Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep Med ; 5(3): 101431, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38378002

ABSTRACT

Sulfasalazine is a prodrug known to be effective for the treatment of inflammatory bowel disease (IBD)-associated peripheral spondyloarthritis (pSpA), but the mechanistic role for the gut microbiome in regulating its clinical efficacy is not well understood. Here, treatment of 22 IBD-pSpA subjects with sulfasalazine identifies clinical responders with a gut microbiome enriched in Faecalibacterium prausnitzii and the capacity for butyrate production. Sulfapyridine promotes butyrate production and transcription of the butyrate synthesis gene but in F. prausnitzii in vitro, which is suppressed by excess folate. Sulfasalazine therapy enhances fecal butyrate production and limits colitis in wild-type and gnotobiotic mice colonized with responder, but not non-responder, microbiomes. F. prausnitzii is sufficient to restore sulfasalazine protection from colitis in gnotobiotic mice colonized with non-responder microbiomes. These findings reveal a mechanistic link between the efficacy of sulfasalazine therapy and the gut microbiome with the potential to guide diagnostic and therapeutic approaches for IBD-pSpA.


Subject(s)
Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Mice , Animals , Sulfasalazine/pharmacology , Sulfasalazine/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Treatment Outcome , Butyrates
2.
bioRxiv ; 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37808658

ABSTRACT

Endurance exercise is an important health modifier. We studied cell-type specific adaptations of human skeletal muscle to acute endurance exercise using single-nucleus (sn) multiome sequencing in human vastus lateralis samples collected before and 3.5 hours after 40 min exercise at 70% VO2max in four subjects, as well as in matched time of day samples from two supine resting circadian controls. High quality same-cell RNA-seq and ATAC-seq data were obtained from 37,154 nuclei comprising 14 cell types. Among muscle fiber types, both shared and fiber-type specific regulatory programs were identified. Single-cell circuit analysis identified distinct adaptations in fast, slow and intermediate fibers as well as LUM-expressing FAP cells, involving a total of 328 transcription factors (TFs) acting at altered accessibility sites regulating 2,025 genes. These data and circuit mapping provide single-cell insight into the processes underlying tissue and metabolic remodeling responses to exercise.

3.
Cell Rep ; 37(10): 110100, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34879281

ABSTRACT

Older age is a strong risk factor for several diseases, including cancer. The etiology and biology of age-associated differences among cancers are poorly understood. To address this knowledge gap, we aim to delineate differences in tumor molecular characteristics between younger and older patients across a variety of tumor types from The Cancer Genome Atlas. We show that these groups exhibit widespread molecular differences in select tumor types. Our work shows that tumors in younger individuals exhibit a dysregulated molecular aging phenotype and are associated with hallmarks of premature senescence. Additionally, we find that these tumors are enriched for driver gene mutations, resulting in homologous recombination defects. Lastly, we observe a trend toward decreased immune infiltration and function in older patients and find that, immunologically, young tumor tissue resembles aged healthy tissue. Taken together, we find that tumors from young individuals possess unique characteristics that may be leveraged for therapy.


Subject(s)
Aging/genetics , Biomarkers, Tumor/genetics , Genomics , Mutation , Neoplasms/genetics , Adult , Age Factors , Aged , Aged, 80 and over , Aging/immunology , Aging/pathology , Cell Proliferation/genetics , Cellular Senescence/genetics , DNA Mutational Analysis , Databases, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Precision Medicine , Signal Transduction , Tumor Microenvironment , Young Adult
4.
Am J Hum Genet ; 108(9): 1752-1764, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34363748

ABSTRACT

An individual's genetics can dramatically influence breast cancer (BC) risk. Although clinical measures for prevention do exist, non-invasive personalized measures for reducing BC risk are limited. Commonly used medications are a promising set of modifiable factors, but no previous study has explored whether a range of widely taken approved drugs modulate BC genetics. In this study, we describe a quantitative framework for exploring the interaction between the genetic susceptibility of BC and medication usage among UK Biobank women. We computed BC polygenic scores (PGSs) that summarize BC genetic risk and find that the PGS explains nearly three-times greater variation in disease risk within corticosteroid users compared to non-users. We map 35 genes significantly interacting with corticosteroid use (FDR < 0.1), highlighting the transcription factor NRF2 as a common regulator of gene-corticosteroid interactions in BC. Finally, we discover a regulatory variant strongly stratifying BC risk according to corticosteroid use. Within risk allele carriers, 18.2% of women taking corticosteroids developed BC, compared to 5.1% of the non-users (with an HR = 3.41 per-allele within corticosteroid users). In comparison, there are no differences in BC risk within the reference allele homozygotes. Overall, this work highlights the clinical relevance of gene-drug interactions in disease risk and provides a roadmap for repurposing biobanks in drug repositioning and precision medicine.


Subject(s)
Adrenal Cortex Hormones/adverse effects , Breast Neoplasms/genetics , Gene-Environment Interaction , Multifactorial Inheritance , NF-E2-Related Factor 2/genetics , Prescription Drugs/adverse effects , Alleles , Biological Specimen Banks , Breast Neoplasms/chemically induced , Breast Neoplasms/diagnosis , Breast Neoplasms/epidemiology , Female , Gene Expression , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Incidence , NF-E2-Related Factor 2/metabolism , Polymorphism, Single Nucleotide , Precision Medicine/methods , Risk Assessment , United Kingdom/epidemiology
5.
Am J Hum Genet ; 108(1): 49-67, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33326753

ABSTRACT

Although thousands of loci have been associated with human phenotypes, the role of gene-environment (GxE) interactions in determining individual risk of human diseases remains unclear. This is partly because of the severe erosion of statistical power resulting from the massive number of statistical tests required to detect such interactions. Here, we focus on improving the power of GxE tests by developing a statistical framework for assessing quantitative trait loci (QTLs) associated with the trait means and/or trait variances. When applying this framework to body mass index (BMI), we find that GxE discovery and replication rates are significantly higher when prioritizing genetic variants associated with the variance of the phenotype (vQTLs) compared to when assessing all genetic variants. Moreover, we find that vQTLs are enriched for associations with other non-BMI phenotypes having strong environmental influences, such as diabetes or ulcerative colitis. We show that GxE effects first identified in quantitative traits such as BMI can be used for GxE discovery in disease phenotypes such as diabetes. A clear conclusion is that strong GxE interactions mediate the genetic contribution to body weight and diabetes risk.


Subject(s)
Biological Variation, Population/genetics , Genome-Wide Association Study/methods , Gene-Environment Interaction , Genotype , Humans , Phenotype , Quantitative Trait Loci/genetics , Quantitative Trait, Heritable
6.
Nat Commun ; 11(1): 2213, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32371927

ABSTRACT

Despite infiltrating immune cells having an essential function in human disease and patients' responses to treatments, mechanisms influencing variability in infiltration patterns remain unclear. Here, using bulk RNA-seq data from 46 tissues in the Genotype-Tissue Expression project, we apply cell-type deconvolution algorithms to evaluate the immune landscape across the healthy human body. We discover that 49 of 189 infiltration-related phenotypes are associated with either age or sex (FDR < 0.1). Genetic analyses further show that 31 infiltration-related phenotypes have genome-wide significant associations (iQTLs) (P < 5.0 × 10-8), with a significant enrichment of same-tissue expression quantitative trait loci in suggested iQTLs (P < 10-5). Furthermore, we find an association between helper T cell content in thyroid tissue and a COMMD3/DNAJC1 regulatory variant (P = 7.5 × 10-10), which is associated with thyroiditis in other cohorts. Together, our results identify key factors influencing inter-individual variability of immune infiltration, to provide insights on potential therapeutic targets.


Subject(s)
Gene Expression Profiling/methods , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Immune System/metabolism , Polymorphism, Single Nucleotide , Quantitative Trait Loci/genetics , Adult , Algorithms , Female , Gene Regulatory Networks/genetics , Gene Regulatory Networks/immunology , Genotype , Humans , Immune System/cytology , Immune System/immunology , Male , Middle Aged , Phenotype , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Thyroid Gland/cytology , Thyroid Gland/immunology , Thyroid Gland/metabolism
7.
Immunity ; 49(6): 1077-1089.e5, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30552020

ABSTRACT

Inflammatory bowel disease (IBD) results from a dysregulated interaction between the microbiota and a genetically susceptible host. Genetic studies have linked TNFSF15 polymorphisms and its protein TNF-like ligand 1A (TL1A) with IBD, but the functional role of TL1A is not known. Here, we found that adherent IBD-associated microbiota induced TL1A release from CX3CR1+ mononuclear phagocytes (MNPs). Using cell-specific genetic deletion models, we identified an essential role for CX3CR1+MNP-derived TL1A in driving group 3 innate lymphoid cell (ILC3) production of interleukin-22 and mucosal healing during acute colitis. In contrast to this protective role in acute colitis, TL1A-dependent expression of co-stimulatory molecule OX40L in MHCII+ ILC3s during colitis led to co-stimulation of antigen-specific T cells that was required for chronic T cell colitis. These results identify a role for ILC3s in activating intestinal T cells and reveal a central role for TL1A in promoting ILC3 barrier immunity during colitis.


Subject(s)
Colitis/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Microbiota/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Adult , Aged , Animals , Colitis/genetics , Colitis/metabolism , Female , Humans , Immunity, Innate/genetics , Interleukins/genetics , Interleukins/immunology , Interleukins/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microbiota/physiology , Middle Aged , Phagocytes/cytology , Phagocytes/immunology , Phagocytes/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Young Adult , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL
...