Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters








Publication year range
1.
Neoplasia ; 58: 101071, 2024 Oct 13.
Article in English | MEDLINE | ID: mdl-39405604

ABSTRACT

Chemotherapy, although effective in treating cancer, can induce various cellular responses, including senescence and drug resistance. Here, we investigate the transcriptomic alterations induced by doxorubicin (DOX), a commonly used chemotherapeutic agent, in human colon cancer cells. Using single-cell RNA sequencing, we identified distinct cell populations and their transcriptional profiles following subtoxic DOX treatment, revealing cell clusters characterized by differential expression of genes involved in cell cycle regulation and interferon (IFN) signaling. DOX-persisting proliferating cells exhibited upregulation of genes reported to be linked to the unphosphorylated form of ISGF3 (U-ISGF3) transcription factor. Furthermore, we found that HSH2D, a poor prognostic marker, was highly upregulated in doxorubicin-surviving proliferative cells, and its expression was correlated with U-ISGF3-related genes. Analysis of transcription kinetics via mathematical modeling revealed that the number of mRNA molecules produced per transcriptional burst was increased for U-ISGF3-related genes. We also observed altered gene co-expression patterns of U-ISGF3-related genes and others upon DOX treatment, which potentially contributes to chemoresistance of DOX-surviving proliferative cells and may influence cancer cell fate after chemotherapy. Our findings highlight U-ISGF3-related genes and the JAK/STAT pathway as potential therapeutic targets for overcoming chemoresistance in colon cancer.

2.
Immunity ; 57(5): 1005-1018.e7, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38697116

ABSTRACT

Cytokine expression during T cell differentiation is a highly regulated process that involves long-range promoter-enhancer and CTCF-CTCF contacts at cytokine loci. Here, we investigated the impact of dynamic chromatin loop formation within the topologically associating domain (TAD) in regulating the expression of interferon gamma (IFN-γ) and interleukin-22 (IL-22); these cytokine loci are closely located in the genome and are associated with complex enhancer landscapes, which are selectively active in type 1 and type 3 lymphocytes. In situ Hi-C analyses revealed inducible TADs that insulated Ifng and Il22 enhancers during Th1 cell differentiation. Targeted deletion of a 17 bp boundary motif of these TADs imbalanced Th1- and Th17-associated immunity, both in vitro and in vivo, upon Toxoplasma gondii infection. In contrast, this boundary element was dispensable for cytokine regulation in natural killer cells. Our findings suggest that precise cytokine regulation relies on lineage- and developmental stage-specific interactions of 3D chromatin architectures and enhancer landscapes.


Subject(s)
CCCTC-Binding Factor , Cell Differentiation , Interferon-gamma , Interleukin-22 , Interleukins , Th1 Cells , Animals , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Th1 Cells/immunology , Mice , Cell Differentiation/immunology , Interferon-gamma/metabolism , Binding Sites , Interleukins/metabolism , Interleukins/genetics , Enhancer Elements, Genetic/genetics , Mice, Inbred C57BL , Chromatin/metabolism , Toxoplasmosis/immunology , Toxoplasmosis/parasitology , Toxoplasmosis/genetics , Gene Expression Regulation , Toxoplasma/immunology , Cytokines/metabolism , Cell Lineage , Th17 Cells/immunology
3.
Sci Immunol ; 9(91): eadg8691, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38241399

ABSTRACT

Allergic diseases are common, affecting more than 20% of the population. Genetic variants in the TGFß pathway are strongly associated with atopy. To interrogate the mechanisms underlying this association, we examined patients and mice with Loeys-Dietz syndrome (LDS) who harbor missense mutations in the kinase domain of TGFΒR1/2. We demonstrate that LDS mutations lead to reduced TGFß signaling and elevated total and allergen-specific IgE, despite the presence of wild-type T regulatory cells in a chimera model. Germinal center activity was enhanced in LDS and characterized by a selective increase in type 2 follicular helper T cells (TFH2). Expression of Pik3cg was increased in LDS TFH cells and associated with reduced levels of the transcriptional repressor SnoN. PI3Kγ/mTOR signaling in LDS naïve CD4+ T cells was elevated after T cell receptor cross-linking, and pharmacologic inhibition of PI3Kγ or mTOR prevented exaggerated TFH2 and antigen-specific IgE responses after oral antigen exposure in an adoptive transfer model. Naïve CD4+ T cells from nonsyndromic allergic individuals also displayed decreased TGFß signaling, suggesting that our mechanistic discoveries may be broadly relevant to allergic patients in general. Thus, TGFß plays a conserved, T cell-intrinsic, and nonredundant role in restraining TFH2 development via the PI3Kγ/mTOR pathway and thereby protects against allergic disease.


Subject(s)
Hypersensitivity , Transforming Growth Factor beta , Animals , Humans , Mice , Hypersensitivity/metabolism , Immunoglobulin E , Th2 Cells , TOR Serine-Threonine Kinases
4.
Nat Immunol ; 24(12): 2080-2090, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37957354

ABSTRACT

Aberrant differentiation of progenitor cells in the hematopoietic system is known to severely impact host immune responsiveness. Here we demonstrate that NOD1, a cytosolic innate sensor of bacterial peptidoglycan, also functions in murine hematopoietic cells as a major regulator of both the generation and differentiation of lymphoid progenitors as well as peripheral T lymphocyte homeostasis. We further show that NOD1 mediates these functions by facilitating STAT5 signaling downstream of hematopoietic cytokines. In steady-state, loss of NOD1 resulted in a modest but significant decrease in numbers of mature T, B and natural killer cells. During systemic protozoan infection this defect was markedly enhanced, leading to host mortality. Lack of functional NOD1 also impaired T cell-dependent anti-tumor immunity while preventing colitis. These findings reveal that, in addition to its classical role as a bacterial ligand receptor, NOD1 plays an important function in regulating adaptive immunity through interaction with a major host cytokine signaling pathway.


Subject(s)
Immunity, Innate , Lymphopoiesis , Animals , Mice , Colitis , Ligands , Signal Transduction
5.
Sci Immunol ; 7(77): eabl9467, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36427325

ABSTRACT

Activated lymphocytes adapt their metabolism to meet the energetic and biosynthetic demands imposed by rapid growth and proliferation. Common gamma chain (cγ) family cytokines are central to these processes, but the role of downstream signal transducer and activator of transcription 5 (STAT5) signaling, which is engaged by all cγ members, is poorly understood. Using genome-, transcriptome-, and metabolome-wide analyses, we demonstrate that STAT5 is a master regulator of energy and amino acid metabolism in CD4+ T helper cells. Mechanistically, STAT5 localizes to an array of enhancers and promoters for genes encoding essential enzymes and transporters, where it facilitates p300 recruitment and epigenetic remodeling. We also find that STAT5 licenses the activity of two other key metabolic regulators, the mTOR signaling pathway and the MYC transcription factor. Building on the latter, we present evidence for transcriptome-wide cooperation between STAT5 and MYC in both normal and transformed T cells. Together, our data provide a molecular framework for transcriptional programing of T cell metabolism downstream of cγ cytokines and highlight the JAK-STAT pathway in mediating cellular growth and proliferation.


Subject(s)
Janus Kinases , STAT5 Transcription Factor , STAT5 Transcription Factor/genetics , Signal Transduction , STAT Transcription Factors , T-Lymphocytes, Helper-Inducer , Cytokines
6.
Cell ; 185(21): 3857-3876, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36240739

ABSTRACT

The discovery of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway arose from investigations of how cells respond to interferons (IFNs), revealing a paradigm in cell signaling conserved from slime molds to mammals. These discoveries revealed mechanisms underlying rapid gene expression mediated by a wide variety of extracellular polypeptides including cytokines, interleukins, and related factors. This knowledge has provided numerous insights into human disease, from immune deficiencies to cancer, and was rapidly translated to new drugs for autoimmune, allergic, and infectious diseases, including COVID-19. Despite these advances, major challenges and opportunities remain.


Subject(s)
COVID-19 , Janus Kinases , Animals , Cytokines/metabolism , Humans , Interferons/metabolism , Janus Kinases/metabolism , Mammals/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction
7.
Biocontrol Sci ; 27(1): 9-19, 2022.
Article in English | MEDLINE | ID: mdl-35314562

ABSTRACT

The aim of this study was to evaluate bacterial decontamination of boiled young sardine by treatment with violet-blue light followed by cooling storage of the irradiated boiled sardine. Viable cell count in the samples was evaluated after irradiation with four types of violet-blue light-emitting diodes (LEDs; peak wavelength at 405, 412, 421 or 455 nm) and subsequent cooling storage for two days. LED (405 nm) exhibited bactericidal and growth suppression effects. The irradiation gave a 47% bactericidal rate in comparison with no irradiation samples (control) and the two-day storage suppressed the increase in cell counts to 24%, while the rate of increase was 545% for the control. Integrated viability (IV) based on growth delay analysis was estimated after irradiation of four isolates from boiled sardine with 405 nm light. The irradiation caused growth delay against all isolates, resulting in smaller IV values for three isolates compared to those viabilities estimated from colony forming units. Exposure (405 nm) at 432 J/cm2 fluence resulted in a decrease in water content, resulting in an increase in salinity of the samples. This study demonstrated the advantages of light emitting a narrow violet region as a non-thermal disinfection technology in the processing and storage of boiled sardines.


Subject(s)
Disinfection , Drug Contamination , Anti-Bacterial Agents
8.
Article in English | MEDLINE | ID: mdl-34001528

ABSTRACT

Not simply an attribute of the adaptive immune system, immunological memory can be viewed on multiple levels. Accordingly, the molecular basis of memory comprises multiple mechanisms. The advent of new sequencing technologies has greatly enhanced the understanding of gene regulation and lymphocyte specification, and improved measurement of chromatin states affords new insights into the epigenomic and transcriptomic programs that underlie memory. Beyond canonical genes, the involvement of long noncoding RNAs (lncRNAs) is becoming increasingly apparent, and it appears that there are more than two to three times as many lncRNAs as protein-coding genes. lncRNAs can directly interact with DNA, RNA, and proteins, and a single lncRNA can contain multiple modular domains and thus interact with different classes of molecules. Yet, most lncRNAs have not been tested for function, and even fewer knockout mice have been generated. It is therefore timely to consider new potential mechanisms that may contribute to immune memory.


Subject(s)
RNA, Long Noncoding , Animals , Chromatin , Epigenomics , Gene Expression Regulation , Lymphocytes , Mice , RNA, Long Noncoding/metabolism
9.
Immunity ; 54(3): 514-525.e6, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33657395

ABSTRACT

MicroRNAs are important regulators of immune responses. Here, we show miR-221 and miR-222 modulate the intestinal Th17 cell response. Expression of miR-221 and miR-222 was induced by proinflammatory cytokines and repressed by the cytokine TGF-ß. Molecular targets of miR-221 and miR-222 included Maf and Il23r, and loss of miR-221 and miR-222 expression shifted the transcriptomic spectrum of intestinal Th17 cells to a proinflammatory signature. Although the loss of miR-221 and miR-222 was tolerated for maintaining intestinal Th17 cell homeostasis in healthy mice, Th17 cells lacking miR-221 and miR-222 expanded more efficiently in response to IL-23. Both global and T cell-specific deletion of miR-221 and miR-222 rendered mice prone to mucosal barrier damage. Collectively, these findings demonstrate that miR-221 and miR-222 are an integral part of intestinal Th17 cell response that are induced after IL-23 stimulation to constrain the magnitude of proinflammatory response.


Subject(s)
Inflammation/immunology , Interleukin-23/metabolism , Intestinal Mucosa/immunology , MicroRNAs/genetics , Th17 Cells/immunology , Animals , Feedback, Physiological , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-maf/metabolism , Receptors, Interleukin/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
10.
Immunity ; 53(4): 745-758.e4, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33010223

ABSTRACT

Innate immune responses rely on rapid and precise gene regulation mediated by accessibility of regulatory regions to transcription factors (TFs). In natural killer (NK) cells and other innate lymphoid cells, competent enhancers are primed during lineage acquisition, and formation of de novo enhancers characterizes the acquisition of innate memory in activated NK cells and macrophages. Here, we investigated how primed and de novo enhancers coordinate to facilitate high-magnitude gene induction during acute activation. Epigenomic and transcriptomic analyses of regions near highly induced genes (HIGs) in NK cells both in vitro and in a model of Toxoplasma gondii infection revealed de novo chromatin accessibility and enhancer remodeling controlled by signal-regulated TFs STATs. Acute NK cell activation redeployed the lineage-determining TF T-bet to de novo enhancers, independent of DNA-sequence-specific motif recognition. Thus, acute stimulation reshapes enhancer function through the combinatorial usage and repurposing of both lineage-determining and signal-regulated TFs to ensure an effective response.


Subject(s)
Enhancer Elements, Genetic/genetics , Enhancer Elements, Genetic/immunology , Killer Cells, Natural/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Animals , Chromatin/genetics , Chromatin/immunology , Female , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Toxoplasma/immunology , Toxoplasmosis/genetics , Toxoplasmosis/immunology
11.
Cell ; 181(7): 1696-1696.e1, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32589961

ABSTRACT

The JAK-STAT pathway is an evolutionarily conserved signal transduction paradigm, providing mechanisms for rapid receptor-to-nucleus communication and transcription control. Discoveries in this field provided insights into primary immunodeficiencies, inherited autoimmune and autoinflammatory diseases, and hematologic and oncologic disorders, giving rise to a new class of drugs, JAK inhibitors (or Jakinibs).


Subject(s)
Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Animals , Cell Membrane/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Janus Kinases/genetics , Janus Kinases/physiology , STAT Transcription Factors/genetics , STAT Transcription Factors/physiology , Signal Transduction/physiology
12.
Mol Cell ; 75(6): 1229-1242.e5, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31377117

ABSTRACT

Interferon gamma (IFN-γ), critical for host defense and tumor surveillance, requires tight control of its expression. Multiple cis-regulatory elements exist around Ifng along with a non-coding transcript, Ifng-as1 (also termed NeST). Here, we describe two genetic models generated to dissect the molecular functions of this locus and its RNA product. DNA deletion within the Ifng-as1 locus disrupted chromatin organization of the extended Ifng locus, impaired Ifng response, and compromised host defense. Insertion of a polyA signal ablated the Ifng-as1 full-length transcript and impaired host defense, while allowing proper chromatin structure. Transient knockdown of Ifng-as1 also reduced IFN-γ production. In humans, discordant expression of IFNG and IFNG-AS1 was evident in memory T cells, with high expression of this long non-coding RNA (lncRNA) and low expression of the cytokine. These results establish Ifng-as1 as an important regulator of Ifng expression, as a DNA element and transcribed RNA, involved in dynamic and cell state-specific responses to infection.


Subject(s)
Gene Expression Regulation/immunology , Immunologic Memory , Infections/immunology , Interferon-gamma/immunology , RNA, Untranslated/immunology , T-Lymphocytes/immunology , Animals , Chromatin/genetics , Chromatin/immunology , Female , Gene Knockdown Techniques , Infections/genetics , Infections/pathology , Interferon-gamma/genetics , Mice , RNA, Untranslated/genetics , T-Lymphocytes/pathology
13.
Immunity ; 51(1): 131-140.e5, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31315031

ABSTRACT

Macrophages play an important role in structural cardiac remodeling and the transition to heart failure following myocardial infarction (MI). Previous research has focused on the impact of blood-derived monocytes on cardiac repair. Here we examined the contribution of resident cavity macrophages located in the pericardial space adjacent to the site of injury. We found that disruption of the pericardial cavity accelerated maladaptive post-MI cardiac remodeling. Gata6+ macrophages in mouse pericardial fluid contributed to the reparative immune response. Following experimental MI, these macrophages invaded the epicardium and lost Gata6 expression but continued to perform anti-fibrotic functions. Loss of this specialized macrophage population enhanced interstitial fibrosis after ischemic injury. Gata6+ macrophages were present in human pericardial fluid, supporting the notion that this reparative function is relevant in human disease. Our findings uncover an immune cardioprotective role for the pericardial tissue compartment and argue for the reevaluation of surgical procedures that remove the pericardium.


Subject(s)
Fibrosis/prevention & control , GATA6 Transcription Factor/metabolism , Heart/physiology , Macrophages/immunology , Myocardial Infarction/immunology , Myocardium/pathology , Pericardium/immunology , Animals , Cell Movement , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ventricular Remodeling
14.
Immunity ; 51(4): 682-695.e6, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31353223

ABSTRACT

Innate lymphocytes maintain tissue homeostasis at mucosal barriers, with group 2 innate lymphoid cells (ILC2s) producing type 2 cytokines and controlling helminth infection. While the molecular understanding of ILC2 responses has advanced, the complexity of microenvironmental factors impacting ILC2s is becoming increasingly apparent. Herein, we used single-cell analysis to explore the diversity of gene expression among lung lymphocytes during helminth infection. Following infection, we identified a subset of ILC2s that preferentially expressed Il5-encoding interleukin (IL)-5, together with Calca-encoding calcitonin gene-related peptide (CGRP) and its cognate receptor components. CGRP in concert with IL-33 and neuromedin U (NMU) supported IL-5 but constrained IL-13 expression and ILC2 proliferation. Without CGRP signaling, ILC2 responses and worm expulsion were enhanced. Collectively, these data point to CGRP as a context-dependent negative regulatory factor that shapes innate lymphocyte responses to alarmins and neuropeptides during type 2 innate immune responses.


Subject(s)
Inflammation/immunology , Lymphocytes/immunology , Nippostrongylus/physiology , Receptors, Calcitonin Gene-Related Peptide/metabolism , Strongylida Infections/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Immunity, Innate , Interleukin-33/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/metabolism , Receptors, Calcitonin Gene-Related Peptide/genetics , Single-Cell Analysis , Th2 Cells/immunology , Transplantation Chimera
15.
Immunity ; 50(1): 106-120.e10, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30650370

ABSTRACT

CD4+ T helper (Th) differentiation is regulated by diverse inputs, including the vitamin A metabolite retinoic acid (RA). RA acts through its receptor RARα to repress transcription of inflammatory cytokines, but is also essential for Th-mediated immunity, indicating complex effects of RA on Th specification and the outcome of the immune response. We examined the impact of RA on the genome-wide transcriptional response during Th differentiation to multiple subsets. RA effects were subset-selective and were most significant in Th9 cells. RA globally antagonized Th9-promoting transcription factors and inhibited Th9 differentiation. RA directly targeted the extended Il9 locus and broadly modified the Th9 epigenome through RARα. RA-RARα activity limited murine Th9-associated pulmonary inflammation, and human allergic inflammation was associated with reduced expression of RA target genes. Thus, repression of the Th9 program is a major function of RA-RARα signaling in Th differentiation, arguing for a role for RA in interleukin 9 (IL-9) related diseases.


Subject(s)
Hypersensitivity/immunology , Lung/physiology , Pneumonia/immunology , Retinoic Acid Receptor alpha/metabolism , T-Lymphocytes, Helper-Inducer/physiology , Animals , Epigenetic Repression , HEK293 Cells , Humans , Hypersensitivity/genetics , Interleukin-9/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/genetics , Retinoic Acid Receptor alpha/genetics , Signal Transduction , Transcription, Genetic , Tretinoin/metabolism
16.
J Leukoc Biol ; 104(3): 499-514, 2018 09.
Article in English | MEDLINE | ID: mdl-29999544

ABSTRACT

In this era, it is axiomatic that cytokines have critical roles in cellular development and differentiation, immune homeostasis, and host defense. Equally, dysregulation of cytokines is known to contribute to diverse inflammatory and immune-mediated disorders. In fact, the past 20 years have witnessed the rapid translation of basic discoveries in cytokine biology to multiple successful biological agents (mAbs and recombinant fusion proteins) that target cytokines. These targeted therapies have not only fundamentally changed the face of multiple immune-mediated diseases but have also unequivocally established the role of specific cytokines in human disease; cytokine biologists have many times over provided remarkable basic advances with direct clinical benefit. Numerous cytokines rely on the JAK-STAT pathway for signaling, and new, safe, and effective small molecule inhibitors have been developed for a range of disorders. In this review, we will briefly summarize basic discoveries in cytokine signaling and briefly comment on some major unresolved issues. We will review clinical data pertaining to the first generation of JAK inhibitors and their clinical indications, discuss additional opportunities for targeting this pathway, and lay out some of the challenges that lie ahead.


Subject(s)
Janus Kinase Inhibitors/pharmacology , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/physiology , Animals , Humans , Signal Transduction/drug effects , Translational Research, Biomedical
17.
Immunity ; 48(4): 614-615, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29669241

ABSTRACT

The molecular basis for T cell memory differentiation remains elusive. Wang et al. (2018) identify Runx3 as an initiating transcription factor that specifies regulatory regions required for cytotoxic T cell (CTL) memory differentiation early after TCR signaling and constrains the ability of T-bet to drive terminal effector generation.


Subject(s)
Chromatin , T-Lymphocytes, Cytotoxic , Cell Differentiation , Gene Expression Regulation , Memory , Transcription Factors
18.
Eur J Immunol ; 48(7): 1174-1180, 2018 07.
Article in English | MEDLINE | ID: mdl-29524223

ABSTRACT

Innate lymphoid cells (ILCs) producing IL-22 and/or IL-17, designated as ILC3, comprise a heterogeneous subset of cells involved in regulation of gut barrier homeostasis and inflammation. Exogenous environmental cues in conjunction with regulated expression of endogenous factors are key determinants of plasticity of ILC3 toward the type 1 fate. Herein, by using mouse models and transcriptomic approaches, we defined at the molecular level, initial events driving ILC3 expressing natural cytotoxicity receptors (NCR+ ILC3) to acquire type 1 features. We observed that NCR+ ILC3 exhibited high basal expression of the signal-dependent transcription factor STAT4 due to T-BET, leading to predisposed potential for the type 1 response. We found that the prototypical inducer of type 3 response, IL-23, played a predominant role over IL-12 by accessing STAT4 and preferentially inducing its phosphorylation in ILC3 expressing T-BET. The early effector program driven by IL-23 was characterized by the expression of IL-22, followed by a production of IFN-γ, which relies on STAT4, T-BET and required chromatin remodeling of the Ifng locus. Altogether, our findings shed light on a feed-forward mechanism involving STAT4 and T-BET that modulates the outcome of IL-23 signaling in ILC3.


Subject(s)
Interleukin-23/metabolism , Intestines/immunology , Lymphocytes/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Ly/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Homeostasis , Humans , Immunity, Mucosal , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Intestines/pathology , Mice , Natural Cytotoxicity Triggering Receptor 1/metabolism , Protein Binding , Signal Transduction , T-Box Domain Proteins/genetics , Th1 Cells/immunology , Interleukin-22
19.
Nat Rev Drug Discov ; 17(1): 78, 2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29282366

ABSTRACT

This corrects the article DOI: 10.1038/nrd.2017.201.

20.
Nat Rev Drug Discov ; 16(12): 843-862, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29104284

ABSTRACT

The discovery of cytokines as key drivers of immune-mediated diseases has spurred efforts to target their associated signalling pathways. Janus kinases (JAKs) are essential signalling mediators downstream of many pro-inflammatory cytokines, and small-molecule inhibitors of JAKs (jakinibs) have gained traction as safe and efficacious options for the treatment of inflammation-driven pathologies such as rheumatoid arthritis, psoriasis and inflammatory bowel disease. Building on the clinical success of first-generation jakinibs, second-generation compounds that claim to be more selective are currently undergoing development and proceeding to clinical trials. However, important questions remain about the advantages and limitations of improved JAK selectivity, optimal routes and dosing regimens and how best to identify patients who will benefit from jakinibs. This Review discusses the biology of jakinibs from a translational perspective, focusing on recent insights from clinical trials, the development of novel agents and the use of jakinibs in a spectrum of immune and inflammatory diseases.


Subject(s)
Immune System Diseases/drug therapy , Inflammation/drug therapy , Janus Kinase Inhibitors/therapeutic use , Animals , Cytokines/immunology , Drug Design , Humans , Immune System Diseases/physiopathology , Inflammation/physiopathology , Janus Kinase Inhibitors/adverse effects , Janus Kinase Inhibitors/pharmacology , Janus Kinases/drug effects , Janus Kinases/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL