Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.169
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 427-53, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38360547

ABSTRACT

The role of the autoimmune regulator (Aire) in central immune tolerance and thymic self-representation was first described more than 20 years ago, but fascinating new insights into its biology continue to emerge, particularly in the era of advanced single-cell genomics. We briefly describe the role of human genetics in the discovery of Aire, as well as insights into its function gained from genotype-phenotype correlations and the spectrum of Aire-associated autoimmunity-including insights from patients with Aire mutations with broad and diverse implications for human health. We then highlight emerging trends in Aire biology, focusing on three topic areas. First, we discuss medullary thymic epithelial diversity and the role of Aire in thymic epithelial development. Second, we highlight recent developments regarding the molecular mechanisms of Aire and its binding partners. Finally, we describe the rapidly evolving biology of the identity and function of extrathymic Aire-expressing cells (eTACs), and a novel eTAC subset called Janus cells, as well as their potential roles in immune homeostasis.


Subject(s)
AIRE Protein , Autoimmunity , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Animals , Thymus Gland/immunology , Thymus Gland/metabolism , Mutation , Immune Tolerance , Epithelial Cells/metabolism , Epithelial Cells/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism
2.
Annu Rev Immunol ; 40: 75-94, 2022 04 26.
Article in English | MEDLINE | ID: mdl-34985929

ABSTRACT

Strong epidemiological evidence now exists that sex is an important biologic variable in immunity. Recent studies, for example, have revealed that sex differences are associated with the severity of symptoms and mortality due to coronavirus disease 2019 (COVID-19). Despite this evidence, much remains to be learned about the mechanisms underlying associations between sex differences and immune-mediated conditions. A growing body of experimental data has made significant inroads into understanding sex-influenced immune responses. As physicians seek to provide more targeted patient care, it is critical to understand how sex-defining factors (e.g., chromosomes, gonadal hormones) alter immune responses in health and disease. In this review, we highlight recent insights into sex differences in autoimmunity; virus infection, specifically severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection; and cancer immunotherapy. A deeper understanding of underlying mechanisms will allow the development of a sex-based approach to disease screening and treatment.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Female , Humans , Male , Sex Characteristics , Sex Factors
3.
Annu Rev Immunol ; 39: 791-817, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33902311

ABSTRACT

Programmed cell death (PCD) is a requisite feature of development and homeostasis but can also be indicative of infections, injuries, and pathologies. In concordance with these heterogeneous contexts, an array of disparate effector responses occur downstream of cell death and its clearance-spanning tissue morphogenesis, homeostatic turnover, host defense, active dampening of inflammation, and tissue repair. This raises a fundamental question of how a single contextually appropriate response ensues after an event of PCD. To explore how complex inputs may together tailor the specificity of the resulting effector response, here we consider (a) the varying contexts during which different cell death modalities are observed, (b) the nature of the information that can be passed on by cell corpses, and (c) the ways by which efferocyte populations synthesize signals from dying cells with those from the surrounding microenvironment.


Subject(s)
Apoptosis , Animals , Cell Death , Homeostasis , Humans
4.
Annu Rev Immunol ; 39: 537-556, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33577346

ABSTRACT

The IL-17 family is an evolutionarily old cytokine family consisting of six members (IL-17A through IL-17F). IL-17 family cytokines signal through heterodimeric receptors that include the shared IL-17RA subunit, which is widely expressed throughout the body on both hematopoietic and nonhematopoietic cells. The founding family member, IL-17A, is usually referred to as IL-17 and has received the most attention for proinflammatory roles in autoimmune diseases like psoriasis. However, IL-17 is associated with a wide array of diseases with perhaps surprisingly variable pathologies. This review focuses on recent advances in the roles of IL-17 during health and in disease pathogenesis. To decipher the functions of IL-17 in diverse disease processes it is useful to first consider the physiological functions that IL-17 contributes to health. We then discuss how these beneficial functions can be diverted toward pathogenic amplification of deleterious pathways driving chronic disease.


Subject(s)
Autoimmune Diseases , Interleukin-17 , Animals , Autoimmune Diseases/etiology , Cytokines , Humans , Intention , Receptors, Interleukin-17
5.
Annu Rev Immunol ; 39: 511-536, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33577348

ABSTRACT

The surfaces of all living organisms and most secreted proteins share a common feature: They are glycosylated. As the outermost-facing molecules, glycans participate in nearly all immunological processes, including driving host-pathogen interactions, immunological recognition and activation, and differentiation between self and nonself through a complex array of pathways and mechanisms. These fundamental immunologic roles are further cast into sharp relief in inflammatory, autoimmune, and cancer disease states in which immune regulation goes awry. Here, we review the broad impact of glycans on the immune system and discuss the changes and clinical opportunities associated with the onset of immunologic disease.


Subject(s)
Host-Pathogen Interactions , Polysaccharides , Animals , Cell Differentiation , Humans
6.
Annu Rev Immunol ; 38: 759-784, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340572

ABSTRACT

The signaling lipid sphingosine 1-phosphate (S1P) plays critical roles in an immune response. Drugs targeting S1P signaling have been remarkably successful in treatment of multiple sclerosis, and they have shown promise in clinical trials for colitis and psoriasis. One mechanism of these drugs is to block lymphocyte exit from lymph nodes, where lymphocytes are initially activated, into circulation, from which lymphocytes can reach sites of inflammation. Indeed, S1P can be considered a circulation marker, signaling to immune cells to help them find blood and lymphatic vessels, and to endothelial cells to stabilize the vasculature. That said, S1P plays pleiotropic roles in the immune response, and it will be important to build an integrated view of how S1P shapes inflammation. S1P can function so effectively because its distribution is exquisitely tightly controlled. Here we review how S1P gradients regulate immune cell exit from tissues, with particular attention to key outstanding questions in the field.


Subject(s)
Cell Movement/immunology , Immune System/immunology , Immune System/metabolism , Lysophospholipids/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Biomarkers , Humans , Immune System/cytology , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Sphingosine/metabolism
7.
Annu Rev Immunol ; 38: 249-287, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340579

ABSTRACT

Since the birth of biotechnology, hundreds of biotherapeutics have been developed and approved by the US Food and Drug Administration (FDA) for human use. These novel medicines not only bring significant benefit to patients but also represent precision tools to interrogate human disease biology. Accordingly, much has been learned from the successes and failures of hundreds of high-quality clinical trials. In this review, we discuss general and broadly applicable themes that have emerged from this collective experience. We base our discussion on insights gained from exploring some of the most important target classes, including interleukin-1 (IL-1), tumor necrosis factor α (TNF-α), IL-6, IL-12/23, IL-17, IL-4/13, IL-5, immunoglobulin E (IgE), integrins and B cells. We also describe current challenges and speculate about how emerging technological capabilities may enable the discovery and development of the next generation of biotherapeutics.


Subject(s)
Biological Products/pharmacology , Biological Products/therapeutic use , Biological Therapy , Drug Development , Animals , Biological Products/history , Biological Therapy/history , Biological Therapy/methods , Biotechnology/history , Biotechnology/methods , Clinical Trials as Topic , Drug Development/history , Drug Discovery/history , Drug Discovery/methods , Drug Evaluation, Preclinical , History, 20th Century , History, 21st Century , Humans
8.
Annu Rev Immunol ; 38: 315-340, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31986068

ABSTRACT

The age-associated B cell subset has been the focus of increasing interest over the last decade. These cells have a unique cell surface phenotype and transcriptional signature, and they rely on TLR7 or TLR9 signals in the context of Th1 cytokines for their formation and activation. Most are antigen-experienced memory B cells that arise during responses to microbial infections and are key to pathogen clearance and control. Their increasing prevalence with age contributes to several well-established features of immunosenescence, including reduced B cell genesis and damped immune responses. In addition, they are elevated in autoimmune and autoinflammatory diseases, and in these settings they are enriched for characteristic autoantibody specificities. Together, these features identify age-associated B cells as a subset with pivotal roles in immunological health, disease, and aging. Accordingly, a detailed understanding of their origins, functions, and physiology should make them tractable translational targets in each of these settings.


Subject(s)
Aging/physiology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Animals , Autoimmunity , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Biomarkers , Cytokines/metabolism , Disease Susceptibility , Homeostasis , Humans , Immunologic Memory , Immunosenescence , Lymphocyte Activation/immunology
9.
Annu Rev Immunol ; 38: 621-648, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32017656

ABSTRACT

Vitiligo is an autoimmune disease of the skin that targets pigment-producing melanocytes and results in patches of depigmentation that are visible as white spots. Recent research studies have yielded a strong mechanistic understanding of this disease. Autoreactive cytotoxic CD8+ T cells engage melanocytes and promote disease progression through the local production of IFN-γ, and IFN-γ-induced chemokines are then secreted from surrounding keratinocytes to further recruit T cells to the skin through a positive-feedback loop. Both topical and systemic treatments that block IFN-γ signaling can effectively reverse vitiligo in humans; however, disease relapse is common after stopping treatments. Autoreactive resident memory T cells are responsible for relapse, and new treatment strategies focus on eliminating these cells to promote long-lasting benefit. Here, we discuss basic, translational, and clinical research studies that provide insight into the pathogenesis of vitiligo, and how this insight has been utilized to create new targeted treatment strategies.


Subject(s)
Vitiligo/etiology , Vitiligo/therapy , Animals , Autoimmunity , Biomarkers , Cytokines/metabolism , Disease Management , Disease Susceptibility , Humans , Immunologic Memory , Vitiligo/diagnosis
10.
Annu Rev Immunol ; 37: 439-456, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31026415

ABSTRACT

Monocytes are innate blood cells that maintain vascular homeostasis and are early responders to pathogens in acute infections. There are three well-characterized classes of monocytes: classical (CD14+CD16- in humans and Ly6Chi in mice), intermediate (CD14+CD16+ in humans and Ly6C+Treml4+ in mice), and nonclassical (CD14-CD16+ in humans and Ly6Clo in mice). Classical monocytes are critical for the initial inflammatory response. Classical monocytes can differentiate into macrophages in tissue and can contribute to chronic disease. Nonclassical monocytes have been widely viewed as anti-inflammatory, as they maintain vascular homeostasis. They are a first line of defense in recognition and clearance of pathogens. However, their roles in chronic disease are less clear. They have been shown to be protective as well as positively associated with disease burden. This review focuses on the state of the monocyte biology field and the functions of monocytes, particularly nonclassical monocytes, in health and disease.


Subject(s)
Arthritis, Rheumatoid/immunology , Atherosclerosis/immunology , Blood Vessels/physiology , Monocytes/immunology , Myocardial Infarction/immunology , Animals , Autoimmunity , Hematopoiesis , Homeostasis , Humans , Inflammation , Mice
11.
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
12.
Annu Rev Immunol ; 37: 599-624, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31026411

ABSTRACT

The intestinal microbiota plays a crucial role in influencing the development of host immunity, and in turn the immune system also acts to regulate the microbiota through intestinal barrier maintenance and immune exclusion. Normally, these interactions are homeostatic, tightly controlled, and organized by both innate and adaptive immune responses. However, a combination of environmental exposures and genetic defects can result in a break in tolerance and intestinal homeostasis. The outcomes of these interactions at the mucosal interface have broad, systemic effects on host immunity and the development of chronic inflammatory or autoimmune disease. The underlying mechanisms and pathways the microbiota can utilize to regulate these diseases are just starting to emerge. Here, we discuss the recent evidence in this area describing the impact of microbiota-immune interactions during inflammation and autoimmunity, with a focus on barrier function and CD4+ T cell regulation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/microbiology , Gastrointestinal Microbiome/immunology , Inflammation/microbiology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/microbiology , Animals , Autoimmunity , Diabetes Mellitus, Type 1/immunology , Homeostasis , Humans , Immune Tolerance , Immunomodulation , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology
13.
Annu Rev Immunol ; 36: 309-338, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677470

ABSTRACT

The complement system is an evolutionarily ancient key component of innate immunity required for the detection and removal of invading pathogens. It was discovered more than 100 years ago and was originally defined as a liver-derived, blood-circulating sentinel system that classically mediates the opsonization and lytic killing of dangerous microbes and the initiation of the general inflammatory reaction. More recently, complement has also emerged as a critical player in adaptive immunity via its ability to instruct both B and T cell responses. In particular, work on the impact of complement on T cell responses led to the surprising discoveries that the complement system also functions within cells and is involved in regulating basic cellular processes, predominantly those of metabolic nature. Here, we review current knowledge about complement's role in T cell biology, with a focus on the novel intracellular and noncanonical activities of this ancient system.


Subject(s)
Complement System Proteins/immunology , Immunomodulation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adaptive Immunity , Animals , Autoimmunity , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Complement Activation/immunology , Energy Metabolism , Host-Pathogen Interactions/immunology , Humans , Immunity, Cellular , Membrane Cofactor Protein/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism
14.
Annu Rev Immunol ; 36: 127-156, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29237129

ABSTRACT

T cells possess an array of functional capabilities important for host defense against pathogens and tumors. T cell effector functions require the T cell antigen receptor (TCR). The TCR has no intrinsic enzymatic activity, and thus signal transduction from the receptor relies on additional signaling molecules. One such molecule is the cytoplasmic tyrosine kinase ZAP-70, which associates with the TCR complex and is required for initiating the canonical biochemical signal pathways downstream of the TCR. In this article, we describe recent structure-based insights into the regulation and substrate specificity of ZAP-70, and then we review novel methods for determining the role of ZAP-70 catalytic activity-dependent and -independent signals in developing and mature T cells. Lastly, we discuss the disease states in mouse models and humans, which range from immunodeficiency to autoimmunity, that are caused by mutations in ZAP-70.


Subject(s)
Disease Susceptibility , Signal Transduction , T-Lymphocytes/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism , Animals , Autoimmunity , Biomarkers , Catalysis , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression Regulation , Humans , Immunity , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Phosphorylation , Protein Transport , Structure-Activity Relationship , Substrate Specificity , T-Lymphocytes/immunology , ZAP-70 Protein-Tyrosine Kinase/antagonists & inhibitors , ZAP-70 Protein-Tyrosine Kinase/chemistry , ZAP-70 Protein-Tyrosine Kinase/genetics
15.
Annu Rev Immunol ; 35: 337-370, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28142321

ABSTRACT

Transcriptomics, the high-throughput characterization of RNAs, has been instrumental in defining pathogenic signatures in human autoimmunity and autoinflammation. It enabled the identification of new therapeutic targets in IFN-, IL-1- and IL-17-mediated diseases. Applied to immunomonitoring, transcriptomics is starting to unravel diagnostic and prognostic signatures that stratify patients, track molecular changes associated with disease activity, define personalized treatment strategies, and generally inform clinical practice. Herein, we review the use of transcriptomics to define mechanistic, diagnostic, and predictive signatures in human autoimmunity and autoinflammation. We discuss some of the analytical approaches applied to extract biological knowledge from high-dimensional data sets. Finally, we touch upon emerging applications of transcriptomics to study eQTLs, B and T cell repertoire diversity, and isoform usage.


Subject(s)
Autoimmune Diseases/diagnosis , Inflammation/diagnosis , Transcriptome , Autoimmune Diseases/immunology , Datasets as Topic , High-Throughput Nucleotide Sequencing , Humans , Inflammation/immunology , Information Storage and Retrieval , Molecular Targeted Therapy , Monitoring, Immunologic , Prognosis
16.
Cell ; 187(9): 2079-2094, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38670066

ABSTRACT

Several conceptual pillars form the foundation of modern immunology, including the clonal selection theory, antigen receptor diversity, immune memory, and innate control of adaptive immunity. However, some immunological phenomena cannot be explained by the current framework. Thus, we still do not know how to design vaccines that would provide long-lasting protective immunity against certain pathogens, why autoimmune responses target some antigens and not others, or why the immune response to infection sometimes does more harm than good. Understanding some of these mysteries may require that we question existing assumptions to develop and test alternative explanations. Immunology is increasingly at a point when, once again, exploring new perspectives becomes a necessity.


Subject(s)
Allergy and Immunology , Humans , Animals , Allergy and Immunology/trends , Adaptive Immunity , Immunity, Innate , Immunologic Memory
17.
Cell ; 187(3): 733-749.e16, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38306984

ABSTRACT

Autoimmune diseases disproportionately affect females more than males. The XX sex chromosome complement is strongly associated with susceptibility to autoimmunity. Xist long non-coding RNA (lncRNA) is expressed only in females to randomly inactivate one of the two X chromosomes to achieve gene dosage compensation. Here, we show that the Xist ribonucleoprotein (RNP) complex comprising numerous autoantigenic components is an important driver of sex-biased autoimmunity. Inducible transgenic expression of a non-silencing form of Xist in male mice introduced Xist RNP complexes and sufficed to produce autoantibodies. Male SJL/J mice expressing transgenic Xist developed more severe multi-organ pathology in a pristane-induced lupus model than wild-type males. Xist expression in males reprogrammed T and B cell populations and chromatin states to more resemble wild-type females. Human patients with autoimmune diseases displayed significant autoantibodies to multiple components of XIST RNP. Thus, a sex-specific lncRNA scaffolds ubiquitous RNP components to drive sex-biased immunity.


Subject(s)
Autoantibodies , Autoimmune Diseases , RNA, Long Noncoding , Animals , Female , Humans , Male , Mice , Autoantibodies/genetics , Autoimmune Diseases/genetics , Autoimmunity/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , X Chromosome/genetics , X Chromosome/metabolism , X Chromosome Inactivation , Sex Characteristics
18.
Annu Rev Immunol ; 34: 265-97, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26907214

ABSTRACT

MHC class II (MHC-II) molecules are critical in the control of many immune responses. They are also involved in most autoimmune diseases and other pathologies. Here, we describe the biology of MHC-II and MHC-II variations that affect immune responses. We discuss the classic cell biology of MHC-II and various perturbations. Proteolysis is a major process in the biology of MHC-II, and we describe the various components forming and controlling this endosomal proteolytic machinery. This process ultimately determines the MHC-II-presented peptidome, including cryptic peptides, modified peptides, and other peptides that are relevant in autoimmune responses. MHC-II also variable in expression, glycosylation, and turnover. We illustrate that MHC-II is variable not only in amino acids (polymorphic) but also in its biology, with consequences for both health and disease.


Subject(s)
Antigen Presentation , Antigens/metabolism , Endosomes/metabolism , Histocompatibility Antigens Class II/metabolism , Immune System Diseases/immunology , Animals , Antigens/immunology , Autoimmunity , Endocytosis , Gene Expression Regulation , Glycosylation , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Peptide Fragments/immunology , Polymorphism, Genetic , Protein Transport , Proteolysis
19.
Cell ; 186(6): 1144-1161.e18, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36868219

ABSTRACT

Germinal centers (GCs) that form within lymphoid follicles during antibody responses are sites of massive cell death. Tingible body macrophages (TBMs) are tasked with apoptotic cell clearance to prevent secondary necrosis and autoimmune activation by intracellular self antigens. We show by multiple redundant and complementary methods that TBMs derive from a lymph node-resident, CD169-lineage, CSF1R-blockade-resistant precursor that is prepositioned in the follicle. Non-migratory TBMs use cytoplasmic processes to chase and capture migrating dead cell fragments using a "lazy" search strategy. Follicular macrophages activated by the presence of nearby apoptotic cells can mature into TBMs in the absence of GCs. Single-cell transcriptomics identified a TBM cell cluster in immunized lymph nodes which upregulated genes involved in apoptotic cell clearance. Thus, apoptotic B cells in early GCs trigger activation and maturation of follicular macrophages into classical TBMs to clear apoptotic debris and prevent antibody-mediated autoimmune diseases.


Subject(s)
Germinal Center , Lymph Nodes , Macrophages , Apoptosis , B-Lymphocytes , Lymph Nodes/cytology , Macrophages/cytology , Macrophages/metabolism
20.
Cell ; 186(7): 1309-1327, 2023 03 30.
Article in English | MEDLINE | ID: mdl-37001498

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of the central nervous system afflicting nearly three million individuals worldwide. Neuroimmune interactions between glial, neural, and immune cells play important roles in MS pathology and offer potential targets for therapeutic intervention. Here, we review underlying risk factors, mechanisms of MS pathogenesis, available disease modifying therapies, and examine the value of emerging technologies, which may address unmet clinical needs and identify novel therapeutic targets.


Subject(s)
Multiple Sclerosis , Humans , Multiple Sclerosis/drug therapy , Central Nervous System , Neuroglia , Cell Physiological Phenomena , Inflammation/pathology
SELECTION OF CITATIONS
SEARCH DETAIL