Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4703, 2023 08 05.
Article in English | MEDLINE | ID: mdl-37543621

ABSTRACT

TGFß signaling is associated with non-response to immune checkpoint blockade in patients with advanced cancers, particularly in the immune-excluded phenotype. While previous work demonstrates that converting tumors from excluded to inflamed phenotypes requires attenuation of PD-L1 and TGFß signaling, the underlying cellular mechanisms remain unclear. Here, we show that TGFß and PD-L1 restrain intratumoral stem cell-like CD8 T cell (TSCL) expansion and replacement of progenitor-exhausted and dysfunctional CD8 T cells with non-exhausted T effector cells in the EMT6 tumor model in female mice. Upon combined TGFß/PD-L1 blockade IFNγhi CD8 T effector cells show enhanced motility and accumulate in the tumor. Ensuing IFNγ signaling transforms myeloid, stromal, and tumor niches to yield an immune-supportive ecosystem. Blocking IFNγ abolishes the anti-PD-L1/anti-TGFß therapy efficacy. Our data suggest that TGFß works with PD-L1 to prevent TSCL expansion and replacement of exhausted CD8 T cells, thereby maintaining the T cell compartment in a dysfunctional state.


Subject(s)
B7-H1 Antigen , Breast Neoplasms , CD8-Positive T-Lymphocytes , Immune Checkpoint Inhibitors , Transforming Growth Factor beta , Female , Animals , Mice , Cell Differentiation , CD8-Positive T-Lymphocytes/immunology , Stem Cells , B7-H1 Antigen/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Interferon-gamma/immunology , T-Cell Exhaustion , Immune Checkpoint Inhibitors/pharmacology , Mice, Inbred BALB C , Cell Line, Tumor , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , RNA-Seq
2.
Clin Exp Immunol ; 211(2): 176-183, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36571811

ABSTRACT

The actions of the immune system are finely tuned, involving complex communication and coordination between diverse immune and non-immune cells across the tissues of the body. A healthy immune system requires a precise balance between immunity and tolerance. Regulatory T cells (Tregs) have long been appreciated as one of the master regulators of this balance; their importance is underscored by the autoimmunity that develops in mice and humans when Tregs are missing or dysfunctional. In addition to the immunoregulatory roles of Tregs in suppressing autoimmunity and inflammation via control of adaptive and innate immune responses, several non-immune modulatory functions of Tregs have been identified in recent years. In this review, we have highlighted the growing literature on the action of Tregs in metabolism, stem cell maintenance, tissue repair, and angiogenesis. Alongside Tregs' immune suppressive role, these non-suppressive activities comprise a key function of Tregs in regulating health and disease. As Tregs receive increasing attention as therapeutic targets, understanding their non-canonical functions may become an important feature of Treg-directed interventions.


Subject(s)
Immune Tolerance , T-Lymphocytes, Regulatory , Humans , Animals , Mice , Autoimmunity
3.
PLoS One ; 16(12): e0260800, 2021.
Article in English | MEDLINE | ID: mdl-34879110

ABSTRACT

The cancer-associated fibroblast (CAF) marker podoplanin (PDPN) is generally correlated with poor clinical outcomes in cancer patients and thus represents a promising therapeutic target. Despite its biomedical relevance, basic aspects of PDPN biology such as its cellular functions and cell surface ligands remain poorly uncharacterized, thus challenging drug development. Here, we utilize a high throughput platform to elucidate the PDPN cell surface interactome, and uncover the neutrophil protein CD177 as a new binding partner. Quantitative proteomics analysis of the CAF phosphoproteome reveals a role for PDPN in cell signaling, growth and actomyosin contractility, among other processes. Moreover, cellular assays demonstrate that CD177 is a functional antagonist, recapitulating the phenotype observed in PDPN-deficient CAFs. In sum, starting from the unbiased elucidation of the PDPN co-receptome, our work provides insights into PDPN functions and reveals the PDPN/CD177 axis as a possible modulator of fibroblast physiology in the tumor microenvironment.


Subject(s)
Biomarkers, Tumor/metabolism , Cancer-Associated Fibroblasts/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Isoantigens/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Tumor Microenvironment , Apoptosis , Biomarkers, Tumor/genetics , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Isoantigens/genetics , Membrane Glycoproteins/genetics , Neutrophils/immunology , Neutrophils/metabolism , Prognosis , Receptors, Cell Surface/genetics , Survival Rate , Tumor Cells, Cultured
4.
Nat Immunol ; 22(5): 571-585, 2021 05.
Article in English | MEDLINE | ID: mdl-33903764

ABSTRACT

Fibroblastic reticular cells (FRCs) are specialized stromal cells that define tissue architecture and regulate lymphocyte compartmentalization, homeostasis, and innate and adaptive immunity in secondary lymphoid organs (SLOs). In the present study, we used single-cell RNA sequencing (scRNA-seq) of human and mouse lymph nodes (LNs) to identify a subset of T cell-zone FRCs defined by the expression of Gremlin1 (Grem1) in both species. Grem1-CreERT2 knock-in mice enabled localization, multi-omics characterization and genetic depletion of Grem1+ FRCs. Grem1+ FRCs primarily localize at T-B cell junctions of SLOs, neighboring pre-dendritic cells and conventional dendritic cells (cDCs). As such, their depletion resulted in preferential loss and decreased homeostatic proliferation and survival of resident cDCs and compromised T cell immunity. Trajectory analysis of human LN scRNA-seq data revealed expression similarities to murine FRCs, with GREM1+ cells marking the endpoint of both trajectories. These findings illuminate a new Grem1+ fibroblastic niche in LNs that functions to maintain the homeostasis of lymphoid tissue-resident cDCs.


Subject(s)
Dendritic Cells, Follicular/immunology , Fibroblasts/immunology , Lymph Nodes/immunology , Stromal Cells/immunology , Aged , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Proliferation/genetics , Cell Survival/genetics , Cell Survival/immunology , Dendritic Cells, Follicular/metabolism , Female , Fibroblasts/metabolism , Gene Expression Regulation/immunology , Gene Knock-In Techniques , Humans , Immunity, Cellular/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lymph Nodes/cytology , Male , Mice , Mice, Transgenic , RNA-Seq , Single-Cell Analysis , Stromal Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
5.
Cancer Immunol Res ; 6(12): 1472-1485, 2018 12.
Article in English | MEDLINE | ID: mdl-30266714

ABSTRACT

Cancer-associated fibroblasts (CAFs) are generally associated with poor clinical outcome. CAFs support tumor growth in a variety of ways and can suppress antitumor immunity and response to immunotherapy. However, a precise understanding of CAF contributions to tumor growth and therapeutic response is lacking. Discrepancies in this field of study may stem from heterogeneity in the composition and function of fibroblasts in the tumor microenvironment. Furthermore, it remains unclear whether CAFs directly interact with and suppress T cells. Here, mouse and human breast tumors were used to examine stromal cells expressing fibroblast activation protein (FAP), a surface marker for CAFs. Two discrete populations of FAP+ mesenchymal cells were identified on the basis of podoplanin (PDPN) expression: a FAP+PDPN+ population of CAFs and a FAP+PDPN- population of cancer-associated pericytes (CAPs). Although both subsets expressed extracellular matrix molecules, the CAF transcriptome was enriched in genes associated with TGFß signaling and fibrosis compared with CAPs. In addition, CAFs were enriched at the outer edge of the tumor, in close contact with T cells, whereas CAPs were localized around vessels. Finally, FAP+PDPN+ CAFs suppressed the proliferation of T cells in a nitric oxide-dependent manner, whereas FAP+PDPN- pericytes were not immunosuppressive. Collectively, these findings demonstrate that breast tumors contain multiple populations of FAP-expressing stromal cells of dichotomous function, phenotype, and location.


Subject(s)
Breast Neoplasms/pathology , Gelatinases/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Stromal Cells/metabolism , Tumor Microenvironment/immunology , Animals , Breast Neoplasms/immunology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Proliferation , Endopeptidases , Female , Gene Expression Regulation , Humans , Membrane Glycoproteins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide/metabolism , Pericytes/metabolism , Pericytes/pathology , Stromal Cells/pathology , T-Lymphocytes/pathology
6.
Nature ; 554(7693): 544-548, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29443960

ABSTRACT

Therapeutic antibodies that block the programmed death-1 (PD-1)-programmed death-ligand 1 (PD-L1) pathway can induce robust and durable responses in patients with various cancers, including metastatic urothelial cancer. However, these responses only occur in a subset of patients. Elucidating the determinants of response and resistance is key to improving outcomes and developing new treatment strategies. Here we examined tumours from a large cohort of patients with metastatic urothelial cancer who were treated with an anti-PD-L1 agent (atezolizumab) and identified major determinants of clinical outcome. Response to treatment was associated with CD8+ T-effector cell phenotype and, to an even greater extent, high neoantigen or tumour mutation burden. Lack of response was associated with a signature of transforming growth factor ß (TGFß) signalling in fibroblasts. This occurred particularly in patients with tumours, which showed exclusion of CD8+ T cells from the tumour parenchyma that were instead found in the fibroblast- and collagen-rich peritumoural stroma; a common phenotype among patients with metastatic urothelial cancer. Using a mouse model that recapitulates this immune-excluded phenotype, we found that therapeutic co-administration of TGFß-blocking and anti-PD-L1 antibodies reduced TGFß signalling in stromal cells, facilitated T-cell penetration into the centre of tumours, and provoked vigorous anti-tumour immunity and tumour regression. Integration of these three independent biological features provides the best basis for understanding patient outcome in this setting and suggests that TGFß shapes the tumour microenvironment to restrain anti-tumour immunity by restricting T-cell infiltration.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , Transforming Growth Factor beta/metabolism , Urologic Neoplasms/drug therapy , Urologic Neoplasms/immunology , Urothelium/pathology , Animals , Antibodies/immunology , Antibodies/pharmacology , Antibodies/therapeutic use , Antibodies, Monoclonal, Humanized , Antigens, Neoplasm/analysis , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle Checkpoints/drug effects , Cohort Studies , Collagen/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Fibroblasts/metabolism , Humans , Immunotherapy , Mice , Mutation , Neoplasm Metastasis , Phenotype , Signal Transduction/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , Treatment Outcome , Tumor Microenvironment/immunology , Urologic Neoplasms/genetics , Urologic Neoplasms/pathology , Urothelium/drug effects , Urothelium/immunology
7.
Immunity ; 43(6): 1160-73, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26682987

ABSTRACT

Tertiary lymphoid tissues (TLTs) have been observed in the meninges of multiple sclerosis (MS) patients, but the stromal cells and molecular signals that support TLTs remain unclear. Here, we show that T helper 17 (Th17) cells induced robust TLTs within the brain meninges that were associated with local demyelination during experimental autoimmune encephalitis (EAE). Th17-cell-induced TLTs were underpinned by a network of stromal cells producing extracellular matrix proteins and chemokines, enabling leukocytes to reside within, rather than simply transit through, the meninges. Within the CNS, interactions between lymphotoxin αß (LTαß) on Th17 cells and LTßR on meningeal radio-resistant cells were necessary for the propagation of de novo interleukin-17 responses, and activated T cells from MS patients expressed elevated levels of LTßR ligands. Therefore, input from both Th17 cells and the lymphotoxin pathway induce the formation of an immune-competent stromal cell niche in the meninges.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphotoxin-alpha/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , Stromal Cells/immunology , Th17 Cells/immunology , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Flow Cytometry , Humans , Immunohistochemistry , Inflammation/immunology , Male , Meninges/cytology , Meninges/immunology , Mice , Mice, Knockout , Polymerase Chain Reaction , Signal Transduction/immunology
8.
Nat Rev Immunol ; 15(11): 669-82, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26471778

ABSTRACT

A dynamic and mutualistic interaction between tumour cells and the surrounding stroma promotes the initiation, progression, metastasis and chemoresistance of solid tumours. Far less understood is the relationship between the stroma and tumour-infiltrating leukocytes; however, emerging evidence suggests that the stromal compartment can shape antitumour immunity and responsiveness to immunotherapy. Thus, there is growing interest in elucidating the immunomodulatory roles of the stroma that evolve within the tumour microenvironment. In this Review, we discuss the evidence that stromal determinants interact with leukocytes and influence antitumour immunity, with emphasis on the immunological attributes of stromal cells that may foster their protumorigenic function.


Subject(s)
Stromal Cells/immunology , Tumor Microenvironment , Animals , Cell Communication , Cell Movement , Cytotoxicity, Immunologic , Extracellular Matrix/physiology , Fibroblasts/physiology , Humans , Leukocytes/physiology , Mesenchymal Stem Cells/physiology
9.
Nat Immunol ; 16(1): 75-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25347465

ABSTRACT

In lymph nodes, fibroblastic reticular cells (FRCs) form a collagen-based reticular network that supports migratory dendritic cells (DCs) and T cells and transports lymph. A hallmark of FRCs is their propensity to contract collagen, yet this function is poorly understood. Here we demonstrate that podoplanin (PDPN) regulates actomyosin contractility in FRCs. Under resting conditions, when FRCs are unlikely to encounter mature DCs expressing the PDPN receptor CLEC-2, PDPN endowed FRCs with contractile function and exerted tension within the reticulum. Upon inflammation, CLEC-2 on mature DCs potently attenuated PDPN-mediated contractility, which resulted in FRC relaxation and reduced tissue stiffness. Disrupting PDPN function altered the homeostasis and spacing of FRCs and T cells, which resulted in an expanded reticular network and enhanced immunity.


Subject(s)
Collagen/metabolism , Fibroblasts/cytology , Lectins, C-Type/metabolism , Lymph Nodes/cytology , Membrane Glycoproteins/metabolism , Amides/pharmacology , Animals , Cell Survival/immunology , Collagen/immunology , Cytoskeleton/immunology , Cytoskeleton/ultrastructure , Enzyme Inhibitors/pharmacology , Female , Fibroblasts/immunology , Fibroblasts/ultrastructure , Lectins, C-Type/immunology , Lymph Nodes/immunology , Lymph Nodes/ultrastructure , Male , Membrane Glycoproteins/immunology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Phosphorylation , Pyridines/pharmacology , Specific Pathogen-Free Organisms
10.
Nature ; 514(7523): 498-502, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25341788

ABSTRACT

After immunogenic challenge, infiltrating and dividing lymphocytes markedly increase lymph node cellularity, leading to organ expansion. Here we report that the physical elasticity of lymph nodes is maintained in part by podoplanin (PDPN) signalling in stromal fibroblastic reticular cells (FRCs) and its modulation by CLEC-2 expressed on dendritic cells. We show in mouse cells that PDPN induces actomyosin contractility in FRCs via activation of RhoA/C and downstream Rho-associated protein kinase (ROCK). Engagement by CLEC-2 causes PDPN clustering and rapidly uncouples PDPN from RhoA/C activation, relaxing the actomyosin cytoskeleton and permitting FRC stretching. Notably, administration of CLEC-2 protein to immunized mice augments lymph node expansion. In contrast, lymph node expansion is significantly constrained in mice selectively lacking CLEC-2 expression in dendritic cells. Thus, the same dendritic cells that initiate immunity by presenting antigens to T lymphocytes also initiate remodelling of lymph nodes by delivering CLEC-2 to FRCs. CLEC-2 modulation of PDPN signalling permits FRC network stretching and allows for the rapid lymph node expansion--driven by lymphocyte influx and proliferation--that is the critical hallmark of adaptive immunity.


Subject(s)
Dendritic Cells/physiology , Fibroblasts/cytology , Lymph Nodes/cytology , Stromal Cells/cytology , Actomyosin/metabolism , Animals , Cell Membrane/metabolism , Cytoskeleton/metabolism , Dendritic Cells/immunology , Female , Fibroblasts/physiology , Inflammation/immunology , Lectins, C-Type/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Stromal Cells/physiology , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein , rhoC GTP-Binding Protein
11.
Front Immunol ; 3: 283, 2012.
Article in English | MEDLINE | ID: mdl-22988448

ABSTRACT

Podoplanin (PDPN) is a well-conserved, mucin-type transmembrane protein expressed in multiple tissues during ontogeny and in adult animals, including the brain, heart, kidney, lungs, osteoblasts, and lymphoid organs. Studies of PDPN-deficient mice have demonstrated that this molecule plays a critical role in development of the heart, lungs, and lymphatic system. PDPN is widely used as a marker for lymphatic endothelial cells and fibroblastic reticular cells of lymphoid organs and for lymphatics in the skin and tumor microenvironment. Much of the mechanistic insight into PDPN biology has been gleaned from studies of tumor cells; tumor cells often upregulate PDPN as they undergo epithelial-mesenchymal transition and this upregulation is correlated with increased motility and metastasis. The physiological role of PDPN that has been most studied is its ability to aggregate and activate CLEC-2-expressing platelets, as PDPN is the only known endogenous ligand for CLEC-2. However, more recent studies have revealed that PDPN also plays crucial roles in the biology of immune cells, including T cells and dendritic cells. This review will provide a comprehensive overview of the diverse roles of PDPN in development, immunology, and cancer.

12.
Proc Natl Acad Sci U S A ; 109(41): 16696-701, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-23010927

ABSTRACT

The AcrAB-TolC multidrug efflux pump confers resistance to a wide variety of antibiotics and other compounds in Escherichia coli. Here we show that AcrZ (formerly named YbhT), a 49-amino-acid inner membrane protein, associates with the AcrAB-TolC complex. Co-purification of AcrZ with AcrB, in the absence of both AcrA and TolC, two-hybrid assays and suppressor mutations indicate that this interaction occurs through the inner membrane protein AcrB. The highly conserved acrZ gene is coregulated with acrAB through induction by the MarA, Rob, and SoxS transcription regulators. In addition, mutants lacking AcrZ are sensitive to many, but not all, of the antibiotics transported by AcrAB-TolC. This differential antibiotic sensitivity suggests that AcrZ may enhance the ability of the AcrAB-TolC pump to export certain classes of substrates.


Subject(s)
Drug Resistance, Microbial , Escherichia coli Proteins/metabolism , Lipoproteins/metabolism , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Conserved Sequence/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Immunoblotting , Lipoproteins/chemistry , Lipoproteins/genetics , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Microbial Sensitivity Tests , Models, Molecular , Molecular Sequence Data , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/genetics , Mutation , Periplasm/metabolism , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Trans-Activators/genetics , Trans-Activators/metabolism , Two-Hybrid System Techniques
13.
Immunity ; 37(2): 276-89, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22884313

ABSTRACT

To initiate adaptive immunity, dendritic cells (DCs) move from parenchymal tissues to lymphoid organs by migrating along stromal scaffolds that display the glycoprotein podoplanin (PDPN). PDPN is expressed by lymphatic endothelial and fibroblastic reticular cells and promotes blood-lymph separation during development by activating the C-type lectin receptor, CLEC-2, on platelets. Here, we describe a role for CLEC-2 in the morphodynamic behavior and motility of DCs. CLEC-2 deficiency in DCs impaired their entry into lymphatics and trafficking to and within lymph nodes, thereby reducing T cell priming. CLEC-2 engagement of PDPN was necessary for DCs to spread and migrate along stromal surfaces and sufficient to induce membrane protrusions. CLEC-2 activation triggered cell spreading via downregulation of RhoA activity and myosin light-chain phosphorylation and triggered F-actin-rich protrusions via Vav signaling and Rac1 activation. Thus, activation of CLEC-2 by PDPN rearranges the actin cytoskeleton in DCs to promote efficient motility along stromal surfaces.


Subject(s)
Cell Movement/physiology , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Actins/metabolism , Adaptive Immunity/physiology , Animals , Antigen-Presenting Cells/metabolism , Blood Platelets/metabolism , Cells, Cultured , Dendritic Cells/immunology , Embryo, Mammalian , Endothelial Cells/metabolism , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/metabolism , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymph Nodes/cytology , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Myosin Light Chains/metabolism , Platelet Activation , Pregnancy , Proto-Oncogene Proteins c-vav/metabolism , Signal Transduction/physiology , Skin/cytology , Skin/metabolism , Tissue Culture Techniques , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
14.
J Bacteriol ; 192(1): 59-67, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19734312

ABSTRACT

More than 80 small regulatory RNAs (sRNAs) and 60 proteins of 16 to 50 amino acids (small proteins) are encoded in the Escherichia coli genome. The vast majority of the corresponding genes have no known function. We screened 125 DNA bar-coded mutants to identify novel cell envelope stress and acute acid shock phenotypes associated with deletions of genes coding for sRNAs and small proteins. Nine deletion mutants (ssrA, micA, ybaM, ryeF, yqcG, sroH, ybhT, yobF, and glmY) were sensitive to cell envelope stress and two were resistant (rybB and blr). Deletion mutants of genes coding for four small proteins (yqgB, mgrB, yobF, and yceO) were sensitive to acute acid stress. We confirmed each of these phenotypes in one-on-one competition assays against otherwise-wild-type lacZ mutant cells. A more detailed investigation of the SsrA phenotype suggests that ribosome release is critical for resistance to cell envelope stress. The bar-coded deletion collection we generated can be screened for sensitivity or resistance to virtually any stress condition.


Subject(s)
Escherichia coli Proteins/physiology , Escherichia coli/drug effects , Escherichia coli/metabolism , Hydrochloric Acid/pharmacology , RNA, Bacterial/physiology , Sodium Dodecyl Sulfate/pharmacology , Cell Wall/drug effects , Cell Wall/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Bacterial/genetics , Genetic Complementation Test , Kanamycin Resistance/physiology , Mutation , Oligonucleotide Array Sequence Analysis , RNA, Bacterial/genetics , Stress, Physiological
SELECTION OF CITATIONS
SEARCH DETAIL
...