Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Australas J Ageing ; 2024 May 21.
Article in English | MEDLINE | ID: mdl-38770595

ABSTRACT

OBJECTIVE: Older inpatients who fall are often frail, with multiple co-morbidities and polypharmacy. Although the causes of falls are multifactorial, sedating and delirium-inducing drugs increase that risk. The aims were to determine whether people who fell had a change in their sedative and anticholinergic medication burden during an admission compared to people who did not fall. A secondary aim was to determine the factors associated with change in drug burden. METHODS: A retrospective, observational, case-control study of inpatients who fell. Two hundred consecutive people who fell were compared with 200 randomly selected people who had not fallen. Demographics, functional ability, frailty and cognition were recorded. For each patient, their total medications and anticholinergic and sedative burden were calculated on admission and on discharge, using the drug burden index (DBI). RESULTS: People who fell were more dependent and cognitively impaired than people who did not fallen. People who fell had a higher DBI on admission, than people who had not fall (mean: .69 vs .43, respectively, p < .001) and discharge (.66 vs .38, p < .001). For both cohorts, the DBI decreased between admission and discharge (-.03 and -.05), but neither were clinically significant. Higher total medications and a higher number DBI medications on admission were both associated with greater DBI changes (p = .003 and <.001, respectively). However, the presence (or absence) of cognitive impairment, dependency, frailty and single vs multiple falls were not significantly associated with DBI changes. CONCLUSIONS: In older people, DBI medications and falls are both common and have serious consequences, yet this study was unable to demonstrate any clinically relevant reduction in average DBI either in people who fell or people who had not fallen during a hospital admission.

2.
bioRxiv ; 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37163060

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) cooperate with adaptive Th2 cells as key organizers of tissue type 2 immune responses, while a spectrum of innate and adaptive lymphocytes coordinate early type 3/17 immunity. Both type 2 and type 3/17 lymphocyte associated cytokines are linked to tissue fibrosis, but how their dynamic and spatial topographies may direct beneficial or pathologic organ remodelling is unclear. Here we used volumetric imaging in models of liver fibrosis, finding accumulation of periportal and fibrotic tract IL-5 + lymphocytes, predominantly ILC2s, in close proximity to expanded type 3/17 lymphocytes and IL-33 high niche fibroblasts. Ablation of IL-5 + lymphocytes worsened carbon tetrachloride-and bile duct ligation-induced liver fibrosis with increased niche IL-17A + type 3/17 lymphocytes, predominantly γδ T cells. In contrast, concurrent ablation of IL-5 + and IL-17A + lymphocytes reduced this progressive liver fibrosis, suggesting a cross-regulation of type 2 and type 3 lymphocytes at specialized fibroblast niches that tunes hepatic fibrosis.

3.
Annu Rev Pathol ; 18: 311-335, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36351364

ABSTRACT

Tuft cells are found in tissues with distinct stem cell compartments, tissue architecture, and luminal exposures but converge on a shared transcriptional program, including expression of taste transduction signaling pathways. Here, we summarize seminal and recent findings on tuft cells, focusing on major categories of function-instigation of type 2 cytokine responses, orchestration of antimicrobial responses, and emerging roles in tissue repair-and describe tuft cell-derived molecules used to affect these functional programs. We review what is known about the development of tuft cells from epithelial progenitors under homeostatic conditions and during disease. Finally, we discuss evidence that immature, or nascent, tuft cells with potential for diverse functions are driven toward dominant effector programs by tissue- or perturbation-specific contextual cues, which may result in heterogeneous mature tuft cell phenotypes both within and between tissues.


Subject(s)
Intestinal Mucosa , Signal Transduction , Humans , Cell Lineage , Intestinal Mucosa/metabolism , Stem Cells , Homeostasis , Epithelial Cells/metabolism
4.
Immunity ; 55(10): 1891-1908.e12, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36044899

ABSTRACT

Demodex mites are commensal parasites of hair follicles (HFs). Normally asymptomatic, inflammatory outgrowth of mites can accompany malnutrition, immune dysfunction, and aging, but mechanisms restricting Demodex outgrowth are not defined. Here, we show that control of mite HF colonization in mice required group 2 innate lymphoid cells (ILC2s), interleukin-13 (IL-13), and its receptor, IL-4Ra-IL-13Ra1. HF-associated ILC2s elaborated IL-13 that attenuated HFs and epithelial proliferation at anagen onset; in their absence, Demodex colonization led to increased epithelial proliferation and replacement of gene programs for repair by aberrant inflammation, leading to the loss of barrier function and HF exhaustion. Humans with rhinophymatous acne rosacea, an inflammatory condition associated with Demodex, had increased HF inflammation with decreased type 2 cytokines, consistent with the inverse relationship seen in mice. Our studies uncover a key role for skin ILC2s and IL-13, which comprise an immune checkpoint that sustains cutaneous integrity and restricts pathologic infestation by colonizing HF mites.


Subject(s)
Mite Infestations , Mites , Animals , Cytokines , Hair Follicle/pathology , Humans , Immunity, Innate , Inflammation , Interleukin-13 , Lymphocytes/pathology , Mice , Mite Infestations/complications , Mite Infestations/parasitology , Mite Infestations/pathology , Symbiosis
5.
Curr Opin Cell Biol ; 76: 102082, 2022 06.
Article in English | MEDLINE | ID: mdl-35468541

ABSTRACT

Tuft cells are sentinel chemosensory cells that monitor the lumen of hollow organs for noxious or infectious stimuli and respond with disease- and tissue-specific effectors. The discovery of critical tuft cell functions in intestinal type 2 immune responses and airway defense has sparked interest in the formation and function of this architecturally unique cell type. Recent advances in single-cell transcriptomics and computational biology allow for new insights into the genetics and environmental cues underlying tuft cell formation and maturation. Here, we summarize the most recent research on tuft cell development and function in various disease states and organ systems.


Subject(s)
Intestinal Mucosa , Cell Differentiation , Intestinal Mucosa/metabolism , Structure-Activity Relationship
6.
Sci Immunol ; 7(69): eabj1080, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35245089

ABSTRACT

Inflammation and dysfunction of the extrahepatic biliary tree are common causes of human pathology, including gallstones and cholangiocarcinoma. Despite this, we know little about the local regulation of biliary inflammation. Tuft cells, rare sensory epithelial cells, are particularly prevalent in the mucosa of the gallbladder and extrahepatic bile ducts. Here, we show that biliary tuft cells express a core genetic tuft cell program in addition to a tissue-specific gene signature and, in contrast to small intestinal tuft cells, decreased postnatally, coincident with maturation of bile acid production. Manipulation of enterohepatic bile acid recirculation revealed that tuft cell abundance is negatively regulated by bile acids, including in a model of obstructive cholestasis in which inflammatory infiltration of the biliary tree correlated with loss of tuft cells. Unexpectedly, tuft cell-deficient mice spontaneously displayed an increased gallbladder epithelial inflammatory gene signature accompanied by neutrophil infiltration that was modulated by the microbiome. We propose that biliary tuft cells function as bile acid-sensitive negative regulators of inflammation in biliary tissues and serve to limit inflammation under homeostatic conditions.


Subject(s)
Bile Acids and Salts , Biliary Tract , Animals , Epithelial Cells/physiology , Inflammation , Mice , Neutrophils
7.
Curr Protoc ; 1(7): e205, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34242484
8.
Curr Protoc ; 1(3): e77, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33740294

ABSTRACT

Recent findings position tuft cells as key mediators of intestinal immunity through their production of the cytokine interleukin (IL)-25 and activation of group 2 innate lymphoid cells (ILC2s). Though tuft cells are found in numerous epithelial tissues, their phenotype and function have been best characterized in the small intestine, where robust in vivo techniques have enabled the dissection of their cellular function, ontogeny, and key signaling pathways. We describe methods for the identification, quantification, and manipulation of tuft cells, focusing on analysis of ILC2s as a readout of tuft cell function. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Ex vivo analysis of small intestinal tuft cells and ILC2 by flow cytometry Alternate Protocol: Ex vivo analysis of small intestinal tuft cells and ILC2 by flow cytometry in the context of type 2 inflammation Basic Protocol 2: Ex vivo analysis of small intestinal tuft cells by imaging of intestinal Swiss roll Basic Protocol 3: Tuft-ILC2 circuit activation by oral gavage of adult Nippostrongylus brasiliensis worms Basic Protocol 4: Circuit activation by colonization with Tritrichomonas spp. Basic Protocol 5: Circuit activation by treatment with succinate in drinking water Basic Protocol 6: Circuit activation by treatment with recombinant IL-25.


Subject(s)
Immunity, Innate , Tritrichomonas , Animals , Intestine, Small , Lymphocytes , Nippostrongylus
9.
Methods Mol Biol ; 2111: 239-256, 2020.
Article in English | MEDLINE | ID: mdl-31933212

ABSTRACT

Ubiquitination is a crucial component of many immune processes. While ubiquitin-mediated degradation is essential to T cell activation via T cell receptor signaling, the specific E3 ligases and substrates involved are not well-understood. Here, we describe a strategy integrating RNA, protein, and posttranslational modification datasets to identify targets of ubiquitin-mediated degradation. When integrated, these assays can provide broad insight into how this posttranslational modification regulates protein function and influences T cell biology.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/metabolism , Ubiquitin-Protein Ligases/metabolism , Lymphocyte Activation , Proteolysis , Signal Transduction , Ubiquitin
10.
Nat Immunol ; 20(6): 747-755, 2019 06.
Article in English | MEDLINE | ID: mdl-31061531

ABSTRACT

Despite gathering evidence that ubiquitylation can direct non-degradative outcomes, most investigations of ubiquitylation in T cells have focused on degradation. Here, we integrated proteomic and transcriptomic datasets from primary mouse CD4+ T cells to establish a framework for predicting degradative or non-degradative outcomes of ubiquitylation. Di-glycine remnant profiling was used to reveal ubiquitylated proteins, which in combination with whole-cell proteomic and transcriptomic data allowed prediction of protein degradation. Analysis of ubiquitylated proteins identified by di-glycine remnant profiling indicated that activation of CD4+ T cells led to an increase in non-degradative ubiquitylation. This correlated with an increase in non-proteasome-targeted K29, K33 and K63 polyubiquitin chains. This study revealed over 1,200 proteins that were ubiquitylated in primary mouse CD4+ T cells and highlighted the relevance of non-proteasomally targeted ubiquitin chains in T cell signaling.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lymphocyte Activation/immunology , Proteome , Proteomics , Animals , Gene Expression Profiling , Lymphocyte Activation/genetics , Mass Spectrometry , Mice , Polyubiquitin/metabolism , Proteomics/methods , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome , Ubiquitination
11.
Nat Rev Immunol ; 19(9): 584-593, 2019 09.
Article in English | MEDLINE | ID: mdl-31114038

ABSTRACT

Tuft cells are rare, secretory epithelial cells that generated scant immunological interest until contemporaneous reports in 2016 linked tuft cells with type 2 immunity in the small intestine. Tuft cells have the capacity to produce an unusual spectrum of biological effector molecules, including IL-25, eicosanoids implicated in allergy (such as cysteinyl leukotrienes and prostaglandin D2) and the neurotransmitter acetylcholine. In most cases, the extracellular signals controlling tuft cell effector function are unknown, but signal transduction is thought to proceed via canonical, G protein-coupled receptor-dependent pathways involving components of the signalling pathway used by type II taste bud cells to sense sweet, bitter and umami compounds. Tuft cells are ideally positioned as chemosensory sentinels that can detect and relay information from diverse luminal substances via what appear to be stereotyped outputs to initiate both positive and aversive responses through populations of immune and neuronal cells. Despite recent insights, numerous questions remain regarding tuft cell lineage, diversity and effector mechanisms and how tuft cells interface with the immunological niche in the tissues where they reside.


Subject(s)
Intestinal Mucosa/immunology , Intestine, Small/cytology , Acetylcholine/metabolism , Animals , Cell Lineage , Eicosanoids/metabolism , Humans , Interleukin-17/physiology , Intestinal Mucosa/cytology , Intestine, Small/immunology , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Succinic Acid/metabolism
12.
Annu Rev Immunol ; 37: 47-72, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30379593

ABSTRACT

Tuft cells-rare solitary chemosensory cells in mucosal epithelia-are undergoing intense scientific scrutiny fueled by recent discovery of unsuspected connections to type 2 immunity. These cells constitute a conduit by which ligands from the external space are sensed via taste-like signaling pathways to generate outputs unique among epithelial cells: the cytokine IL-25, eicosanoids associated with allergic immunity, and the neurotransmitter acetylcholine. The classic type II taste cell transcription factor POU2F3 is lineage defining, suggesting a conceptualization of these cells as widely distributed environmental sensors with effector functions interfacing type 2 immunity and neural circuits. Increasingly refined single-cell analytics have revealed diversity among tuft cells that extends from nasal epithelia and type II taste cells to ex-Aire-expressing medullary thymic cells and small-intestine cells that mediate tissue remodeling in response to colonizing helminths and protists.


Subject(s)
Epithelium/physiology , Helminthiasis/immunology , Helminths/physiology , Octamer Transcription Factors/metabolism , Sensory Receptor Cells/physiology , Th2 Cells/immunology , Animals , Humans , Immune System , Interleukin-17/metabolism , Nervous System , Neuroimmunomodulation , Octamer Transcription Factors/genetics , Signal Transduction , TRPM Cation Channels/metabolism
13.
Cell ; 174(2): 271-284.e14, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29887373

ABSTRACT

The small intestinal tuft cell-ILC2 circuit mediates epithelial responses to intestinal helminths and protists by tuft cell chemosensory-like sensing and IL-25-mediated activation of lamina propria ILC2s. Small intestine ILC2s constitutively express the IL-25 receptor, which is negatively regulated by A20 (Tnfaip3). A20 deficiency in ILC2s spontaneously triggers the circuit and, unexpectedly, promotes adaptive small-intestinal lengthening and remodeling. Circuit activation occurs upon weaning and is enabled by dietary polysaccharides that render mice permissive for Tritrichomonas colonization, resulting in luminal accumulation of acetate and succinate, metabolites of the protist hydrogenosome. Tuft cells express GPR91, the succinate receptor, and dietary succinate, but not acetate, activates ILC2s via a tuft-, TRPM5-, and IL-25-dependent pathway. Also induced by parasitic helminths, circuit activation and small intestinal remodeling impairs infestation by new helminths, consistent with the phenomenon of concomitant immunity. We describe a metabolic sensing circuit that may have evolved to facilitate mutualistic responses to luminal pathosymbionts.


Subject(s)
Intestine, Small/physiology , Tritrichomonas/metabolism , Acetates/metabolism , Animals , Dietary Fiber/metabolism , Energy Metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/parasitology , Interleukins/genetics , Interleukins/metabolism , Intestinal Mucosa/cytology , Intestine, Small/microbiology , Intestine, Small/parasitology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microbiota , Plasmids/genetics , Plasmids/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Interleukin/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , Succinic Acid/metabolism , TRPM Cation Channels/metabolism , Tritrichomonas/growth & development , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
14.
Nat Commun ; 8: 15677, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28580955

ABSTRACT

Foxp3+ T regulatory (Treg) cells suppress immune cell activation and establish normal immune homeostasis. How Treg cells maintain their identity is not completely understood. Here we show that Ndfip1, a coactivator of Nedd4-family E3 ubiquitin ligases, is required for Treg cell stability and function. Ndfip1 deletion in Treg cells results in autoinflammatory disease. Ndfip1-deficient Treg cells are highly proliferative and are more likely to lose Foxp3 expression to become IL-4-producing TH2 effector cells. Proteomic analyses indicate altered metabolic signature of Ndfip1-deficient Treg cells and metabolic profiling reveals elevated glycolysis and increased mTORC1 signalling. Ndfip1 restricts Treg cell metabolism and IL-4 production via distinct mechanisms, as IL-4 deficiency does not prevent hyperproliferation or elevated mTORC1 signalling in Ndfip1-deficient Treg cells. Thus, Ndfip1 preserves Treg lineage stability and immune homeostasis by preventing the expansion of highly proliferative and metabolically active Treg cells and by preventing pathological secretion of IL-4 from Treg cells.


Subject(s)
Carrier Proteins/metabolism , Inflammation/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Membrane Proteins/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation , Cell Membrane/metabolism , Cell Proliferation , Female , Forkhead Transcription Factors/metabolism , Glycolysis , Hyaluronan Receptors/metabolism , Inflammation/immunology , Intercellular Signaling Peptides and Proteins , Interleukin-4/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Proteomics , Th2 Cells/immunology , Ubiquitination
15.
J Exp Med ; 214(1): 27-37, 2017 01.
Article in English | MEDLINE | ID: mdl-28011865

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) and type 2 helper T cells (Th2 cells) are the primary source of interleukin 5 (IL-5) and IL-13 during type 2 (allergic) inflammation in the lung. In Th2 cells, T cell receptor (TCR) signaling activates the transcription factors nuclear factor of activated T cells (NFAT), nuclear factor κB (NF-κB), and activator protein 1 (AP-1) to induce type 2 cytokines. ILC2s lack a TCR and respond instead to locally produced cytokines such as IL-33. Although IL-33 induces AP-1 and NF-κB, NFAT signaling has not been described in ILC2s. In this study, we report a nonredundant NFAT-dependent role for lipid-derived leukotrienes (LTs) in the activation of lung ILC2s. Using cytokine reporter and LT-deficient mice, we find that complete disruption of LT signaling markedly diminishes ILC2 activation and downstream responses during type 2 inflammation. Type 2 responses are equivalently attenuated in IL-33- and LT-deficient mice, and optimal ILC2 activation reflects potent synergy between these pathways. These findings expand our understanding of ILC2 regulation and may have important implications for the treatment of airways disease.


Subject(s)
Interleukin-33/pharmacology , Leukotrienes/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , NFATC Transcription Factors/physiology , Animals , Drug Synergism , Homeostasis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/physiology
16.
Nat Commun ; 7: 11226, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27088444

ABSTRACT

Nedd4 family E3 ubiquitin ligases have been shown to restrict T-cell function and impact T-cell differentiation. We show here that Ndfip1 and Ndfip2, activators of Nedd4 family ligases, together limit accumulation and function of effector CD4+ T cells. Using a three-part proteomics approach in primary T cells, we identify stabilization of Jak1 in Ndfip1/2-deficient T cells stimulated through the TCR. Jak1 degradation is aborted in activated T cells that lack Ndfips. In wild-type cells, Jak1 degradation lessens CD4+ cell sensitivity to cytokines during TCR stimulation, while in Ndfip-deficient cells cytokine responsiveness persists, promoting increased expansion and survival of pathogenic effector T cells. Thus, Ndfip1/Ndfip2 regulate the cross talk between the T-cell receptor and cytokine signalling pathways to limit inappropriate T-cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Carrier Proteins/metabolism , Cytokines/metabolism , Janus Kinase 1/metabolism , Membrane Proteins/metabolism , Signal Transduction , Animals , CD4-Positive T-Lymphocytes/cytology , Carrier Proteins/genetics , Cell Proliferation , Cells, Cultured , Colitis/genetics , Colitis/metabolism , Female , Immunoblotting , Intercellular Signaling Peptides and Proteins , Lymphocyte Count , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proteolysis , Proteomics/methods , Reverse Transcriptase Polymerase Chain Reaction
17.
Oncoimmunology ; 5(1): e1061175, 2016.
Article in English | MEDLINE | ID: mdl-26942073

ABSTRACT

Neutrophils are important innate immune cells involved in microbial clearance at the sites of infection. However, their role in cancer development is unclear. We hypothesized that neutrophils mediate antitumor effects in early tumorigenesis. To test this, we first studied the cytotoxic effects of neutrophils in vitro. Neutrophils were cytotoxic against tumor cells, with neutrophils isolated from tumor-bearing mice trending to have increased cytotoxic activities. We then injected an ELR+ CXC chemokine-producing tumor cell line into C57BL/6 and Cxcr2-/- mice, the latter lacking the receptors for neutrophil chemokines. We observed increased tumor growth in Cxcr2-/- mice. As expected, tumors from Cxcr2-/- mice contained fewer neutrophils. Surprisingly, these tumors also contained fewer CD8+ T cells, but more IL-17-producing cells. Replenishment of functional neutrophils was correlated with decreased IL-17-producing cells, increased CD8+ T cells, and decreased tumor size in Cxcr2-/- mice, while depletion of neutrophils in C57BL/6 mice showed the opposite effects. Results from a non-ELR+ CXC chemokine producing tumor further supported that functional neutrophils indirectly mediate tumor control by suppressing IL-17A production. We further studied the correlation of IL-17A and CD8+ T cells in vitro. IL-17A suppressed proliferation and IFNγ production of CD8+ T cells, while CD11b+Ly6G+ neutrophils did not suppress CD8+ T cell function. Taken together, these data demonstrate that, while neutrophils could control tumor growth by direct cytotoxic effects, the primary mechanism by which neutrophils exert antitumor effects is to regulate IL-17 production, through which they indirectly promote CD8+ T cell responses.

18.
J Biol Chem ; 290(39): 23875-87, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26245901

ABSTRACT

Nedd4-family E3 ubiquitin ligases regulate an array of biologic processes. Autoinhibition maintains these catalytic ligases in an inactive state through several mechanisms. However, although some Nedd4 family members are activated by binding to Nedd4 family-interacting proteins (Ndfips), how binding activates E3 function remains unclear. Our data reveal how these two regulatory processes are linked functionally. In the absence of Ndfip1, the Nedd4 family member Itch can bind an E2 but cannot accept ubiquitin onto its catalytic cysteine. This is because Itch is autoinhibited by an intramolecular interaction between its HECT (homologous to the E6-AP carboxy terminus domain) and two central WW domains. Ndfip1 binds these WW domains to release the HECT, allowing trans-thiolation and Itch catalytic activity. This molecular switch also regulates the closely related family member WWP2. Importantly, multiple PY motifs are required for Ndfip1 to activate Itch, functionally distinguishing Ndfips from single PY-containing substrates. These data establish a novel mechanism for control of the function of a subfamily of Nedd4 E3 ligases at the level of E2-E3 trans-thiolation.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Humans , Intercellular Signaling Peptides and Proteins , Jurkat Cells , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nedd4 Ubiquitin Protein Ligases , Protein Structure, Tertiary , Repressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics
19.
Front Immunol ; 6: 618, 2015.
Article in English | MEDLINE | ID: mdl-26732666

ABSTRACT

T cell receptor (TCR) signaling must be precisely tuned to limit collateral damage and prevent reactivity to self, while still allowing robust protective immune responses that control pathogen invasion. One process that can be used to promote, modify, or terminate TCR signaling is ubiquitylation. During ubiquitylation, ubiquitin is covalently attached to target proteins through a multistep process, in which E3 ubiquitin ligases promote the formation of ubiquitin chains on selected substrates. Ubiquitylation can facilitate protein-protein interactions, direct a protein to a specific subcellular location, or initiate protein destruction. Like phosphorylation, ubiquitylation is a reversible process - deubiquitylating enzymes counteract ligase function by removing ubiquitin chains. This reversibility also allows for ubiquitin chain "editing." Based on an emerging wealth of information from genetic loss-of-function studies showing that deregulation of ubiquitylation pathways leads to immune dysfunction, it has become increasingly apparent that the dynamic process of ubiquitylation is critical for normal immune cell function. In this review, we will describe how ubiquitylation acts as a key modulator and integrator of signaling downstream of TCR engagement. Specifically, we highlight the known roles of the substrate-specific E3 ligases and deubiquitylating enzymes in TCR signaling and T cell activation. While it is clear that ubiquitin enzymes tune T cell signaling and T cell function, elucidating the molecular mechanisms by which these proteins modulate T cells has met with significant challenges. Identifying substrates of these enzymes has been a particular challenge, and thus substrates of many E3 ligases and deubiquitylating enzymes remain largely unknown. To that end, we discuss the promise, and some practical considerations, of using proteomics-based techniques for unbiased identification of putative substrates of ubiquitin cascade proteins within primary T cells. These methods provide an exciting opportunity for further defining how TCR signals are regulated and for identifying new targets for therapeutic modulation.

20.
Nat Med ; 20(5): 524-30, 2014 May.
Article in English | MEDLINE | ID: mdl-24747744

ABSTRACT

Neonatal colonization by microbes, which begins immediately after birth, is influenced by gestational age and the mother's microbiota and is modified by exposure to antibiotics. In neonates, prolonged duration of antibiotic therapy is associated with increased risk of late-onset sepsis (LOS), a disorder controlled by neutrophils. A role for the microbiota in regulating neutrophil development and susceptibility to sepsis in the neonate remains unclear. We exposed pregnant mouse dams to antibiotics in drinking water to limit transfer of maternal microbes to the neonates. Antibiotic exposure of dams decreased the total number and composition of microbes in the intestine of the neonates. This was associated with decreased numbers of circulating and bone marrow neutrophils and granulocyte/macrophage-restricted progenitor cells in the bone marrow of antibiotic-treated and germ-free neonates. Antibiotic exposure of dams reduced the number of interleukin-17 (IL-17)-producing cells in the intestine and production of granulocyte colony-stimulating factor (G-CSF). Granulocytopenia was associated with impaired host defense and increased susceptibility to Escherichia coli K1 and Klebsiella pneumoniae sepsis in antibiotic-treated neonates, which could be partially reversed by administration of G-CSF. Transfer of a normal microbiota into antibiotic-treated neonates induced IL-17 production by group 3 innate lymphoid cells (ILCs) in the intestine, increasing plasma G-CSF levels and neutrophil numbers in a Toll-like receptor 4 (TLR4)- and myeloid differentiation factor 88 (MyD88)-dependent manner and restored IL-17-dependent resistance to sepsis. Specific depletion of ILCs prevented IL-17- and G-CSF-dependent granulocytosis and resistance to sepsis. These data support a role for the intestinal microbiota in regulation of granulocytosis, neutrophil homeostasis and host resistance to sepsis in neonates.


Subject(s)
Escherichia coli Infections/microbiology , Microbiota , Neutrophils/pathology , Sepsis/microbiology , Animals , Anti-Bacterial Agents/administration & dosage , Escherichia coli/immunology , Escherichia coli/pathogenicity , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Escherichia coli Infections/pathology , Female , Granulocyte Colony-Stimulating Factor/metabolism , Homeostasis , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interleukin-17/metabolism , Intestines/immunology , Intestines/microbiology , Klebsiella pneumoniae/pathogenicity , Mice , Neutrophils/microbiology , Pregnancy , Sepsis/genetics , Sepsis/immunology , Sepsis/pathology , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...