology ,medicine.disease_cause ,Severity of Illness Index ,Asymptomatic ,Article ,Autoimmunity ,03 medical and health sciences ,0302 clinical medicine ,Severity of illness ,medicine ,otorhinolaryngologic diseases ,Humans ,Immunology and Allergy ,Myeloid Cells ,Endothelium ,Child ,Autoantibodies ,SARS-CoV-2 ,alarmins ,TRBV11-2 ,T-cell receptor ,COVID-19 ,Immune dysregulation ,Systemic Inflammatory Response Syndrome ,Killer Cells, Natural ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,pediatric ,inflammation ,Child, Preschool ,030220 oncology & carcinogenesis ,cytotoxicity ,medicine.symptom ,CD8 - Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a life-threatening post-infectious complication occurring unpredictably weeks after mild or asymptomatic SARS-CoV-2 infection. We profiled MIS-C, adult COVID-19, and healthy pediatric and adult individuals using single-cell RNA sequencing, flow cytometry, antigen receptor repertoire analysis, and unbiased serum proteomics, which collectively identified a signature in MIS-C patients that correlated with disease severity. Despite having no evidence of active infection, MIS-C patients had elevated S100A-family alarmins and decreased antigen presentation signatures, indicative of myeloid dysfunction. MIS-C patients showed elevated expression of cytotoxicity genes in NK and CD8+ T cells and expansion of specific IgG-expressing plasmablasts. Clinically severe MIS-C patients displayed skewed memory T cell TCR repertoires and autoimmunity characterized by endothelium-reactive IgG. The alarmin, cytotoxicity, TCR repertoire, and plasmablast signatures we defined have potential for application in the clinic to better diagnose and potentially predict disease severity early in the course of MIS-C., Graphical abstract, Multisystem inflammatory syndrome in children (MIS-C) is a life-threatening and unpredictable condition of unknown etiology. Ramaswamy et al. use peripheral blood single-cell transcriptomic profiling along with other techniques to define key innate and adaptive signatures that characterize MIS-C.
- Published
- 2021
21. PD-1highCXCR5–CD4+ Peripheral Helper T (Tph) cells Promote Tissue-Homing Plasmablasts in COVID-19
- Author
-
William Ruff, Brinda Emu, Ruth R. Montgomery, Tomokazu Sumida, Albert C. Shaw, Akiko Iwasaki, Inessa Cohen, Charles S. Dela Cruz, Subhasis Mohanty, David A. Hafler, Avraham Unterman, Omkar Chaudhary, Kenneth B. Hoehn, Sarah Coffey, Michela Comi, Naftali Kaminski, Khadir Raddassi, Steven H. Kleinstein, Hiromitsu Asashima, and Patrick Wong
- Subjects
biology ,Chemistry ,CXCR3 ,Chemokine receptor ,medicine.anatomical_structure ,Immune system ,Immunity ,Immunology ,biology.protein ,medicine ,Antibody ,Receptor ,B cell ,Homing (hematopoietic) - Abstract
SummaryA dysregulated immune response against coronavirus-2 (SARS-CoV-2) plays a critical role in the outcome of patients with coronavirus disease 2019 (COVID-19). A significant increase in circulating plasmablasts is characteristic of COVID-19 though the underlying mechanisms and its prognostic implications are not known. Here, we demonstrate that in the acute phase of COVID-19, activated PD-1highCXCR5−CD4+ T cells, peripheral helper T cells, (Tph) are significantly increased and promote inflammatory tissue-homing plasmablasts in patients with stable but not severe COVID-19. Analysis of scRNA-seq data revealed that plasmablasts in stable patients express higher levels of tissue-homing receptors including CXCR3. The increased Tph cells exhibited “B cell help” signatures similar to that of circulating T follicular helper (cTfh) cells and promoted B cell differentiation in vitro. Compared with cTfh cells, Tph cells produced more IFNγ, inducing tissue-homing chemokine receptors on plasmablasts. Finally, expansion of activated Tph cells was correlated with the frequency of CXCR3+ plasmablasts in the acute phase of patients with stable disease. Our results demonstrate a novel role for Tph cells in acute viral immunity by inducing ectopic, antibody secreting plasmablasts.
- Published
- 2021
22. Elevated N-glycosylation of immunoglobulin G variable regions in myasthenia gravis highlights a commonality across autoantibody-associated diseases
- Author
-
Neil L. Kelleher, Steven H. Kleinstein, Caleigh Mandel-Brehm, Miriam L. Fichtner, Minh C. Pham, Joseph L. DeRisi, Kevin C. O’Connor, Valerie J. Winton, Kenneth B. Hoehn, Richard Nowak, Michael R. Wilson, Sara E. Vazquez, and Roy Jiang
- Subjects
Autoimmune disease ,Glycosylation ,medicine.drug_class ,Autoantibody ,Somatic hypermutation ,Biology ,Monoclonal antibody ,medicine.disease ,medicine.disease_cause ,Immunoglobulin G ,Autoimmunity ,chemistry.chemical_compound ,chemistry ,N-linked glycosylation ,Immunology ,medicine ,biology.protein - Abstract
Elevated N-linked glycosylation of immunoglobulin G variable regions (IgG-VN-Glyc) is an emerging molecular phenotype associated with autoimmune disorders. To test the broader specificity of elevated IgG-VN-Glyc, we studied patients with distinct subtypes of myasthenia gravis (MG), a B cell-mediated autoimmune disease. Our experimental design included adaptive immune receptor repertoire sequencing to quantify and characterize N-glycosylation sites in the global B cell receptor repertoire, proteomics to examine glycosylation patterns of the circulating IgG, and production of human-derived recombinant autoantibodies, which were studied with mass spectrometry and antigen binding assays to confirm occupation of glycosylation sites and determine whether they alter binding. We found that the frequency of IgG-VN-Glyc motifs was increased in the B cell repertoire of MG patients when compared to healthy donors. Motifs were introduced by both biased V gene segment usage and somatic hypermutation. IgG-VN-Glyc could be observed in the circulating IgG in a subset of MG patients. Autoantigen binding, by patient-derived MG autoantigen-specific monoclonal antibodies with experimentally confirmed presence of IgG-VN-Glyc, was not altered by the glycosylation. Our findings extend prior work on patterns of variable region N-linked glycosylation in autoimmunity to MG subtypes. Although occupied IgG-VN-Glyc motifs are found on MG autoantigen-specific monoclonal antibodies, they are not required for binding to the autoantigen in this disease.
- Published
- 2021
23. Single-cell immunophenotyping of the skin lesion erythema migrans identifies IgM memory B cells
- Author
-
Steven H. Kleinstein, Alex K. Shalek, David A. Hafler, Kenneth R. Dardick, Ruth R. Montgomery, Ira Fleming, Hailong Meng, Kenneth B. Hoehn, Linda K. Bockenstedt, Khadir Raddassi, Ruoyi Jiang, and Alexia A. Belperron
- Subjects
0301 basic medicine ,Adult ,Male ,Antigen presentation ,B-cell receptor ,Immunology ,Adaptive immunity ,Somatic hypermutation ,Biology ,Immunophenotyping ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Bacterial infections ,medicine ,Humans ,RNA-Seq ,B cell ,Aged ,Skin ,B-Lymphocytes ,Lyme Disease ,Infectious disease ,Histocompatibility Antigens Class II ,General Medicine ,Middle Aged ,Acquired immune system ,030104 developmental biology ,medicine.anatomical_structure ,Immunoglobulin M ,030220 oncology & carcinogenesis ,biology.protein ,Erythema Chronicum Migrans ,Female ,Antibody ,Single-Cell Analysis ,Transcriptome ,Research Article - Abstract
The skin lesion erythema migrans (EM) is an initial sign of the Ixodes tick-transmitted Borreliella spirochetal infection known as Lyme disease. T cells and innate immune cells have previously been shown to predominate the EM lesion and promote the reaction. Despite the established importance of B cells and antibodies in preventing infection, the role of B cells in the skin immune response to Borreliella is unknown. Here, we used single-cell RNA-Seq in conjunction with B cell receptor (BCR) sequencing to immunophenotype EM lesions and their associated B cells and BCR repertoires. We found that B cells were more abundant in EM in comparison with autologous uninvolved skin; many were clonally expanded and had circulating relatives. EM-associated B cells upregulated the expression of MHC class II genes and exhibited preferential IgM isotype usage. A subset also exhibited low levels of somatic hypermutation despite a gene expression profile consistent with memory B cells. Our study demonstrates that single-cell gene expression with paired BCR sequencing can be used to interrogate the sparse B cell populations in human skin and reveals that B cells in the skin infection site in early Lyme disease expressed a phenotype consistent with local antigen presentation and antibody production.
- Published
- 2021
24. Cutting Edge: Distinct B Cell Repertoires Characterize Patients with Mild and Severe COVID-19
- Author
-
Steven H. Kleinstein, Palaniappan Ramanathan, David A. Hafler, Stuart C. Sealfon, Alexander Bukreyev, Avraham Unterman, Hiromitsu Asashima, Naftali Kaminski, Kenneth B. Hoehn, Charles S. Dela Cruz, and Tomokazu Sumida
- Subjects
Protective immunity ,2019-20 coronavirus outbreak ,B-Lymphocytes ,Coronavirus disease 2019 (COVID-19) ,SARS-CoV-2 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Immunology ,RNA ,Germinal center ,COVID-19 ,Biology ,Article ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Immunity ,medicine ,Immunology and Allergy ,Humans ,B cell ,030215 immunology - Abstract
Protective immunity against COVID-19 likely depends on the production of SARS-CoV-2–specific plasma cells and memory B cells postinfection or postvaccination. Previous work has found that germinal center reactions are disrupted in severe COVID-19. This may adversely affect long-term immunity against reinfection. Consistent with an extrafollicular B cell response, patients with severe COVID-19 have elevated frequencies of clonally expanded, class-switched, unmutated plasmablasts. However, it is unclear whether B cell populations in individuals with mild COVID-19 are similarly skewed. In this study, we use single-cell RNA sequencing of B cells to show that in contrast to patients with severe COVID-19, subjects with mildly symptomatic COVID-19 have B cell repertoires enriched for clonally diverse, somatically hypermutated memory B cells ∼30 d after the onset of symptoms. This provides evidence that B cell responses are less disrupted in mild COVID-19 and result in the production of memory B cells.
- Published
- 2021
25. Human B cell lineages engaged by germinal centers following influenza vaccination are measurably evolving
- Author
-
Ruoyi Jiang, Kenneth B. Hoehn, Frederick I. Miller, Ali H. Ellebedy, Jackson S. Turner, Oliver G. Pybus, and Steven H. Kleinstein
- Subjects
Affinity maturation ,Vaccination ,medicine.anatomical_structure ,Phylogenetic tree ,Immunity ,medicine ,Somatic hypermutation ,Germinal center ,Biology ,Virology ,B cell ,Virus - Abstract
Poor efficacy of seasonal influenza virus vaccines is often attributed to pre-existing immunity interfering with the persistence and maturation of vaccine-induced B cell responses.1 Consistent with this notion, no significant increase in somatic hypermutation (SHM) among circulating influenza-binding lineages was detected following seasonal vaccination in humans.2 A more recent study showed that at least a subset of vaccine-induced B cell lineages are recruited into germinal centers (GCs) following vaccination, suggesting that affinity maturation of these lineages can occur.3 Crucially, however, it has not been demonstrated whether these GC-engaged lineages actually accumulate additional SHM. Here, we address this point using a phylogenetic test of measurable evolution. We first validate this test through simulations and demonstrate measurable B cell evolution in known examples of affinity maturation such as the response to HIV infection. We then show that lineages in the blood are rarely measurably evolving following influenza vaccination, but that GC-engaged lineages - likely derived from memory B cells - are frequently measurably evolving. These findings confirm that seasonal influenza virus vaccination can stimulate additional SHM among responding B cell lineages.
- Published
- 2021
26. Post-infectious inflammatory disease in MIS-C features elevated cytotoxicity signatures and autoreactivity that correlates with severity
- Author
-
Steven H. Kleinstein, John S. Tsang, Neha Bansal, Neal G. Ravindra, Hiromitsu Asashima, David A. Hafler, Michela Comi, Xiting Yan, William W. Lau, Nina N. Brodsky, Carrie L. Lucas, Jason Bishai, Zuoheng Wang, Avraham Unterman, Yunqing Liu, Alamzeb Khan, Merrick Lopez, Richard W. Pierce, Anjali Ramaswamy, Brian Sellers, Aagam Shah, Kenneth B. Hoehn, David van Dijk, Harsha K. Chandnani, Rachel Sparks, Ningshan Li, Charles S. Dela Cruz, Naftali Kaminski, Tomokazu Sumida, Jason Catanzaro, Victoria Habet, and Andrew J. Rice
- Subjects
Immune system ,business.industry ,Immunopathology ,Immunology ,Cytotoxic T cell ,Medicine ,Disease ,medicine.symptom ,Complication ,business ,Cytotoxicity ,Asymptomatic ,Blood proteins - Abstract
SUMMARYMultisystem inflammatory syndrome in children (MIS-C) is a life-threatening post-infectious complication occurring unpredictably weeks after mild or asymptomatic SARS-CoV2 infection in otherwise healthy children. Here, we define immune abnormalities in MIS-C compared to adult COVID-19 and pediatric/adult healthy controls using single-cell RNA sequencing, antigen receptor repertoire analysis, unbiased serum proteomics, andin vitroassays. Despite no evidence of active infection, we uncover elevated S100A-family alarmins in myeloid cells and marked enrichment of serum proteins that map to myeloid cells and pathways including cytokines, complement/coagulation, and fluid shear stress in MIS-C patients. Moreover, NK and CD8 T cell cytotoxicity genes are elevated, and plasmablasts harboring IgG1 and IgG3 are expanded. Consistently, we detect elevated binding of serum IgG from severe MIS-C patients to activated human cardiac microvascular endothelial cells in culture. Thus, we define immunopathology features of MIS-C with implications for predicting and managing this SARS-CoV2-induced critical illness in children.
- Published
- 2020
27. Single-Cell Omics Reveals Dyssynchrony of the Innate and Adaptive Immune System in Progressive COVID-19
- Author
-
Santos Bermejo, Jonas C. Schupp, Guilin Wang, David van Dijk, Nima Nouri, Giuseppe DeIuliis, Anne L. Wyllie, Victor Gasque, Arnau Casanovas-Massana, Akiko Iwasaki, Wenxuan Deng, Patrick Wong, Xiting Yan, Avraham Unterman, Yunqing Liu, Anthony Melillo, Micha Sam Brickman Raredon, Shelli F. Farhadian, Neal G. Ravindra, Christopher Castaldi, Nathan D. Grubaugh, Carlos Cosme, John Fournier, Hiromitsu Asashima, Naftali Kaminski, Albert C. Shaw, Albert I. Ko, Maksym Minasyan, Chantal B.F. Vogels, Zuoheng Wang, David A. Hafler, Amy Y Zhao, Ming Chen, Hailong Meng, Ruth R. Montgomery, Steven H. Kleinstein, Laura E. Niklason, Lokesh Sharma, Ningshan Li, Charles S. Dela Cruz, Tomokazu Sumida, Hongyu Zhao, and Kenneth B. Hoehn
- Subjects
Immune system ,Innate immune system ,Interferon ,Immunopathology ,Immunology ,medicine ,Somatic hypermutation ,Cytotoxic T cell ,Biology ,Acquired immune system ,Virus ,medicine.drug - Abstract
A dysregulated immune response against the SARS-CoV-2 virus plays a critical role in severe COVID-19. However, the molecular and cellular mechanisms by which the virus causes lethal immunopathology are poorly understood. Here, we utilize multiomics single-cell analysis to probe dynamic immune responses in patients with stable or progressive manifestations of COVID-19, and assess the effects of tocilizumab, an anti-IL-6 receptor monoclonal antibody. Coordinated profiling of gene expression and cell lineage protein markers reveals a prominent type-1 interferon response across all immune cells, especially in progressive patients. An anti-inflammatory innate immune response and a pre-exhaustion phenotype in activated T cells are hallmarks of progressive disease. Skewed T cell receptor repertoires in CD8+ T cells and uniquely enriched V(D)J sequences are also identified in COVID-19 patients. B cell repertoire and somatic hypermutation analysis are consistent with a primary immune response, with possible contribution from memory B cells. Our in-depth immune profiling reveals dyssynchrony of the innate and adaptive immune interaction in progressive COVID-19, which may contribute to delayed virus clearance and has implications for therapeutic intervention.
- Published
- 2020
- Full Text
- View/download PDF
28. Single-cell repertoire tracing identifies rituximab-resistant B cells during myasthenia gravis relapses
- Author
-
Ruoyi Jiang, Kevin C. O’Connor, Minh C. Pham, Richard Nowak, Panos Stathopoulos, Kenneth B. Hoehn, Steven H. Kleinstein, and Miriam L. Fichtner
- Subjects
0301 basic medicine ,medicine.medical_treatment ,B-cell receptor ,Cell ,B-Lymphocyte Subsets ,medicine.disease_cause ,Autoimmunity ,Autoimmune Diseases ,03 medical and health sciences ,0302 clinical medicine ,Myasthenia Gravis ,medicine ,Humans ,Receptors, Cholinergic ,B cell ,Autoantibodies ,B-Lymphocytes ,Kinase ,business.industry ,Receptor Protein-Tyrosine Kinases ,General Medicine ,Immunotherapy ,medicine.disease ,Myasthenia gravis ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Rituximab ,Single-Cell Analysis ,business ,Transcriptome ,medicine.drug ,Research Article - Abstract
Rituximab, a B cell-depleting therapy, is indicated for treating a growing number of autoantibody-mediated autoimmune disorders. However, relapses can occur after treatment, and autoantibody-producing B cell subsets may be found during relapses. It is not understood whether these autoantibody-producing B cell subsets emerge from the failed depletion of preexisting B cells or are generated de novo. To further define the mechanisms that cause postrituximab relapse, we studied patients with autoantibody-mediated muscle-specific kinase (MuSK) myasthenia gravis (MG) who relapsed after treatment. We carried out single-cell transcriptional and B cell receptor profiling on longitudinal B cell samples. We identified clones present before therapy that persisted during relapse. Persistent B cell clones included both antibody-secreting cells and memory B cells characterized by gene expression signatures associated with B cell survival. A subset of persistent antibody-secreting cells and memory B cells were specific for the MuSK autoantigen. These results demonstrate that rituximab is not fully effective at eliminating autoantibody-producing B cells and provide a mechanistic understanding of postrituximab relapse in MuSK MG.
- Published
- 2020
29. BTG1 Mutation Promotes Aggressive Lymphoma Development By Lowering the Threshold to MYC Activation and Generating 'Super-Competitor' B Cells
- Author
-
Christopher R. Chin, Ekaterina Kots, Jonatan Ersching, Juhee Pae, Wyatt Morgan, Chen Zhengming, George Khelashvili, Huimin Geng, Jonah Melnick, Michael G. Kharas, Amy Chadburn, Ari Melnick, Hannah M Isles, Michael Meyer-Hermann, Ersilia Barin, Matthew R. Teater, Diu T.T. Nguyen, Kenneth B. Hoehn, Arnab Bandyopadhyay, David Scott, Coraline Mlynarczyk, Ling Wang, Theinmozhi Arulraj, Muhammad H Rehman, Hui Xian Poh, Samie R. Jaffrey, Hao Shen, Gabriel D. Victora, Olivier Elemento, and Ashley S. Doane
- Subjects
Immunology ,Mutation (genetic algorithm) ,Cancer research ,Aggressive lymphoma ,Cell Biology ,Hematology ,Biology ,Biochemistry ,BTG1 - Abstract
Somatic missense mutations of BTG1 are exclusive to germinal center (GC)-derived B cell lymphomas (~12% of DLBCLs) and are most prevalent in ABC-DLBCL (p=0.0184 vs GCB-DLBCL), particularly in the MCD/cluster 5 subtype, which features extranodal dissemination and unfavorable outcome. However, the relevance, mechanism of action and biological contribution of BTG1 mutations have not been studied. Using a rigorous genomic covariate analysis, we identified BTG1 mutations as a top genetic driver in DLBCL. Furthermore, molecular dynamics simulations indicated that BTG1 recurrent mutations, including the most frequent Q36H, disrupted the protein structure, with likely deleterious functional consequences. To investigate the effect of BTG1 mutation in GC B cells, we generated a conditional Btg1Q36H knock in mouse crossed to the B cell specific Cd19Cre line. Surprisingly, there was no apparent phenotype in GC B cells or other B cell populations. However, placing Btg1 Q36H and WT GC B cells in competition within the same mouse through adoptive transfer revealed a dramatic competitive advantage of Btg1 Q36H cells, virtually taking over the GC reaction. To gain further insight into this striking fitness advantage, we performed RNAseq in Btg1 Q36H GCs, which showed marked enrichment for genes induced in positively selected GC B cells, including MYC targets and biosynthetic pathways. The same genes were also enriched in BTG1 mutant DLBCL patients in 2 independent cohorts. Furthermore, Btg1 Q36H GC B cells displayed greater RNA content and cell size, reflecting increased fitness. Positive selection normally triggers a brief Myc pulse in GC B cells. We therefore crossed our Btg1Q36H mice to the MycGFPprotein fusion reporter and observed higher proportion of Myc GFP+ cells in Btg1 Q36H GCs. For mechanistic studies, we generated isogenic BTG1 Q36H or BTG1 WT human DLBCL cell lines. BTG1 Q36H cells exhibited enrichment for the same positively selected GC B and MYC target genes, as well as greater RNA content and cell size. BTG1 family members were suggested to interact with RNA. Performing RNA-immunoprecipitation, we discovered that ~800 transcripts associated with BTG1 WT, but not BTG1 Q36H. Notably, these corresponded to the same positively selected GC B and MYC target genes, including MYC itself. BTG1 was shown to regulate mRNA stability in other cell types. However, BTG1 Q36H did not alter MYC mRNA stability and instead facilitated MYC protein synthesis, thus disrupting a novel GC context-specific checkpoint mechanism, whereby BTG1 normally attenuates spurious MYC translation to tightly restrict fitness potential. In GC B cells, Myc induction coincides with S phase entry, but G2/M progression requires re-entry into the proliferative dark zone. To characterize GC dynamics in vivo, we performed targeted single cell RNAseq in competing Btg1 Q36H and WT GC B cells and noted earlier and higher proportion of positively selected Btg1 Q36H GC B cells having committed to G2/M and the proliferative program. We confirmed faster S phase completion in competing Btg1 Q36H GC B cells by in vivo EdU/BrdU labelling and greater re-entry into the proliferative dark zone by in vivo antigen delivery to synchronize GC B cells at the time of positive selection. Given that MCD-DLBCLs express high levels of BCL2, we crossed our Btg1Q36H mice to the VavP-Bcl2 model. As compared to VavP-Bcl2, VavP-Bcl2+Btg1 Q36H mice displayed shorter survival (p=0.0005), earlier onset of lymphoma, dysplastic B cell infiltration into non lymphoid organs and they contained highly mutated, selected and clonal tumor B cells. Moribund VavP-Bcl2+Btg1 Q36H mice uniquely featured sheets of large, immunoblastic lymphoma cells, characteristic of ABC-DLBCLs. Most notably, examining ABC-DLBCLs from 5 independent cohorts showed inferior clinical outcome for BTG1 mutant patients (p=0.0011) and independent association of BTG1 mutation with inferior overall survival by multivariable Cox regression (p=0.0190). Collectively, we find that BTG1 mutations mediate lymphomagenesis through an entirely novel mechanism of action that recapitulates the embryonic MYC-dependent "super-competitive" phenotype originally described in Drosophila imaginal disc cells. In the GC, "super-competition" is provided by BTG1 mutation via a subtle acceleration of MYC induction and GC dynamics, conferring dramatic fitness and the potential to transform into aggressive lymphomas. Disclosures Hoehn: Prellis Biologics: Consultancy. Elemento: Janssen: Research Funding; Freenome: Consultancy, Other: Current equity holder in a privately-held company; Volastra Therapeutics: Consultancy, Other: Current equity holder, Research Funding; Owkin: Consultancy, Other: Current equity holder; Champions Oncology: Consultancy; One Three Biotech: Consultancy, Other: Current equity holder; Eli Lilly: Research Funding; AstraZeneca: Research Funding; Johnson and Johnson: Research Funding. Scott: NanoString Technologies: Patents & Royalties: Patent describing measuring the proliferation signature in MCL using gene expression profiling.; BC Cancer: Patents & Royalties: Patent describing assigning DLBCL COO by gene expression profiling--licensed to NanoString Technologies. Patent describing measuring the proliferation signature in MCL using gene expression profiling. ; AstraZeneca: Consultancy; Abbvie: Consultancy; Celgene: Consultancy; Incyte: Consultancy; Janssen: Consultancy, Research Funding; Rich/Genentech: Research Funding. Melnick: Constellation: Consultancy; Epizyme: Consultancy; Daiichi Sankyo: Research Funding; Sanofi: Research Funding; Janssen Pharmaceuticals: Research Funding; KDAC Pharma: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2021
30. Overexpression of T-bet in HIV infection is associated with accumulation of B cells outside germinal centers and poor affinity maturation
- Author
-
Yuxing Li, Ryan G. Swanson, John S. Tsang, Kenneth B. Hoehn, Margery G. Smelkinson, Nicole A. Doria-Rose, Michael C. Sneller, Steven H. Kleinstein, James W. Austin, Yiannis Dimopoulos, Andrew J. Martins, Clarisa M. Buckner, Valerie A. Melson, Alexander Chassiakos, Sijy O'Dell, Tae-Wook Chun, Stefania Pittaluga, Wei Wang, Susan Moir, Lela Kardava, Constantinos Petrovas, Adrian B. McDermott, Richard Kwan, J. Nick Fisk, Catherine Seamon, Julian Q. Zhou, and Xiaozhen Zhang
- Subjects
Adult ,Male ,Antigens, CD19 ,B-cell receptor ,Antibody Affinity ,Receptors, Antigen, B-Cell ,chemical and pharmacologic phenomena ,HIV Infections ,Viremia ,Biology ,Article ,Affinity maturation ,Transcriptome ,Young Adult ,Mutation Rate ,medicine ,Humans ,Receptor ,Memory B cell ,B-Lymphocytes ,breakpoint cluster region ,Germinal center ,hemic and immune systems ,T-Lymphocytes, Helper-Inducer ,General Medicine ,Middle Aged ,Germinal Center ,medicine.disease ,Antibodies, Neutralizing ,Phenotype ,Immunology ,Cytokines ,Female ,Lymph Nodes ,T-Box Domain Proteins ,Immunologic Memory - Abstract
Nearly all chronic human infections are associated with alterations in the memory B cell (MBC) compartment, including a large expansion of CD19(hi)T-bet(hi) MBC in the peripheral blood of HIV-infected individuals with chronic viremia. Despite their prevalence, it is unclear how these B cells arise and whether they contribute to the inefficiency of antibody-mediated immunity in chronic infectious diseases. We addressed these questions by characterizing T-bet-expressing B cells in lymph nodes (LN) and identifying a strong T-bet signature among HIV-specific MBC associated with poor immunologic outcome. Confocal microscopy and quantitative imaging revealed that T-bet(hi) B cells in LN of HIV-infected chronically viremic individuals distinctly accumulated outside germinal centers (GC), which are critical for optimal antibody responses. In single-cell analyses, LN T-bet(hi) B cells of HIV-infected individuals were almost exclusively found among CD19(hi) MBC and expressed reduced GC-homing receptors. Furthermore, HIV-specific B cells of infected individuals were enriched among LN CD19(hi)T-bet(hi) MBC and displayed a distinct transcriptome, with features similar to CD19(hi)T-bet(hi) MBC in blood and LN GC B cells (GCBC). LN CD19(hi)T-bet(hi) MBC were also related to GCBC by B cell receptor (BCR)–based phylogenetic linkage but had lower BCR mutation frequencies and reduced HIV-neutralizing capacity, consistent with diminished participation in GC-mediated affinity selection. Thus, in the setting of chronic immune activation associated with HIV viremia, failure of HIV-specific B cells to enter or remain in GC may help explain the rarity of high-affinity protective antibodies.
- Published
- 2019
31. Single-cell repertoire tracing identifies rituximab refractory B cells during myasthenia gravis relapses
- Author
-
Panos Stathopoulos, Ruoyi Jiang, Kevin C. O’Connor, Steven H. Kleinstein, Richard Nowak, Miriam L. Fichtner, and Kenneth B. Hoehn
- Subjects
medicine.drug_class ,business.industry ,Cell ,B-cell receptor ,Autoantibody ,Somatic hypermutation ,Monoclonal antibody ,Isotype ,medicine.anatomical_structure ,medicine ,Cancer research ,Rituximab ,business ,B cell ,medicine.drug - Abstract
Rituximab, an anti-CD20 B cell-depleting therapy, is indicated to treat a growing number of autoimmune disorders. However, disease relapses can occur when the B cell compartment is re-established after treatment. To explore the mechanism of relapse, we studied patients with muscle-specific kinase (MuSK) myasthenia gravis (MG), a paradigm for B cell-mediated autoimmune diseases in which pathology is directly associated with autoantibody production. Whether the failed depletion of specific clones leads to the persistence of autoantibody producing B cell subsets has yet to be elucidated. Here we show that rituximab (RTX) therapy is not fully effective at eliminating disease-associated B cells and provide a mechanistic understanding of relapse. We carried out single-cell transcriptional and B cell receptor (BCR) profiling on longitudinal B cell samples from the peripheral blood of MuSK MG patients who relapsed after rituximab therapy. Computational analysis of these data identified individual B cells that were refractory to rituximab depletion by linking them to clones that were present prior to therapy and continued to persist after therapy. These persistent B cell clones were present at high frequency, and included both antibody-secreting cells and memory B cells. They were disproportionately isotype switched with elevated somatic hypermutation frequencies and a gene expression signature associated with antigen-experience and B cell clonal expansion. We further identified both antibody-secreting cells and memory B cells with specificity for MuSK autoantigen that were clonally expanded during relapse and persistent. Our results provide evidence that post-rituximab relapse in MuSK MG is associated with the failed depletion of autoantigen-specific B cell subsets. This work provides insight into the durability of human B cells during treatment with therapeutic CD20-specific monoclonal antibodies. The transcriptional signatures associated with B cell resistance may represent new therapeutic avenues for treatment and biomarkers of depletion efficacy.
- Published
- 2019
- Full Text
- View/download PDF
32. Mutant EZH2 Induces a Pre-malignant Lymphoma Niche by Reprogramming the Immune Response
- Author
-
Steven H. Kleinstein, Christopher E. Mason, Leandro Venturutti, Christian Steidl, Kenneth B. Hoehn, María T. Calvo-Fernández, Olivier Elemento, Katsuyoshi Takata, Neil L. Kelleher, C. David Allis, Johana Gutierrez, Alexey A. Soshnev, Jude M. Phillip, Wendy Béguelin, Cem Meydan, Martín A. Rivas, Jeannie M. Camarillo, Matt Teater, Ari Melnick, Karin Tarte, Cornell University [New York], Yale University School of Medicine, Rockefeller University [New York], Northwestern University [Evanston], British Columbia, Microenvironment, Cell Differentiation, Immunology and Cancer (MICMAC), Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Weill Medical College of Cornell University [New York], NCI CCSG [P30 CA060553], NCIUnited States Department of Health and Human ServicesNational Institutes of Health (NIH) - USANIH National Cancer Institute (NCI) [R35 CA220499, LLS TRP 6572-19, LLS SCOR 7013-17, RO1 CA198089], Damon Runyon Cancer Research Foundation, PhRMA Foundation Post-doctoral Fellowship in Informatics, LLS Career Development Program fellowship, Uehara Memorial FoundationUehara Memorial Foundation, Michael Smith Foundation for Health Research (Career Investigator Award)Michael Smith Foundation for Health Research, Terry Fox Research Institute, Canada LLS, [P41 GM108569], ASH Junior Faculty Scholar Award, Follicular Lymphoma Consortium, Chemotherapy Foundation, NIHUnited States Department of Health and Human ServicesNational Institutes of Health (NIH) - USA [RO1A1104739], Yale School of Medicine [New Haven, Connecticut] (YSM), and Université de Rennes (UR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique )
- Subjects
0301 basic medicine ,Cancer Research ,Lymphoma, B-Cell ,Follicular lymphoma ,immune microenvironment ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,macromolecular substances ,Biology ,Malignant transformation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,follicular lymphoma ,medicine ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,EZH2 ,Lymphoma, Follicular ,B cell ,B-Lymphocytes ,Follicular dendritic cells ,Germinal center ,Dendritic Cells ,medicine.disease ,Cellular Reprogramming ,Germinal Center ,Lymphoma ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,epigenetic dysregulation ,Reprogramming - Abstract
International audience; Follicular lymphomas (FLs) are slow-growing, indolent tumors containing extensive follicular dendritic cell (FDC) networks and recurrent EZH2 gain-of-function mutations. Paradoxically, FLs originate from highly proliferative germinal center (GC) B cells with proliferation strictly dependent on interactions with T follicular helper cells. Herein, we show that EZH2 mutations initiate FL by attenuating GC B cell requirement for T cell help and driving slow expansion of GC centrocytes that become enmeshed with and dependent on FDCs. By impairing T cell help, mutant EZH2 prevents induction of proliferative MYC programs. Thus, EZH2 mutation fosters malignant transformation by epigenetically reprograming B cells to form an aberrant immunological niche that reflects characteristic features of human FLs, explaining how indolent tumors arise from GC B cells.
- Published
- 2019
33. A framework for detecting natural selection on traits above the species level
- Author
-
Kenneth B. Hoehn, V. Louise Roth, and Paul G. Harnik
- Subjects
0106 biological sciences ,Extinction event ,010506 paleontology ,Natural selection ,Extinction ,Ecological Modeling ,Biology ,010603 evolutionary biology ,01 natural sciences ,Frequency ,Nested set model ,Statistics ,Trait ,Taxonomic rank ,Ecology, Evolution, Behavior and Systematics ,Selection (genetic algorithm) ,0105 earth and related environmental sciences - Abstract
Summary To what extent can natural selection act on groupings above the species level? Despite extensive theoretical discussion and growing practical concerns over increased rates of global ecological turnover, the question has largely evaded empirical resolution. A flexible and robust hypothesis-testing framework for detecting the phenomenon could facilitate significant progress in resolving this issue. We introduce a permutation-based approach, implemented in the R package perspectev, which provides an explicit test of whether empirical patterns of correlation between upper level trait values and survivorship are reducible to correlations manifested at lower levels. The package is applicable to virtually any nested set of upper- and lower level groupings, a wide variety of upper level traits, and both historical and contemporary occurrence data. We apply this approach to five paleontological data sets that represent different magnitudes of extinction and differ in taxonomic breadth, geological timing and geographic extent. Using simulations, we demonstrate that this method is a robust means of detecting irreducibility in the relationship between upper level traits and survivorship, and outline circumstances in which the method is less effective. We also find evidence consistent with previous findings of selection above the species level for geographic range size in North American K-Pg molluscs and show that this phenomenon was evident for the same molluscan genera globally. Ultimately, we conclude that at certain points in history, some higher level taxonomic groups have survived differentially with respect to geographic range size in a manner that is not explained by the same trait at the species level, and we show that evidence for this phenomenon varies across taxa and extinction events. We release our method as a flexible and easy-to-use R package that will allow others to help determine the relative frequency of this macroevolutionary phenomenon, both in the fossil record and in estimates of contemporary extinction risk.
- Published
- 2015
34. A phylogenetic codon substitution model for antibody lineages
- Author
-
Kenneth B. Hoehn, Oliver G. Pybus, and Gerton Lunter
- Subjects
Genetics ,0303 health sciences ,Mutation rate ,Phylogenetic tree ,Broadly neutralizing antibody ,Somatic hypermutation ,food and beverages ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Substitution model ,Codon substitution model ,Evolutionary biology ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Phylogenetic methods have shown promise in understanding the development of broadly neutralizing antibody lineages (bNAbs). However, the mutational process that generates these lineages – somatic hypermutation (SHM) – is biased by hotspot motifs, which violates important assumptions in most phylogenetic substitution models. Here, we develop a modified GY94-type substitution model that partially accounts for this context-dependency while preserving independence of sites during calculation. This model shows a substantially better fit to three well-characterized bNAb lineages than the standard GY94 model. We show through simulations that accounting for hotspot motifs can lead to reduced bias of other substitution parameters, and more accurate ancestral state reconstructions. We also demonstrate how our model can be used to test hypotheses concerning the roles of different hotspot and coldspot motifs in the evolution of B-cell lineages. Further, we explore the consequences of the idea that the number of hotspot motifs – and perhaps the mutation rate in general – is expected to decay over time in individual bNAb lineages.
- Published
- 2016
- Full Text
- View/download PDF
35. A Phylogenetic Codon Substitution Model for Antibody Lineages
- Author
-
Kenneth B, Hoehn, Gerton, Lunter, and Oliver G, Pybus
- Subjects
B-Lymphocytes ,Models, Genetic ,Investigations ,Genetics of Immunity ,Antibodies, Neutralizing ,Evolution, Molecular ,B-cell receptor ,antibody ,Mutation ,evolution ,Humans ,Cell Lineage ,phylogenetic tree ,Codon ,Phylogeny ,lineage - Abstract
Phylogenetic methods have shown promise in understanding the development of broadly neutralizing antibody lineages (bNAbs). However, the mutational process that generates these lineages, somatic hypermutation, is biased by hotspot motifs which violates important assumptions in most phylogenetic substitution models. Here, we develop a modified GY94-type substitution model that partially accounts for this context dependency while preserving independence of sites during calculation. This model shows a substantially better fit to three well-characterized bNAb lineages than the standard GY94 model. We also demonstrate how our model can be used to test hypotheses concerning the roles of different hotspot and coldspot motifs in the evolution of B-cell lineages. Further, we explore the consequences of the idea that the number of hotspot motifs, and perhaps the mutation rate in general, is expected to decay over time in individual bNAb lineages.
- Published
- 2016
36. Modelling the Decay of Hotspot Motifs in Broadly Neutralizing Antibody Lineages
- Author
-
Oliver G. Pybus, Gerton Lunter, and Kenneth B. Hoehn
- Subjects
Genetics ,0303 health sciences ,Mutation rate ,Phylogenetic tree ,Broadly neutralizing antibody ,Somatic hypermutation ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Substitution model ,Evolutionary biology ,Hotspot (geology) ,030304 developmental biology ,030215 immunology - Abstract
Phylogenetic methods have shown great promise in understanding the development of broadly neutralizing antibody lineages (bNAbs). However, mutational process for generating these lineages - somatic hypermutation (SHM) - is biased by hotspot motifs, which violates important assumptions in most phylogenetic substitution models. Here, we develop a modified GY94-type substitution model which partially accounts for this context-dependency while preserving independence of sites in calculation. This model shows a substantially better fit to three well-characterized bNAb lineages than the standard GY94 model. We show through simulations that accounting for this can lead to reduced bias of other substitution parameters, and more accurate ancestral state reconstructions. We further explore other implications of this model; namely, that the number of hotspot motifs - and therefore likely the mutation rate in general - is expected to decay over time in individual bNAb lineages.
- Published
- 2016
37. The Diversity and Molecular Evolution of B-Cell Receptors during Infection
- Author
-
Kenneth B, Hoehn, Anna, Fowler, Gerton, Lunter, and Oliver G, Pybus
- Subjects
B-Lymphocytes ,molecular evolution ,Antibody Affinity ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,Immunoglobulins ,Membrane Proteins ,Receptors, Antigen, B-Cell ,Adaptive Immunity ,Infections ,diversity ,Evolution, Molecular ,B-cell receptor ,Mutation ,infection ,Humans ,Fast Track ,immunoglobulin - Abstract
B-cell receptors (BCRs) are membrane-bound immunoglobulins that recognize and bind foreign proteins (antigens). BCRs are formed through random somatic changes of germline DNA, creating a vast repertoire of unique sequences that enable individuals to recognize a diverse range of antigens. After encountering antigen for the first time, BCRs undergo a process of affinity maturation, whereby cycles of rapid somatic mutation and selection lead to improved antigen binding. This constitutes an accelerated evolutionary process that takes place over days or weeks. Next-generation sequencing of the gene regions that determine BCR binding has begun to reveal the diversity and dynamics of BCR repertoires in unprecedented detail. Although this new type of sequence data has the potential to revolutionize our understanding of infection dynamics, quantitative analysis is complicated by the unique biology and high diversity of BCR sequences. Models and concepts from molecular evolution and phylogenetics that have been applied successfully to rapidly evolving pathogen populations are increasingly being adopted to study BCR diversity and divergence within individuals. However, BCR dynamics may violate key assumptions of many standard evolutionary methods, as they do not descend from a single ancestor, and experience biased mutation. Here, we review the application of evolutionary models to BCR repertoires and discuss the issues we believe need be addressed for this interdisciplinary field to flourish.
- Published
- 2016
38. How big is your Y? A genome sequence-based estimate of the size of the male-specific region in Megaselia scalaris
- Author
-
Mohamed A. F. Noor and Kenneth B. Hoehn
- Subjects
Male ,neo-Y chromosome ,Genome, Insect ,Biology ,Investigations ,Y chromosome ,Genome ,Transposition (music) ,Evolution, Molecular ,Megaselia scalaris ,Y Chromosome ,Genetics ,Animals ,sex chromosome evolution ,Molecular Biology ,Small supernumerary marker chromosome ,Genetics (clinical) ,Whole genome sequencing ,Diptera ,Chromosome ,biology.organism_classification ,Female ,YGS ,male-specific ,Sex linkage - Abstract
The scuttle fly, Megaselia scalaris, is often cited as a model in which to study early sex chromosome evolution because of its homomorphic sex chromosomes, low but measurable molecular differentiation between sex chromosomes, and occasional transposition of the male-determining element to different chromosomes in laboratory cultures. Counterintuitively, natural isolates consistently show sex linkage to the second chromosome. Frequent natural transposition of the male-determining element should lead to the loss of male specificity of any nontransposed material on the previous sex-linked chromosome pair. Using next-generation sequencing data from a newly obtained natural isolate of M. scalaris, we show that even highly conservative estimates for the size of the male-specific genome are likely too large to be contained within a transposable element. This result strongly suggests that transposition of the male-determining region either is extremely rare or has not persisted recently in natural populations, allowing for differentiation of the sex chromosomes of this species.
- Published
- 2014
39. Effects of premature termination codon polymorphisms in the Drosophila pseudoobscura subclade
- Author
-
Mohamed A. F. Noor, Suzanne E. McGaugh, and Kenneth B. Hoehn
- Subjects
endocrine system diseases ,Nonsense mutation ,Genome, Insect ,Genome ,Polymerase Chain Reaction ,Article ,Drosophila pseudoobscura ,Genetics ,Animals ,Least-Squares Analysis ,Molecular Biology ,Gene ,Ecology, Evolution, Behavior and Systematics ,Recombination, Genetic ,biology ,Computational Biology ,Reproducibility of Results ,Subclade ,Sequence Analysis, DNA ,biology.organism_classification ,Null allele ,Codon, Nonsense ,Organ Specificity ,Codon usage bias ,Drosophila ,Reference genome - Abstract
Premature termination codon (PTC) mutations can have dramatic effects--both adaptive and deleterious--on gene expression and function. Here, we examine the number and selective effects of PTC mutations within the Drosophila pseudoobscura subclade using 18 resequenced genomes aligned to the reference genome. We located and characterized 1,679 PTC mutations in 605 genes across each of these genomes relative to the D. pseudoobscura reference genome, and use RT-PCR to confirm transcription of a subset of these genes containing PTC mutations. We confirm previous findings that genes containing PTC mutations are less selectively constrained and less broadly expressed than non-PTC-containing genes, suggesting that the most of these mutations are at least mildly deleterious. Further, we find highly significant codon usage bias in regions downstream of the PTC in 38 of these PTC-containing genes, suggesting that some of these PTC mutations--if not alternatively spliced out of the transcript--have neutral effects. Ultimately, these analyzes support the view that the PTC mutations are mostly detrimental, but are nonetheless common enough in genomes that a subset could be effectively neutral.
- Published
- 2012
40. Effects of inversions on within- and between-species recombination and divergence
- Author
-
Mohamed A. F. Noor, Kenneth B. Hoehn, and Laurie S. Stevison
- Subjects
0106 biological sciences ,Heterozygote ,010603 evolutionary biology ,01 natural sciences ,Genome ,Polymorphism, Single Nucleotide ,Chromosomes ,Drosophila pseudoobscura ,Evolution, Molecular ,03 medical and health sciences ,chemistry.chemical_compound ,Gene Frequency ,Species Specificity ,Genetics ,Animals ,Ecology, Evolution, Behavior and Systematics ,Crosses, Genetic ,Research Articles ,030304 developmental biology ,Hybrid ,Chromosomal inversion ,Drosophila persimilis ,Recombination, Genetic ,0303 health sciences ,biology ,Models, Genetic ,Chimera ,Chromosome ,DNA ,Sequence Analysis, DNA ,biology.organism_classification ,chromosomal speciation ,recombination ,chemistry ,Chromosome Inversion ,inversions ,Drosophila ,interchromosomal effect ,Recombination - Abstract
Chromosomal inversions disrupt recombination in heterozygotes by both reducing crossing-over within inverted regions and increasing it elsewhere in the genome. The reduction of recombination in inverted regions facilitates the maintenance of hybridizing species, as outlined by various models of chromosomal speciation. We present a comprehensive comparison of the effects of inversions on recombination rates and on nucleotide divergence. Within an inversion differentiating Drosophila pseudoobscura and Drosophila persimilis, we detected one double recombinant among 9,739 progeny from F(1) hybrids screened, consistent with published double-crossover frequencies observed within species. Despite similar rates of exchange within and between species, we found no sequence-based evidence of ongoing gene exchange between species within this inversion, but significant exchange was inferred within species. We also observed greater differentiation at regions near inversion breakpoints between species versus within species. Moreover, we observed strong "interchromosomal effect" (higher recombination in inversion heterozygotes between species) with up to 9-fold higher recombination rates along collinear segments of chromosome two in hybrids. Further, we observed that regions most susceptible to changes in recombination rates corresponded to regions with lower recombination rates in homokaryotypes. Finally, we showed that interspecies nucleotide divergence is lower in regions with greater increases in recombination rate, potentially resulting from greater interspecies exchange. Overall, we have identified several similarities and differences between inversions segregating within versus between species in their effects on recombination and divergence. We conclude that these differences are most likely due to lower frequency of heterokaryotypes and to fitness consequences from the accumulation of various incompatibilities between species. Additionally, we have identified possible effects of inversions on interspecies gene exchange that had not been considered previously.
- Published
- 2011
41. A Method To Identify Copy Number Aberrations (CNAs) From Whole Exome Sequence (WES) Data and Its Application To Multiple Myeloma Cell Lines and Patient Samples
- Author
-
Jonathan J Keats, Kevin A Kwei, Brian B Tuch, Kenneth B. Hoehn, Jeremiah D. Degenhardt, and Christopher J. Kirk
- Subjects
Whole genome sequencing ,Immunology ,Cancer ,Cell Biology ,Hematology ,Computational biology ,Biology ,Bioinformatics ,medicine.disease ,Biochemistry ,Carfilzomib ,chemistry.chemical_compound ,chemistry ,medicine ,Copy number aberration ,Exome ,Multiple myeloma ,Exome sequencing ,Sequence (medicine) - Abstract
Duplications and deletions of large-scale genomic regions, including whole chromosomes, are a hallmark of many malignant tumors. Gross cytogenetic changes of this type were among the earliest genomic lesions to be observed in tumors and represent an important predictor of outcome for many tumor types. In multiple myeloma, a core set of structural variants are regularly used for disease prognosis (e.g., del17p, t(4;14), t(14,16)). The presence of one or more of these lesions is predictive of a more aggressive disease type and poor outcomes for patients. The application of next-generation sequencing (NGS) to cancer samples has enhanced our ability to detect copy number aberrations (CNAs) and identify driver mutations. The NGS approach can generate higher resolution maps of these mutations than traditional methods, which in turn enables faster identification of the functionally relevant genes they harbor. Most NGS-based methods for the identification of CNAs in tumors require whole genome sequencing (WGS) as input. Relatively fewer methods exist for the detection of CNAs from exome sequencing and, to our knowledge, these methods all rely on having matched normal data. However, WES is still far more common than WGS and matched normal samples are frequently not available. Therefore, we have developed a novel method for the identification of CNAs from single-sample WES data. Our method combines LOWESS smoothing & discrete-wavelet-transformation to normalize the exome coverage data with an HMM for coverage segmentation. We have applied our new method to WES data from 32 myeloma cell-lines for which aCGH copy number calls were also available for training and validation. Our method shows a high correlation with the aCGH calls (mean = 95%). We are currently applying the method to a set of 102 CD138+ tumor cell samples that were collected during patient screening on three Onyx-sponsored Phase 2 clinical trials of the proteasome inhibitor carfilzomib. We will report on the frequency of known and novel lesions in these samples, comparing to what we’ve observed in cell lines and other publically available CNA datasets. Disclosures: Degenhardt: Onyx Pharmaceuticals: Employment, Equity Ownership. Hoehn:Onyx Pharmaceuticals: Employment. Kwei:Onyx Pharmaceuticals: Employment, Equity Ownership. Kirk:Onyx Pharmaceuticals: Employment, Equity Ownership. Tuch:Onyx Pharmaceuticals: Employment, Equity Ownership.
- Published
- 2013
42. Erratum to: Effects of Premature Termination Codon Polymorphisms in the Drosophila pseudoobscura Subclade
- Author
-
Mohamed A. F. Noor, Suzanne E. McGaugh, and Kenneth B. Hoehn
- Subjects
Drosophila pseudoobscura ,Genetics ,biology ,Evolutionary biology ,Subclade ,Premature Termination Codon ,biology.organism_classification ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics - Abstract
Erratum to: J Mol Evol (2012) 75:141–150DOI 10.1007/s00239-012-9528-xOur original paper neglected to acknowledge that SteveSchaeffer and Andre Wallace genotyped and provided thePikes Peak lines of Drosophila pseudoobscura forsequencing. We thank them for this contribution andapologies for neglecting to mention it directly.
- Published
- 2012
43. Human B cell lineages associated with germinal centers following influenza vaccination are measurably evolving
- Author
-
Kenneth B Hoehn, Jackson S Turner, Frederick I Miller, Ruoyi Jiang, Oliver G Pybus, Ali H Ellebedy, and Steven H Kleinstein
- Subjects
B cell ,somatic hypermutation ,phylogenetics ,B cell receptor ,measurable evolution ,temporal evolution ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The poor efficacy of seasonal influenza virus vaccines is often attributed to pre-existing immunity interfering with the persistence and maturation of vaccine-induced B cell responses. We previously showed that a subset of vaccine-induced B cell lineages are recruited into germinal centers (GCs) following vaccination, suggesting that affinity maturation of these lineages against vaccine antigens can occur. However, it remains to be determined whether seasonal influenza vaccination stimulates additional evolution of vaccine-specific lineages, and previous work has found no significant increase in somatic hypermutation among influenza-binding lineages sampled from the blood following seasonal vaccination in humans. Here, we investigate this issue using a phylogenetic test of measurable immunoglobulin sequence evolution. We first validate this test through simulations and survey measurable evolution across multiple conditions. We find significant heterogeneity in measurable B cell evolution across conditions, with enrichment in primary response conditions such as HIV infection and early childhood development. We then show that measurable evolution following influenza vaccination is highly compartmentalized: while lineages in the blood are rarely measurably evolving following influenza vaccination, lineages containing GC B cells are frequently measurably evolving. Many of these lineages appear to derive from memory B cells. We conclude from these findings that seasonal influenza virus vaccination can stimulate additional evolution of responding B cell lineages, and imply that the poor efficacy of seasonal influenza vaccination is not due to a complete inhibition of vaccine-specific B cell evolution.
- 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.