Try a new search

Format these results:

Searched for:

person:littmd01

in-biosketch:yes

Total Results:

367


Multimodal single-cell datasets characterize antigen-specific CD8+ T cells across SARS-CoV-2 vaccination and infection

Zhang, Bingjie; Upadhyay, Rabi; Hao, Yuhan; Samanovic, Marie I; Herati, Ramin S; Blair, John D; Axelrad, Jordan; Mulligan, Mark J; Littman, Dan R; Satija, Rahul
The immune response to SARS-CoV-2 antigen after infection or vaccination is defined by the durable production of antibodies and T cells. Population-based monitoring typically focuses on antibody titer, but there is a need for improved characterization and quantification of T cell responses. Here, we used multimodal sequencing technologies to perform a longitudinal analysis of circulating human leukocytes collected before and after immunization with the mRNA vaccine BNT162b2. Our data indicated distinct subpopulations of CD8+ T cells, which reliably appeared 28 days after prime vaccination. Using a suite of cross-modality integration tools, we defined their transcriptome, accessible chromatin landscape and immunophenotype, and we identified unique biomarkers within each modality. We further showed that this vaccine-induced population was SARS-CoV-2 antigen-specific and capable of rapid clonal expansion. Moreover, we identified these CD8+ T cell populations in scRNA-seq datasets from COVID-19 patients and found that their relative frequency and differentiation outcomes were predictive of subsequent clinical outcomes.
PMID: 37735591
ISSN: 1529-2916
CID: 5606242

Enforced gut homing of murine regulatory T cells reduces early graft-versus-host disease severity

Larson, Jemma H; Jin, Sujeong; Loschi, Michael; Wagers, Sara Bolivar; Thangavelu, Govindarajan; Zaiken, Michael C; McDonald-Hyman, Cameron; Saha, Asim; Aguilar, Ethan G; Koehn, Brent; Osborn, Mark J; Panoskaltsis-Mortari, Angela; Macdonald, Kelli P A; Hill, Geoffrey R; Murphy, William J; Serody, Jonathan S; Maillard, Ivan; Kean, Leslie S; Kim, Sangwon V; Littman, Dan R; Blazar, Bruce R
Damage to the gastrointestinal tract following allogeneic hematopoietic stem cell transplantation is a significant contributor to the severity and perpetuation of graft-versus-host disease. In preclinical models and clinical trials, we showed that infusing high numbers of regulatory T-cells reduces graft-versus-host disease incidence. Despite no change in in vitro suppressive function, transfer of ex vivo expanded regulatory T-cells transduced to overexpress G-protein coupled receptor 15 or C-C Motif Chemokine Receptor 9, specific homing receptors for colon or small intestine, respectively, lessened graft-versus-host disease severity in mice. Increased regulatory T-cell frequency and retention within the gastrointestinal tissues of mice that received gut homing T-cells correlated with lower inflammation and gut damage early post-transplant, decreased graft-versus-host disease severity and prolonged survival compared to those receiving control transduced regulatory T-cells. These data provide evidence that enforced targeting of ex vivo expanded regulatory T-cells to the gastrointestinal tract diminishes gut injury and is associated with decreased graft-versus-host disease severity.
PMID: 36878433
ISSN: 1600-6143
CID: 5432592

Alterations in the gut microbiome implicate key taxa and metabolic pathways across inflammatory arthritis phenotypes

Thompson, Kelsey N; Bonham, Kevin S; Ilott, Nicholas E; Britton, Graham J; Colmenero, Paula; Bullers, Samuel J; McIver, Lauren J; Ma, Siyuan; Nguyen, Long H; Filer, Andrew; Brough, India; Pearson, Claire; Moussa, Caroline; Kumar, Vinod; Lam, Lilian H; Jackson, Matthew A; Pawluk, April; ,; Kiriakidis, Serafim; Taylor, Peter C; Wedderburn, Lucy R; Marsden, Brian; Young, Stephen P; Littman, Dan R; Faith, Jeremiah J; Pratt, Arthur G; Bowness, Paul; Raza, Karim; Powrie, Fiona; Huttenhower, Curtis; ,
Musculoskeletal diseases affect up to 20% of adults worldwide. The gut microbiome has been implicated in inflammatory conditions, but large-scale metagenomic evaluations have not yet traced the routes by which immunity in the gut affects inflammatory arthritis. To characterize the community structure and associated functional processes driving gut microbial involvement in arthritis, the Inflammatory Arthritis Microbiome Consortium investigated 440 stool shotgun metagenomes comprising 221 adults diagnosed with rheumatoid arthritis, ankylosing spondylitis, or psoriatic arthritis and 219 healthy controls and individuals with joint pain without an underlying inflammatory cause. Diagnosis explained about 2% of gut taxonomic variability, which is comparable in magnitude to inflammatory bowel disease. We identified several candidate microbes with differential carriage patterns in patients with elevated blood markers for inflammation. Our results confirm and extend previous findings of increased carriage of typically oral and inflammatory taxa and decreased abundance and prevalence of typical gut clades, indicating that distal inflammatory conditions, as well as local conditions, correspond to alterations to the gut microbial composition. We identified several differentially encoded pathways in the gut microbiome of patients with inflammatory arthritis, including changes in vitamin B salvage and biosynthesis and enrichment of iron sequestration. Although several of these changes characteristic of inflammation could have causal roles, we hypothesize that they are mainly positive feedback responses to changes in host physiology and immune homeostasis. By connecting taxonomic alternations to functional alterations, this work expands our understanding of the shifts in the gut ecosystem that occur in response to systemic inflammation during arthritis.
PMID: 37494472
ISSN: 1946-6242
CID: 5592322

Multimodal characterization of antigen-specific CD8 + T cells across SARS-CoV-2 vaccination and infection

Zhang, Bingjie; Upadhyay, Rabi; Hao, Yuhan; Samanovic, Marie I; Herati, Ramin S; Blair, John; Axelrad, Jordan; Mulligan, Mark J; Littman, Dan R; Satija, Rahul
The human immune response to SARS-CoV-2 antigen after infection or vaccination is defined by the durable production of antibodies and T cells. Population-based monitoring typically focuses on antibody titer, but there is a need for improved characterization and quantification of T cell responses. Here, we utilize multimodal sequencing technologies to perform a longitudinal analysis of circulating human leukocytes collected before and after BNT162b2 immunization. Our data reveal distinct subpopulations of CD8 + T cells which reliably appear 28 days after prime vaccination (7 days post boost). Using a suite of cross-modality integration tools, we define their transcriptome, accessible chromatin landscape, and immunophenotype, and identify unique biomarkers within each modality. By leveraging DNA-oligo-tagged peptide-MHC multimers and T cell receptor sequencing, we demonstrate that this vaccine-induced population is SARS-CoV-2 antigen-specific and capable of rapid clonal expansion. Moreover, we also identify these CD8 + populations in scRNA-seq datasets from COVID-19 patients and find that their relative frequency and differentiation outcomes are predictive of subsequent clinical outcomes. Our work contributes to our understanding of T cell immunity, and highlights the potential for integrative and multimodal analysis to characterize rare cell populations.
PMCID:9900816
PMID: 36747786
ISSN: 2692-8205
CID: 5522692

TH17 cell heterogeneity and its role in tissue inflammation

Schnell, Alexandra; Littman, Dan R; Kuchroo, Vijay K
Since their discovery almost two decades ago, interleukin-17-producing CD4+ T cells (TH17 cells) have been implicated in the pathogenesis of multiple autoimmune and inflammatory disorders. In addition, TH17 cells have been found to play an important role in tissue homeostasis, especially in the intestinal mucosa. Recently, the use of single-cell technologies, along with fate mapping and various mutant mouse models, has led to substantial progress in the understanding of TH17 cell heterogeneity in tissues and of TH17 cell plasticity leading to alternative T cell states and differing functions. In this Review, we discuss the heterogeneity of TH17 cells and the role of this heterogeneity in diverse functions of TH17 cells from homeostasis to tissue inflammation. In addition, we discuss TH17 cell plasticity and its incorporation into the current understanding of T cell subsets and alternative views on the role of TH17 cells in autoimmune and inflammatory diseases.
PMID: 36596896
ISSN: 1529-2916
CID: 5409922

Transcription factor RORα enforces stability of the Th17 cell effector program by binding to a Rorc cis-regulatory element

Hall, Jason A; Pokrovskii, Maria; Kroehling, Lina; Kim, Bo-Ram; Kim, Seung Yong; Wu, Lin; Lee, June-Yong; Littman, Dan R
T helper 17 (Th17) cells regulate mucosal barrier defenses but also promote multiple autoinflammatory diseases. Although many molecular determinants of Th17 cell differentiation have been elucidated, the transcriptional programs that sustain Th17 cells in vivo remain obscure. The transcription factor RORγt is critical for Th17 cell differentiation; however, it is not clear whether the closely related RORα, which is co-expressed in Th17 cells, has a distinct role. Here, we demonstrated that although dispensable for Th17 cell differentiation, RORα was necessary for optimal Th17 responses in peripheral tissues. The absence of RORα in T cells led to reductions in both RORγt expression and effector function among Th17 cells. Cooperative binding of RORα and RORγt to a previously unidentified Rorc cis-regulatory element was essential for Th17 lineage maintenance in vivo. These data point to a non-redundant role of RORα in Th17 lineage maintenance via reinforcement of the RORγt transcriptional program.
PMID: 36243007
ISSN: 1097-4180
CID: 5358332

Gut microbiome dysbiosis in antibiotic-treated COVID-19 patients is associated with microbial translocation and bacteremia

Bernard-Raichon, Lucie; Venzon, Mericien; Klein, Jon; Axelrad, Jordan E; Zhang, Chenzhen; Sullivan, Alexis P; Hussey, Grant A; Casanovas-Massana, Arnau; Noval, Maria G; Valero-Jimenez, Ana M; Gago, Juan; Putzel, Gregory; Pironti, Alejandro; Wilder, Evan; Thorpe, Lorna E; Littman, Dan R; Dittmann, Meike; Stapleford, Kenneth A; Shopsin, Bo; Torres, Victor J; Ko, Albert I; Iwasaki, Akiko; Cadwell, Ken; Schluter, Jonas
Although microbial populations in the gut microbiome are associated with COVID-19 severity, a causal impact on patient health has not been established. Here we provide evidence that gut microbiome dysbiosis is associated with translocation of bacteria into the blood during COVID-19, causing life-threatening secondary infections. We first demonstrate SARS-CoV-2 infection induces gut microbiome dysbiosis in mice, which correlated with alterations to Paneth cells and goblet cells, and markers of barrier permeability. Samples collected from 96 COVID-19 patients at two different clinical sites also revealed substantial gut microbiome dysbiosis, including blooms of opportunistic pathogenic bacterial genera known to include antimicrobial-resistant species. Analysis of blood culture results testing for secondary microbial bloodstream infections with paired microbiome data indicates that bacteria may translocate from the gut into the systemic circulation of COVID-19 patients. These results are consistent with a direct role for gut microbiome dysbiosis in enabling dangerous secondary infections during COVID-19.
PMID: 36319618
ISSN: 2041-1723
CID: 5358262

Provocateurs of autoimmunity within the gut microbiota

Upadhyay, Rabi; Littman, Dan R
An arthritogenic strain of Subdoligranulum in the gut elicits a local immune response, a precursor to systemic autoimmunity (Chriswell et al.).
PMID: 36288277
ISSN: 1946-6242
CID: 5348492

A RORgammat+ cell instructs gut microbiota-specific Treg cell differentiation

Kedmi, Ranit; Najar, Tariq A; Mesa, Kailin R; Grayson, Allyssa; Kroehling, Lina; Hao, Yuhan; Hao, Stephanie; Pokrovskii, Maria; Xu, Mo; Talbot, Jhimmy; Wang, Jiaxi; Germino, Joe; Lareau, Caleb A; Satpathy, Ansuman T; Anderson, Mark S; Laufer, Terri M; Aifantis, Iannis; Bartleson, Juliet M; Allen, Paul M; Paidassi, Helena; Gardner, James M; Stoeckius, Marlon; Littman, Dan R
The mutualistic relationship of gut-resident microbiota and the host immune system promotes homeostasis that ensures maintenance of the microbial community and of a largely non-aggressive immune cell compartment1,2. The consequences of disturbing this balance include proximal inflammatory conditions, such as Crohn's disease, and systemic illnesses. This equilibrium is achieved in part through the induction of both effector and suppressor arms of the adaptive immune system. Helicobacter species induce T regulatory (Treg) and T follicular helper (TFH) cells under homeostatic conditions, but induce inflammatory T helper 17 (TH17) cells when induced Treg (iTreg) cells are compromised3,4. How Helicobacter and other gut bacteria direct T cells to adopt distinct functions remains poorly understood. Here we investigated the cells and molecular components required for iTreg cell differentiation. We found that antigen presentation by cells expressing RORγt, rather than by classical dendritic cells, was required and sufficient for induction of Treg cells. These RORγt+ cells-probably type 3 innate lymphoid cells and/or Janus cells5-require the antigen-presentation machinery, the chemokine receptor CCR7 and the TGFβ activator αv integrin. In the absence of any of these factors, there was expansion of pathogenic TH17 cells instead of iTreg cells, induced by CCR7-independent antigen-presenting cells. Thus, intestinal commensal microbes and their products target multiple antigen-presenting cells with pre-determined features suited to directing appropriate T cell differentiation programmes, rather than a common antigen-presenting cell that they endow with appropriate functions.
PMID: 36071167
ISSN: 1476-4687
CID: 5332522

Publisher Correction: A RORγt+ cell instructs gut microbiota-specific Treg cell differentiation

Kedmi, Ranit; Najar, Tariq A; Mesa, Kailin R; Grayson, Allyssa; Kroehling, Lina; Hao, Yuhan; Hao, Stephanie; Pokrovskii, Maria; Xu, Mo; Talbot, Jhimmy; Wang, Jiaxi; Germino, Joe; Lareau, Caleb A; Satpathy, Ansuman T; Anderson, Mark S; Laufer, Terri M; Aifantis, Iannis; Bartleson, Juliet M; Allen, Paul M; Paidassi, Helena; Gardner, James M; Stoeckius, Marlon; Littman, Dan R
PMID: 36151471
ISSN: 1476-4687
CID: 5335832