Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 222
Filter
Add more filters

Publication year range
1.
Cell ; 184(15): 4048-4063.e32, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34233165

ABSTRACT

Microglia, the resident immune cells of the brain, have emerged as crucial regulators of synaptic refinement and brain wiring. However, whether the remodeling of distinct synapse types during development is mediated by specialized microglia is unknown. Here, we show that GABA-receptive microglia selectively interact with inhibitory cortical synapses during a critical window of mouse postnatal development. GABA initiates a transcriptional synapse remodeling program within these specialized microglia, which in turn sculpt inhibitory connectivity without impacting excitatory synapses. Ablation of GABAB receptors within microglia impairs this process and leads to behavioral abnormalities. These findings demonstrate that brain wiring relies on the selective communication between matched neuronal and glial cell types.


Subject(s)
Microglia/metabolism , Neural Inhibition/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Behavior, Animal , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Parvalbumins/metabolism , Phenotype , Receptors, GABA-B/metabolism , Synapses/physiology , Transcription, Genetic
3.
Cell ; 163(2): 381-93, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26411290

ABSTRACT

RORγt(+) Th17 cells are important for mucosal defenses but also contribute to autoimmune disease. They accumulate in the intestine in response to microbiota and produce IL-17 cytokines. Segmented filamentous bacteria (SFB) are Th17-inducing commensals that potentiate autoimmunity in mice. RORγt(+) T cells were induced in mesenteric lymph nodes early after SFB colonization and distributed across different segments of the gastrointestinal tract. However, robust IL-17A production was restricted to the ileum, where SFB makes direct contact with the epithelium and induces serum amyloid A proteins 1 and 2 (SAA1/2), which promote local IL-17A expression in RORγt(+) T cells. We identified an SFB-dependent role of type 3 innate lymphoid cells (ILC3), which secreted IL-22 that induced epithelial SAA production in a Stat3-dependent manner. This highlights the critical role of tissue microenvironment in activating effector functions of committed Th17 cells, which may have important implications for how these cells contribute to inflammatory disease.


Subject(s)
Gastrointestinal Microbiome , Interleukins/metabolism , Intestines/immunology , Receptors, Interleukin/metabolism , Serum Amyloid A Protein/metabolism , Th17 Cells/immunology , Animals , Immunity, Innate , Interleukins/immunology , Intestines/anatomy & histology , Intestines/microbiology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Receptors, Interleukin/immunology , Signal Transduction , Interleukin-22
4.
Nat Immunol ; 18(4): 412-421, 2017 04.
Article in English | MEDLINE | ID: mdl-28166218

ABSTRACT

Type 1 regulatory T cells (Tr1 cells) are induced by interleukin-27 (IL-27) and have critical roles in the control of autoimmunity and resolution of inflammation. We found that the transcription factors IRF1 and BATF were induced early on after treatment with IL-27 and were required for the differentiation and function of Tr1 cells in vitro and in vivo. Epigenetic and transcriptional analyses revealed that both transcription factors influenced chromatin accessibility and expression of the genes required for Tr1 cell function. IRF1 and BATF deficiencies uniquely altered the chromatin landscape, suggesting that these factors serve a pioneering function during Tr1 cell differentiation.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/immunology , Chromatin/metabolism , Interferon Regulatory Factor-1/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmunity , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Chromatin/genetics , Cluster Analysis , Cytokines/metabolism , Cytokines/pharmacology , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Interferon Regulatory Factor-1/genetics , Mice , Mice, Knockout , Promoter Regions, Genetic , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , Transcription Factors/metabolism , Transcriptome
5.
Immunity ; 51(1): 185-197.e6, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31278058

ABSTRACT

Innate lymphoid cells (ILCs) promote tissue homeostasis and immune defense but also contribute to inflammatory diseases. ILCs exhibit phenotypic and functional plasticity in response to environmental stimuli, yet the transcriptional regulatory networks (TRNs) that control ILC function are largely unknown. Here, we integrate gene expression and chromatin accessibility data to infer regulatory interactions between transcription factors (TFs) and genes within intestinal type 1, 2, and 3 ILC subsets. We predicted the "core" TFs driving ILC identities, organized TFs into cooperative modules controlling distinct gene programs, and validated roles for c-MAF and BCL6 as regulators affecting type 1 and type 3 ILC lineages. The ILC network revealed alternative-lineage-gene repression, a mechanism that may contribute to reported plasticity between ILC subsets. By connecting TFs to genes, the TRNs suggest means to selectively regulate ILC effector functions, while our network approach is broadly applicable to identifying regulators in other in vivo cell populations.


Subject(s)
Intestines/physiology , Lymphocyte Subsets/physiology , Lymphocytes/physiology , Animals , Cell Differentiation , Cell Lineage , Cell Plasticity , Chromatin Assembly and Disassembly , Epigenetic Repression , Gene Regulatory Networks , Immunity, Innate , Immunomodulation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-maf/genetics , Transcriptome
6.
Nat Methods ; 21(8): 1514-1524, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38744917

ABSTRACT

AlphaFold2 revolutionized structural biology with the ability to predict protein structures with exceptionally high accuracy. Its implementation, however, lacks the code and data required to train new models. These are necessary to (1) tackle new tasks, like protein-ligand complex structure prediction, (2) investigate the process by which the model learns and (3) assess the model's capacity to generalize to unseen regions of fold space. Here we report OpenFold, a fast, memory efficient and trainable implementation of AlphaFold2. We train OpenFold from scratch, matching the accuracy of AlphaFold2. Having established parity, we find that OpenFold is remarkably robust at generalizing even when the size and diversity of its training set is deliberately limited, including near-complete elisions of classes of secondary structure elements. By analyzing intermediate structures produced during training, we also gain insights into the hierarchical manner in which OpenFold learns to fold. In sum, our studies demonstrate the power and utility of OpenFold, which we believe will prove to be a crucial resource for the protein modeling community.


Subject(s)
Models, Molecular , Protein Folding , Proteins , Proteins/chemistry , Computational Biology/methods , Software , Protein Conformation , Algorithms , Protein Structure, Secondary
7.
Cell ; 151(2): 289-303, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23021777

ABSTRACT

Th17 cells have critical roles in mucosal defense and are major contributors to inflammatory disease. Their differentiation requires the nuclear hormone receptor RORγt working with multiple other essential transcription factors (TFs). We have used an iterative systems approach, combining genome-wide TF occupancy, expression profiling of TF mutants, and expression time series to delineate the Th17 global transcriptional regulatory network. We find that cooperatively bound BATF and IRF4 contribute to initial chromatin accessibility and, with STAT3, initiate a transcriptional program that is then globally tuned by the lineage-specifying TF RORγt, which plays a focal deterministic role at key loci. Integration of multiple data sets allowed inference of an accurate predictive model that we computationally and experimentally validated, identifying multiple new Th17 regulators, including Fosl2, a key determinant of cellular plasticity. This interconnected network can be used to investigate new therapeutic approaches to manipulate Th17 functions in the setting of inflammatory disease.


Subject(s)
Gene Regulatory Networks , Th17 Cells/cytology , Th17 Cells/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/immunology , Fos-Related Antigen-2/immunology , Fos-Related Antigen-2/metabolism , Genome-Wide Association Study , Humans , Interferon Regulatory Factors/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Th17 Cells/immunology
8.
Nature ; 597(7878): 693-697, 2021 09.
Article in English | MEDLINE | ID: mdl-34552240

ABSTRACT

One of the hallmarks of the cerebral cortex is the extreme diversity of interneurons1-3. The two largest subtypes of cortical interneurons, parvalbumin- and somatostatin-positive cells, are morphologically and functionally distinct in adulthood but arise from common lineages within the medial ganglionic eminence4-11. This makes them an attractive model for studying the generation of cell diversity. Here we examine how developmental changes in transcription and chromatin structure enable these cells to acquire distinct identities in the mouse cortex. Generic interneuron features are first detected upon cell cycle exit through the opening of chromatin at distal elements. By constructing cell-type-specific gene regulatory networks, we observed that parvalbumin- and somatostatin-positive cells initiate distinct programs upon settling within the cortex. We used these networks to model the differential transcriptional requirement of a shared regulator, Mef2c, and confirmed the accuracy of our predictions through experimental loss-of-function experiments. We therefore reveal how a common molecular program diverges to enable these neuronal subtypes to acquire highly specialized properties by adulthood. Our methods provide a framework for examining the emergence of cellular diversity, as well as for quantifying and predicting the effect of candidate genes on cell-type-specific development.


Subject(s)
Cerebral Cortex/cytology , Epigenesis, Genetic , Gene Regulatory Networks , Interneurons/cytology , Neurogenesis , Animals , Cell Differentiation , Cell Movement , Female , MEF2 Transcription Factors/genetics , Male , Mice , Mice, Knockout , Parvalbumins/metabolism , RNA-Seq , Single-Cell Analysis , Somatostatin/metabolism
10.
Nature ; 566(7744): E7, 2019 02.
Article in English | MEDLINE | ID: mdl-30723268

ABSTRACT

In this Letter, the 'Competing interests' statement should have stated: 'D.R.L. consults for and has equity in Vedanta Biosciences.' The original Letter has not been corrected.

11.
J Allergy Clin Immunol ; 153(4): 954-968, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38295882

ABSTRACT

Studies of asthma and allergy are generating increasing volumes of omics data for analysis and interpretation. The National Institute of Allergy and Infectious Diseases (NIAID) assembled a workshop comprising investigators studying asthma and allergic diseases using omics approaches, omics investigators from outside the field, and NIAID medical and scientific officers to discuss the following areas in asthma and allergy research: genomics, epigenomics, transcriptomics, microbiomics, metabolomics, proteomics, lipidomics, integrative omics, systems biology, and causal inference. Current states of the art, present challenges, novel and emerging strategies, and priorities for progress were presented and discussed for each area. This workshop report summarizes the major points and conclusions from this NIAID workshop. As a group, the investigators underscored the imperatives for rigorous analytic frameworks, integration of different omics data types, cross-disciplinary interaction, strategies for overcoming current limitations, and the overarching goal to improve scientific understanding and care of asthma and allergic diseases.


Subject(s)
Asthma , Hypersensitivity , United States , Humans , National Institute of Allergy and Infectious Diseases (U.S.) , Hypersensitivity/genetics , Asthma/etiology , Genomics , Proteomics , Metabolomics
12.
Bioinformatics ; 39(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36477794

ABSTRACT

MOTIVATION: T cells use T cell receptors (TCRs) to recognize small parts of antigens, called epitopes, presented by major histocompatibility complexes. Once an epitope is recognized, an immune response is initiated and T cell activation and proliferation by clonal expansion begin. Clonal populations of T cells with identical TCRs can remain in the body for years, thus forming immunological memory and potentially mappable immunological signatures, which could have implications in clinical applications including infectious diseases, autoimmunity and tumor immunology. RESULTS: We introduce TCRconv, a deep learning model for predicting recognition between TCRs and epitopes. TCRconv uses a deep protein language model and convolutions to extract contextualized motifs and provides state-of-the-art TCR-epitope prediction accuracy. Using TCR repertoires from COVID-19 patients, we demonstrate that TCRconv can provide insight into T cell dynamics and phenotypes during the disease. AVAILABILITY AND IMPLEMENTATION: TCRconv is available at https://github.com/emmijokinen/tcrconv. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
COVID-19 , Humans , Epitopes , Receptors, Antigen, T-Cell , T-Lymphocytes , Antigens , Epitopes, T-Lymphocyte
13.
Biomacromolecules ; 25(1): 258-271, 2024 01 08.
Article in English | MEDLINE | ID: mdl-38110299

ABSTRACT

Protein hydrogels represent an important and growing biomaterial for a multitude of applications, including diagnostics and drug delivery. We have previously explored the ability to engineer the thermoresponsive supramolecular assembly of coiled-coil proteins into hydrogels with varying gelation properties, where we have defined important parameters in the coiled-coil hydrogel design. Using Rosetta energy scores and Poisson-Boltzmann electrostatic energies, we iterate a computational design strategy to predict the gelation of coiled-coil proteins while simultaneously exploring five new coiled-coil protein hydrogel sequences. Provided this library, we explore the impact of in silico energies on structure and gelation kinetics, where we also reveal a range of blue autofluorescence that enables hydrogel disassembly and recovery. As a result of this library, we identify the new coiled-coil hydrogel sequence, Q5, capable of gelation within 24 h at 4 °C, a more than 2-fold increase over that of our previous iteration Q2. The fast gelation time of Q5 enables the assessment of structural transition in real time using small-angle X-ray scattering (SAXS) that is correlated to coarse-grained and atomistic molecular dynamics simulations revealing the supramolecular assembling behavior of coiled-coils toward nanofiber assembly and gelation. This work represents the first system of hydrogels with predictable self-assembly, autofluorescent capability, and a molecular model of coiled-coil fiber formation.


Subject(s)
Molecular Dynamics Simulation , Proteins , Scattering, Small Angle , X-Ray Diffraction , Proteins/chemistry , Hydrogels
14.
Nature ; 554(7692): 373-377, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29414937

ABSTRACT

Both microbial and host genetic factors contribute to the pathogenesis of autoimmune diseases. There is accumulating evidence that microbial species that potentiate chronic inflammation, as in inflammatory bowel disease, often also colonize healthy individuals. These microorganisms, including the Helicobacter species, can induce pathogenic T cells and are collectively referred to as pathobionts. However, how such T cells are constrained in healthy individuals is not yet understood. Here we report that host tolerance to a potentially pathogenic bacterium, Helicobacter hepaticus, is mediated by the induction of RORγt+FOXP3+ regulatory T (iTreg) cells that selectively restrain pro-inflammatory T helper 17 (TH17) cells and whose function is dependent on the transcription factor c-MAF. Whereas colonization of wild-type mice by H. hepaticus promoted differentiation of RORγt-expressing microorganism-specific iTreg cells in the large intestine, in disease-susceptible IL-10-deficient mice, there was instead expansion of colitogenic TH17 cells. Inactivation of c-MAF in the Treg cell compartment impaired differentiation and function, including IL-10 production, of bacteria-specific iTreg cells, and resulted in the accumulation of H. hepaticus-specific inflammatory TH17 cells and spontaneous colitis. By contrast, RORγt inactivation in Treg cells had only a minor effect on the bacteria-specific Treg and TH17 cell balance, and did not result in inflammation. Our results suggest that pathobiont-dependent inflammatory bowel disease is driven by microbiota-reactive T cells that have escaped this c-MAF-dependent mechanism of iTreg-TH17 homeostasis.


Subject(s)
Colitis/immunology , Colitis/microbiology , Helicobacter hepaticus/immunology , Immune Tolerance , Intestines/immunology , Intestines/microbiology , Proto-Oncogene Proteins c-maf/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Bioengineering , Colitis/pathology , Female , Forkhead Transcription Factors/metabolism , Helicobacter hepaticus/genetics , Helicobacter hepaticus/pathogenicity , Homeostasis , Host-Pathogen Interactions , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Interleukin-10/immunology , Male , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proto-Oncogene Proteins c-maf/deficiency , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/cytology , Th17 Cells/immunology
15.
Nature ; 562(7725): 150, 2018 10.
Article in English | MEDLINE | ID: mdl-29973715

ABSTRACT

Change History: This Article has been retracted; see accompanying Retraction. Corrected online 20 January: In this Article, author Frank Rigo was incorrectly listed with a middle initial; this has been corrected in the online versions of the paper.

16.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34131075

ABSTRACT

Despite the belief that social media is altering intergroup dynamics-bringing people closer or further alienating them from one another-the impact of social media on interethnic attitudes has yet to be rigorously evaluated, especially within areas with tenuous interethnic relations. We report results from a randomized controlled trial in Bosnia and Herzegovina (BiH), exploring the effects of exposure to social media during 1 wk around genocide remembrance in July 2019 on a set of interethnic attitudes of Facebook users. We find evidence that, counter to preregistered expectations, people who deactivated their Facebook profiles report lower regard for ethnic outgroups than those who remained active. Moreover, we present additional evidence suggesting that this effect is likely conditional on the level of ethnic heterogeneity of respondents' residence. We also extend the analysis to include measures of subjective well-being and knowledge of news. Here, we find that Facebook deactivation leads to suggestive improvements in subjective wellbeing and a decrease in knowledge of current events, replicating results from recent research in the United States in a very different context, thus increasing our confidence in the generalizability of these effects.


Subject(s)
Ethnicity , Social Media , Attitude , Genocide , Health , Humans , Intention to Treat Analysis , Knowledge , Surveys and Questionnaires
17.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Article in English | MEDLINE | ID: mdl-33723038

ABSTRACT

The rise of antibiotic resistance calls for new therapeutics targeting resistance factors such as the New Delhi metallo-ß-lactamase 1 (NDM-1), a bacterial enzyme that degrades ß-lactam antibiotics. We present structure-guided computational methods for designing peptide macrocycles built from mixtures of l- and d-amino acids that are able to bind to and inhibit targets of therapeutic interest. Our methods explicitly consider the propensity of a peptide to favor a binding-competent conformation, which we found to predict rank order of experimentally observed IC50 values across seven designed NDM-1- inhibiting peptides. We were able to determine X-ray crystal structures of three of the designed inhibitors in complex with NDM-1, and in all three the conformation of the peptide is very close to the computationally designed model. In two of the three structures, the binding mode with NDM-1 is also very similar to the design model, while in the third, we observed an alternative binding mode likely arising from internal symmetry in the shape of the design combined with flexibility of the target. Although challenges remain in robustly predicting target backbone changes, binding mode, and the effects of mutations on binding affinity, our methods for designing ordered, binding-competent macrocycles should have broad applicability to a wide range of therapeutic targets.


Subject(s)
Drug Design , Models, Molecular , Peptides/chemistry , Peptides/pharmacology , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/chemistry , Binding Sites , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Molecular Conformation , Molecular Docking Simulation , Molecular Structure , Protein Binding , Structure-Activity Relationship
18.
Bioinformatics ; 38(20): 4762-4770, 2022 10 14.
Article in English | MEDLINE | ID: mdl-35997560

ABSTRACT

MOTIVATION: Machine learning models for predicting cell-type-specific transcription factor (TF) binding sites have become increasingly more accurate thanks to the increased availability of next-generation sequencing data and more standardized model evaluation criteria. However, knowledge transfer from data-rich to data-limited TFs and cell types remains crucial for improving TF binding prediction models because available binding labels are highly skewed towards a small collection of TFs and cell types. Transfer prediction of TF binding sites can potentially benefit from a multitask learning approach; however, existing methods typically use shallow single-task models to generate low-resolution predictions. Here, we propose NetTIME, a multitask learning framework for predicting cell-type-specific TF binding sites with base-pair resolution. RESULTS: We show that the multitask learning strategy for TF binding prediction is more efficient than the single-task approach due to the increased data availability. NetTIME trains high-dimensional embedding vectors to distinguish TF and cell-type identities. We show that this approach is critical for the success of the multitask learning strategy and allows our model to make accurate transfer predictions within and beyond the training panels of TFs and cell types. We additionally train a linear-chain conditional random field (CRF) to classify binding predictions and show that this CRF eliminates the need for setting a probability threshold and reduces classification noise. We compare our method's predictive performance with two state-of-the-art methods, Catchitt and Leopard, and show that our method outperforms previous methods under both supervised and transfer learning settings. AVAILABILITY AND IMPLEMENTATION: NetTIME is freely available at https://github.com/ryi06/NetTIME and the code is also archived at https://doi.org/10.5281/zenodo.6994897. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
High-Throughput Nucleotide Sequencing , Transcription Factors , Base Pairing , Binding Sites , High-Throughput Nucleotide Sequencing/methods , Protein Binding , Transcription Factors/metabolism
19.
Bioinformatics ; 38(9): 2519-2528, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35188184

ABSTRACT

MOTIVATION: Gene regulatory networks define regulatory relationships between transcription factors and target genes within a biological system, and reconstructing them is essential for understanding cellular growth and function. Methods for inferring and reconstructing networks from genomics data have evolved rapidly over the last decade in response to advances in sequencing technology and machine learning. The scale of data collection has increased dramatically; the largest genome-wide gene expression datasets have grown from thousands of measurements to millions of single cells, and new technologies are on the horizon to increase to tens of millions of cells and above. RESULTS: In this work, we present the Inferelator 3.0, which has been significantly updated to integrate data from distinct cell types to learn context-specific regulatory networks and aggregate them into a shared regulatory network, while retaining the functionality of the previous versions. The Inferelator is able to integrate the largest single-cell datasets and learn cell-type-specific gene regulatory networks. Compared to other network inference methods, the Inferelator learns new and informative Saccharomyces cerevisiae networks from single-cell gene expression data, measured by recovery of a known gold standard. We demonstrate its scaling capabilities by learning networks for multiple distinct neuronal and glial cell types in the developing Mus musculus brain at E18 from a large (1.3 million) single-cell gene expression dataset with paired single-cell chromatin accessibility data. AVAILABILITY AND IMPLEMENTATION: The inferelator software is available on GitHub (https://github.com/flatironinstitute/inferelator) under the MIT license and has been released as python packages with associated documentation (https://inferelator.readthedocs.io/). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Gene Regulatory Networks , Software , Animals , Mice , Genomics , Genome , Chromatin
20.
Ann Rheum Dis ; 82(4): 507-514, 2023 04.
Article in English | MEDLINE | ID: mdl-36600182

ABSTRACT

OBJECTIVES: To investigate the cutaneous microbiome spanning the entire psoriatic disease spectrum, and to evaluate distinguishing features of psoriasis (PsO) and psoriatic arthritis (PsA). METHODS: Skin swabs were collected from upper and lower extremities of healthy individuals and patients with PsO and PsA. Psoriatic patients contributed both lesional (L) and contralateral non-lesional (NL) samples. Microbiota were analysed using 16S rRNA sequencing. RESULTS: Compared with healthy skin, alpha diversity in psoriatic NL and L skin was significantly reduced (p<0.05) and samples clustered separately in plots of beta diversity (p<0.05). Kocuria and Cutibacterium were enriched in healthy subjects, while Staphylococcus was enriched in psoriatic disease. Microbe-microbe association networks revealed a higher degree of similarity between psoriatic NL and L skin compared with healthy skin despite the absence of clinically evident inflammation. Moreover, the relative abundance of Corynebacterium was higher in NL PsA samples compared with NL PsO samples (p<0.05), potentially serving as a biomarker for disease progression. CONCLUSIONS: These findings show differences in diversity, bacterial composition and microbe-microbe interactions between healthy and psoriatic skin, both L and NL. We further identified bacterial biomarkers that differentiate disease phenotypes, which could potentially aid in predicting the transition from PsO to PsA.


Subject(s)
Arthritis, Psoriatic , Microbiota , Psoriasis , Humans , Arthritis, Psoriatic/microbiology , RNA, Ribosomal, 16S/genetics , Skin , Bacteria , Biomarkers
SELECTION OF CITATIONS
SEARCH DETAIL