Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164.215
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
Annu Rev Immunol ; 38: 785-808, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32126183

ABSTRACT

Primary atopic disorders describes a series of monogenic diseases that have allergy- or atopic effector-related symptoms as a substantial feature. The underlying pathogenic genetic lesions help illustrate fundamental pathways in atopy, opening up diagnostic and therapeutic options for further study in those patients, but ultimately for common allergic diseases as well. Key pathways affected in these disorders include T cell receptor and B cell receptor signaling, cytokine signaling, skin barrier function, and mast cell function, as well as pathways that have not yet been elucidated. While comorbidities such as classically syndromic presentation or immune deficiency are often present, in some cases allergy alone is the presenting symptom, suggesting that commonly encountered allergic diseases exist on a spectrum of monogenic and complex genetic etiologies that are impacted by environmental risk factors.


Subject(s)
Disease Susceptibility , Hypersensitivity, Immediate/etiology , Hypersensitivity, Immediate/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Cytokines/metabolism , Disease Management , Environment , Genetic Predisposition to Disease , Humans , Hypersensitivity, Immediate/diagnosis , Mast Cells/immunology , Mast Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
Annu Rev Immunol ; 37: 571-597, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30698999

ABSTRACT

CRISPR technology has opened a new era of genome interrogation and genome engineering. Discovered in bacteria, where it protects against bacteriophage by cleaving foreign nucleic acid sequences, the CRISPR system has been repurposed as an adaptable tool for genome editing and multiple other applications. CRISPR's ease of use, precision, and versatility have led to its widespread adoption, accelerating biomedical research and discovery in human cells and model organisms. Here we review CRISPR-based tools and discuss how they are being applied to decode the genetic circuits that control immune function in health and disease. Genetic variation in immune cells can affect autoimmune disease risk, infectious disease pathogenesis, and cancer immunotherapies. CRISPR provides unprecedented opportunities for functional mechanistic studies of coding and noncoding genome sequence function in immunity. Finally, we discuss the potential of CRISPR technology to engineer synthetic cellular immunotherapies for a wide range of human diseases.


Subject(s)
Autoimmune Diseases/immunology , Cell- and Tissue-Based Therapy/methods , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Infections/immunology , Neoplasms/immunology , Animals , Autoimmune Diseases/genetics , CRISPR-Cas Systems , Gene Editing , Genetic Predisposition to Disease , Genetic Variation , Humans , Immunity , Infections/genetics , Neoplasms/genetics
3.
Annu Rev Immunol ; 36: 519-548, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29394121

ABSTRACT

Natural killer (NK) cells have vital functions in human immunity and reproduction. In the innate and adaptive immune responses to infection, particularly by viruses, NK cells respond by secreting inflammatory cytokines and killing infected cells. In reproduction, NK cells are critical for genesis of the placenta, the organ that controls the supply of oxygen and nutrients to the growing fetus. Controlling NK cell functions are interactions of HLA class I with inhibitory NK cell receptors. First evolved was the conserved interaction of HLA-E with CD94:NKG2A; later established were diverse interactions of HLA-A, -B, and -C with killer cell immunoglobulin-like receptors. Characterizing the latter interactions is rapid evolution, which distinguishes human populations and all species of higher primate. Driving this evolution are the different and competing selections imposed by pathogens on NK cell-mediated immunity and by the constraints of human reproduction on NK cell-mediated placentation. Promoting rapid evolution is independent segregation of polymorphic receptors and ligands throughout human populations.


Subject(s)
Genetic Predisposition to Disease , Immunity , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Biological Evolution , Genetic Loci , Genomics/methods , Haplotypes , Humans , Major Histocompatibility Complex/genetics , Receptors, KIR/genetics , Receptors, KIR/metabolism
4.
Annu Rev Immunol ; 36: 755-781, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677472

ABSTRACT

Inflammatory bowel disease (IBD) defines a spectrum of complex disorders. Understanding how environmental risk factors, alterations of the intestinal microbiota, and polygenetic and epigenetic susceptibility impact on immune pathways is key for developing targeted therapies. Mechanistic understanding of polygenic IBD is complemented by Mendelian disorders that present with IBD, pharmacological interventions that cause colitis, autoimmunity, and multiple animal models. Collectively, this multifactorial pathogenesis supports a concept of immune checkpoints that control microbial-host interactions in the gut by modulating innate and adaptive immunity, as well as epithelial and mesenchymal cell responses. In addition to classical immunosuppressive strategies, we discuss how resetting the microbiota and restoring innate immune responses, in particular autophagy and epithelial barrier function, might be key for maintaining remission or preventing IBD. Targeting checkpoints in genetically stratified subgroups of patients with Mendelian disorder-associated IBD increasingly directs treatment strategies as part of personalized medicine.


Subject(s)
Disease Susceptibility/immunology , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/therapy , Animals , Biomarkers , Chronic Disease , Disease Management , Disease Models, Animal , Drug-Related Side Effects and Adverse Reactions , Dysbiosis , Gastrointestinal Microbiome , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/prevention & control , Molecular Targeted Therapy , Translational Research, Biomedical
5.
Annu Rev Immunol ; 35: 1-30, 2017 04 26.
Article in English | MEDLINE | ID: mdl-27912315

ABSTRACT

Genome technologies have defined a complex genetic architecture in major infectious, inflammatory, and autoimmune disorders. High density marker arrays and Immunochips have powered genome-wide association studies (GWAS) that have mapped nearly 450 genetic risk loci in 22 major inflammatory diseases, including a core of common genes that play a central role in pathological inflammation. Whole-exome and whole-genome sequencing have identified more than 265 genes in which mutations cause primary immunodeficiencies and rare forms of severe inflammatory bowel disease. Combined analysis of inflammatory disease GWAS and primary immunodeficiencies point to shared proteins and pathways that are required for immune cell development and protection against infections and are also associated with pathological inflammation. Finally, sequencing of chromatin immunoprecipitates containing specific transcription factors, with parallel RNA sequencing, has charted epigenetic regulation of gene expression by proinflammatory transcription factors in immune cells, providing complementary information to characterize morbid genes at infectious and inflammatory disease loci.


Subject(s)
Autoimmune Diseases/genetics , Immunologic Deficiency Syndromes/genetics , Infections/genetics , Inflammation/genetics , Vaccines/immunology , Animals , Epigenesis, Genetic , Exome/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Immunity/genetics , Infections/immunology , Risk
6.
Cell ; 187(2): 464-480.e10, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38242088

ABSTRACT

Primary open-angle glaucoma (POAG), the leading cause of irreversible blindness worldwide, disproportionately affects individuals of African ancestry. We conducted a genome-wide association study (GWAS) for POAG in 11,275 individuals of African ancestry (6,003 cases; 5,272 controls). We detected 46 risk loci associated with POAG at genome-wide significance. Replication and post-GWAS analyses, including functionally informed fine-mapping, multiple trait co-localization, and in silico validation, implicated two previously undescribed variants (rs1666698 mapping to DBF4P2; rs34957764 mapping to ROCK1P1) and one previously associated variant (rs11824032 mapping to ARHGEF12) as likely causal. For individuals of African ancestry, a polygenic risk score (PRS) for POAG from our mega-analysis (African ancestry individuals) outperformed a PRS from summary statistics of a much larger GWAS derived from European ancestry individuals. This study quantifies the genetic architecture similarities and differences between African and non-African ancestry populations for this blinding disease.


Subject(s)
Genome-Wide Association Study , Glaucoma, Open-Angle , Humans , Genetic Predisposition to Disease , Glaucoma, Open-Angle/genetics , Black People/genetics , Polymorphism, Single Nucleotide/genetics
7.
Cell ; 187(2): 273-275, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38242084

ABSTRACT

Although the blinding eye disease glaucoma is more common in people of African ancestry, previous genetic studies predominantly involved European subjects. In this issue of Cell, O'Brien et al. report a genome-wide association study for glaucoma in individuals of African ancestry, showing overlap with European studies and refining an African polygenic risk score.


Subject(s)
Genome-Wide Association Study , Glaucoma , Humans , Glaucoma/genetics , Black People/genetics , Research , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide
8.
Annu Rev Immunol ; 34: 31-64, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27168239

ABSTRACT

Inflammatory bowel disease (IBD), including Crohn disease and ulcerative colitis, is characterized by chronic intestinal inflammation due to a complex interaction of genetic determinants, disruption of mucosal barriers, aberrant inflammatory signals, loss of tolerance, and environmental triggers. Importantly, the incidence of pediatric IBD is rising, particularly in children younger than 10 years. In this review, we discuss the clinical presentation of these patients and highlight environmental exposures that may affect disease risk, particularly among people with a background genetic risk. With regard to both children and adults, we review advancements in understanding the intestinal epithelium, the mucosal immune system, and the resident microbiota, describing how dysfunction at any level can lead to diseases like IBD. We conclude with future directions for applying advances in IBD genetics to better understand pathogenesis and develop therapeutics targeting key pathogenic nodes.


Subject(s)
Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Immunity, Mucosal , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Adult , Animals , Child , Child, Preschool , Environmental Exposure/adverse effects , Gene-Environment Interaction , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/therapy , Molecular Targeted Therapy
9.
Cell ; 186(21): 4514-4527.e14, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37757828

ABSTRACT

Autozygosity is associated with rare Mendelian disorders and clinically relevant quantitative traits. We investigated associations between the fraction of the genome in runs of homozygosity (FROH) and common diseases in Genes & Health (n = 23,978 British South Asians), UK Biobank (n = 397,184), and 23andMe. We show that restricting analysis to offspring of first cousins is an effective way of reducing confounding due to social/environmental correlates of FROH. Within this group in G&H+UK Biobank, we found experiment-wide significant associations between FROH and twelve common diseases. We replicated associations with type 2 diabetes (T2D) and post-traumatic stress disorder via within-sibling analysis in 23andMe (median n = 480,282). We estimated that autozygosity due to consanguinity accounts for 5%-18% of T2D cases among British Pakistanis. Our work highlights the possibility of widespread non-additive genetic effects on common diseases and has important implications for global populations with high rates of consanguinity.


Subject(s)
Consanguinity , Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/genetics , Homozygote , Phenotype , Polymorphism, Single Nucleotide , Biological Specimen Banks , Genome, Human , Genetic Predisposition to Disease , United Kingdom
10.
Cell ; 186(23): 5165-5182.e33, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37852259

ABSTRACT

Schizophrenia (SCZ) is a highly heritable mental disorder with thousands of associated genetic variants located mostly in the noncoding space of the genome. Translating these associations into insights regarding the underlying pathomechanisms has been challenging because the causal variants, their mechanisms of action, and their target genes remain largely unknown. We implemented a massively parallel variant annotation pipeline (MVAP) to perform SCZ variant-to-function mapping at scale in disease-relevant neural cell types. This approach identified 620 functional variants (1.7%) that operate in a highly developmental context and neuronal-activity-dependent manner. Multimodal integration of epigenomic and CRISPRi screening data enabled us to link these functional variants to target genes, biological processes, and ultimately alterations of neuronal physiology. These results provide a multistage prioritization strategy to map functional single-nucleotide polymorphism (SNP)-to-gene-to-endophenotype relations and offer biological insights into the context-dependent molecular processes modulated by SCZ-associated genetic variation.


Subject(s)
Schizophrenia , Humans , Genetic Predisposition to Disease , Genome-Wide Association Study , Neurons/metabolism , Polymorphism, Single Nucleotide/genetics , Schizophrenia/genetics , Animals , Mice , High-Throughput Nucleotide Sequencing
11.
Annu Rev Immunol ; 33: 823-74, 2015.
Article in English | MEDLINE | ID: mdl-25706096

ABSTRACT

Patients with autoinflammatory diseases present with noninfectious fever flares and systemic and/or disease-specific organ inflammation. Their excessive proinflammatory cytokine and chemokine responses can be life threatening and lead to organ damage over time. Studying such patients has revealed genetic defects that have helped unravel key innate immune pathways, including excessive IL-1 signaling, constitutive NF-κB activation, and, more recently, chronic type I IFN signaling. Discoveries of monogenic defects that lead to activation of proinflammatory cytokines have inspired the use of anticytokine-directed treatment approaches that have been life changing for many patients and have led to the approval of IL-1-blocking agents for a number of autoinflammatory conditions. In this review, we describe the genetically characterized autoinflammatory diseases, we summarize our understanding of the molecular pathways that drive clinical phenotypes and that continue to inspire the search for novel treatment targets, and we provide a conceptual framework for classification.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Genetic Predisposition to Disease , Inflammation/genetics , Inflammation/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmunity , Disease Models, Animal , Humans , Immunity, Innate , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Inflammation/metabolism , Inflammation Mediators/metabolism , Interferons/metabolism , Interleukin-1/metabolism , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , NF-kappa B/metabolism , Signal Transduction
12.
Cell ; 185(22): 4216-4232.e16, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36240780

ABSTRACT

Genotype-phenotype associations for common diseases are often compounded by pleiotropy and metabolic state. Here, we devised a pooled human organoid-panel of steatohepatitis to investigate the impact of metabolic status on genotype-phenotype association. En masse population-based phenotypic analysis under insulin insensitive conditions predicted key non-alcoholic steatohepatitis (NASH)-genetic factors including the glucokinase regulatory protein (GCKR)-rs1260326:C>T. Analysis of NASH clinical cohorts revealed that GCKR-rs1260326-T allele elevates disease severity only under diabetic state but protects from fibrosis under non-diabetic states. Transcriptomic, metabolomic, and pharmacological analyses indicate significant mitochondrial dysfunction incurred by GCKR-rs1260326, which was not reversed with metformin. Uncoupling oxidative mechanisms mitigated mitochondrial dysfunction and permitted adaptation to increased fatty acid supply while protecting against oxidant stress, forming a basis for future therapeutic approaches for diabetic NASH. Thus, "in-a-dish" genotype-phenotype association strategies disentangle the opposing roles of metabolic-associated gene variant functions and offer a rich mechanistic, diagnostic, and therapeutic inference toolbox toward precision hepatology. VIDEO ABSTRACT.


Subject(s)
Genetic Predisposition to Disease , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/genetics , Organoids , Genetic Association Studies , Alleles , Liver
13.
Cell ; 185(23): 4409-4427.e18, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36368308

ABSTRACT

Fully understanding autism spectrum disorder (ASD) genetics requires whole-genome sequencing (WGS). We present the latest release of the Autism Speaks MSSNG resource, which includes WGS data from 5,100 individuals with ASD and 6,212 non-ASD parents and siblings (total n = 11,312). Examining a wide variety of genetic variants in MSSNG and the Simons Simplex Collection (SSC; n = 9,205), we identified ASD-associated rare variants in 718/5,100 individuals with ASD from MSSNG (14.1%) and 350/2,419 from SSC (14.5%). Considering genomic architecture, 52% were nuclear sequence-level variants, 46% were nuclear structural variants (including copy-number variants, inversions, large insertions, uniparental isodisomies, and tandem repeat expansions), and 2% were mitochondrial variants. Our study provides a guidebook for exploring genotype-phenotype correlations in families who carry ASD-associated rare variants and serves as an entry point to the expanded studies required to dissect the etiology in the ∼85% of the ASD population that remain idiopathic.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Humans , Autism Spectrum Disorder/genetics , Genetic Predisposition to Disease , DNA Copy Number Variations/genetics , Genomics
14.
Nat Immunol ; 25(6): 969-980, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38831104

ABSTRACT

Rare genetic variants in toll-like receptor 7 (TLR7) are known to cause lupus in humans and mice. UNC93B1 is a transmembrane protein that regulates TLR7 localization into endosomes. In the present study, we identify two new variants in UNC93B1 (T314A, located proximally to the TLR7 transmembrane domain, and V117L) in a cohort of east Asian patients with childhood-onset systemic lupus erythematosus. The V117L variant was associated with increased expression of type I interferons and NF-κB-dependent cytokines in patient plasma and immortalized B cells. THP-1 cells expressing the variant UNC93B1 alleles exhibited exaggerated responses to stimulation of TLR7/-8, but not TLR3 or TLR9, which could be inhibited by targeting the downstream signaling molecules, IRAK1/-4. Heterozygous mice expressing the orthologous Unc93b1V117L variant developed a spontaneous lupus-like disease that was more severe in homozygotes and again hyperresponsive to TLR7 stimulation. Together, this work formally identifies genetic variants in UNC93B1 that can predispose to childhood-onset systemic lupus erythematosus.


Subject(s)
Genetic Predisposition to Disease , Lupus Erythematosus, Systemic , Toll-Like Receptor 7 , Lupus Erythematosus, Systemic/genetics , Humans , Animals , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Mice , Child , Female , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Male , Age of Onset , Genetic Variation , NF-kappa B/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Adolescent , THP-1 Cells , Interferon Type I/metabolism
15.
Annu Rev Immunol ; 32: 227-55, 2014.
Article in English | MEDLINE | ID: mdl-24655295

ABSTRACT

The skin is the front line of defense against insult and injury and contains many epidermal and immune elements that comprise the skin-associated lymphoid tissue (SALT). The reaction of these components to injury allows an effective cutaneous response to restore homeostasis. Psoriasis vulgaris is the best-understood and most accessible human disease that is mediated by T cells and dendritic cells. Inflammatory myeloid dendritic cells release IL-23 and IL-12 to activate IL-17-producing T cells, Th1 cells, and Th22 cells to produce abundant psoriatic cytokines IL-17, IFN-γ, TNF, and IL-22. These cytokines mediate effects on keratinocytes to amplify psoriatic inflammation. Therapeutic studies with anticytokine antibodies have shown the importance of the key cytokines IL-23, TNF, and IL-17 in this process. We discuss the genetic background of psoriasis and its relationship to immune function, specifically genetic mutations, key PSORS loci, single nucleotide polymorphisms, and the skin transcriptome. The association between comorbidities and psoriasis is reviewed by correlating the skin transcriptome and serum proteins. Psoriasis-related cytokine-response pathways are considered in the context of the transcriptome of different mouse models. This approach offers a model for other inflammatory skin and autoimmune diseases.


Subject(s)
Psoriasis/immunology , Animals , Comorbidity , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Mice , Psoriasis/diagnosis , Psoriasis/genetics , Skin/immunology , Skin/pathology , Skin Physiological Phenomena/immunology
16.
Annu Rev Immunol ; 32: 83-119, 2014.
Article in English | MEDLINE | ID: mdl-24364806

ABSTRACT

Inheritance of a coding variant of the protein tyrosine phosphatase nonreceptor type 22 (PTPN22) gene is associated with increased susceptibility to autoimmunity and infection. Efforts to elucidate the mechanisms by which the PTPN22-C1858T variant modulates disease risk revealed that PTPN22 performs a signaling function in multiple biochemical pathways and cell types. Capable of both enzymatic activity and adaptor functions, PTPN22 modulates signaling through antigen and innate immune receptors. PTPN22 plays roles in lymphocyte development and activation, establishment of tolerance, and innate immune cell-mediated host defense and immunoregulation. The disease-associated PTPN22-R620W variant protein is likely involved in multiple stages of the pathogenesis of autoimmunity. Establishment of a tolerant B cell repertoire is disrupted by PTPN22-R620W action during immature B cell selection, and PTPN22-R620W alters mature T cell responsiveness. However, after autoimmune attack has initiated tissue injury, PTPN22-R620W may foster inflammation through modulating the balance of myeloid cell-produced cytokines.


Subject(s)
Immunity/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Signal Transduction , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Genetic Predisposition to Disease , Humans , Immune System/physiology , Immune Tolerance , Immunomodulation , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics
17.
Annu Rev Immunol ; 32: 433-59, 2014.
Article in English | MEDLINE | ID: mdl-24499275

ABSTRACT

Although new activation and regulatory mechanisms are still being identified, the basic architecture of the complement system has been known for decades. Two major roles of complement are to control certain bacterial infections and to promote clearance of apoptotic cells. In addition, although inappropriate complement activation has long been proposed to cause tissue damage in human inflammatory and autoimmune diseases, whether this is indeed true has been uncertain. However, recent studies in humans, especially those using newly available biological therapeutics, have now clearly demonstrated the pathophysiologic importance of the complement system in several rare diseases. Beyond these conditions, recent genetic studies have strongly supported an injurious role for complement in a wide array of human inflammatory, degenerative, and autoimmune diseases. This review includes an overview of complement activation, regulatory, and effector mechanisms. It then focuses on new understandings gained from genetic studies, ex vivo analyses, therapeutic trials, and animal models as well as on new research opportunities.


Subject(s)
Complement System Proteins/immunology , Complement System Proteins/metabolism , Receptors, Complement/metabolism , Animals , Carrier Proteins/metabolism , Complement Activation/immunology , Complement System Proteins/genetics , Disease Models, Animal , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Humans , Protein Binding , Signal Transduction
18.
Cell ; 184(18): 4784-4818.e17, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34450027

ABSTRACT

Osteoarthritis affects over 300 million people worldwide. Here, we conduct a genome-wide association study meta-analysis across 826,690 individuals (177,517 with osteoarthritis) and identify 100 independently associated risk variants across 11 osteoarthritis phenotypes, 52 of which have not been associated with the disease before. We report thumb and spine osteoarthritis risk variants and identify differences in genetic effects between weight-bearing and non-weight-bearing joints. We identify sex-specific and early age-at-onset osteoarthritis risk loci. We integrate functional genomics data from primary patient tissues (including articular cartilage, subchondral bone, and osteophytic cartilage) and identify high-confidence effector genes. We provide evidence for genetic correlation with phenotypes related to pain, the main disease symptom, and identify likely causal genes linked to neuronal processes. Our results provide insights into key molecular players in disease processes and highlight attractive drug targets to accelerate translation.


Subject(s)
Genetic Predisposition to Disease , Genetics, Population , Osteoarthritis/genetics , Female , Genome-Wide Association Study , Humans , Osteoarthritis/drug therapy , Phenotype , Polymorphism, Single Nucleotide/genetics , Risk Factors , Sex Characteristics , Signal Transduction/genetics
19.
Cell ; 184(15): 4090-4104.e15, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34129837

ABSTRACT

The oral mucosa remains an understudied barrier tissue. This is a site of rich exposure to antigens and commensals, and a tissue susceptible to one of the most prevalent human inflammatory diseases, periodontitis. To aid in understanding tissue-specific pathophysiology, we compile a single-cell transcriptome atlas of human oral mucosa in healthy individuals and patients with periodontitis. We uncover the complex cellular landscape of oral mucosal tissues and identify epithelial and stromal cell populations with inflammatory signatures that promote antimicrobial defenses and neutrophil recruitment. Our findings link exaggerated stromal cell responsiveness with enhanced neutrophil and leukocyte infiltration in periodontitis. Our work provides a resource characterizing the role of tissue stroma in regulating mucosal tissue homeostasis and disease pathogenesis.


Subject(s)
Immunity, Mucosal , Mouth Mucosa/cytology , Mouth Mucosa/immunology , Neutrophils/cytology , Adult , Epithelial Cells/cytology , Gene Expression Regulation , Genetic Predisposition to Disease , Gingiva/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Microbiota , Myeloid Cells/cytology , Periodontitis/genetics , Periodontitis/immunology , Periodontitis/pathology , Single-Cell Analysis , Stromal Cells/cytology , T-Lymphocytes/cytology
20.
Cell ; 184(11): 3006-3021.e17, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33930287

ABSTRACT

Genetic studies have revealed many variant loci that are associated with immune-mediated diseases. To elucidate the disease pathogenesis, it is essential to understand the function of these variants, especially under disease-associated conditions. Here, we performed a large-scale immune cell gene-expression analysis, together with whole-genome sequence analysis. Our dataset consists of 28 distinct immune cell subsets from 337 patients diagnosed with 10 categories of immune-mediated diseases and 79 healthy volunteers. Our dataset captured distinctive gene-expression profiles across immune cell types and diseases. Expression quantitative trait loci (eQTL) analysis revealed dynamic variations of eQTL effects in the context of immunological conditions, as well as cell types. These cell-type-specific and context-dependent eQTLs showed significant enrichment in immune disease-associated genetic variants, and they implicated the disease-relevant cell types, genes, and environment. This atlas deepens our understanding of the immunogenetic functions of disease-associated variants under in vivo disease conditions.


Subject(s)
Gene Expression Regulation/genetics , Gene Expression/immunology , Immune System Diseases/genetics , Adult , Female , Gene Expression/genetics , Gene Expression Regulation/immunology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Humans , Immune System/cytology , Immune System/metabolism , Immune System Diseases/metabolism , Immune System Diseases/physiopathology , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Quantitative Trait Loci/immunology , Transcriptome/genetics , Whole Genome Sequencing/methods
SELECTION OF CITATIONS
SEARCH DETAIL