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










Publication year range
1.
Nat Commun ; 13(1): 2027, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440118

ABSTRACT

Innate lymphoid cells (ILCs) govern immune cell homeostasis in the intestine and protect the host against microbial pathogens. Various cell-intrinsic pathways have been identified that determine ILC development and differentiation. However, the cellular components that regulate ILC sustenance and function in the intestinal lamina propria are less known. Using single-cell transcriptomic analysis of lamina propria fibroblasts, we identify fibroblastic reticular cells (FRCs) that underpin cryptopatches (CPs) and isolated lymphoid follicles (ILFs). Genetic ablation of lymphotoxin-ß receptor expression in Ccl19-expressing FRCs blocks the maturation of CPs into mature ILFs. Interactome analysis shows the major niche factors and processes underlying FRC-ILC crosstalk. In vivo validation confirms that a sustained lymphotoxin-driven feedforward loop of FRC activation including IL-7 generation is critical for the maintenance of functional ILC populations. In sum, our study indicates critical fibroblastic niches within the intestinal lamina propria that control ILC homeostasis and functionality and thereby secure protective gut immunity.


Subject(s)
Immunity, Innate , Lymphocytes , Fibroblasts , Homeostasis , Intestines
2.
Trends Immunol ; 42(9): 782-794, 2021 09.
Article in English | MEDLINE | ID: mdl-34362676

ABSTRACT

Fibroblastic reticular cells (FRCs) are a crucial part of the stromal cell infrastructure of secondary lymphoid organs (SLOs). Lymphoid organ fibroblasts form specialized niches for immune cell interactions and thereby govern lymphocyte activation and differentiation. Moreover, FRCs produce and ensheath a network of extracellular matrix (ECM) microfibers called the conduit system. FRC-generated conduits contribute to fluid and immune cell control by funneling fluids containing antigens and inflammatory mediators through the SLOs. We review recent progress in FRC biology that has advanced our understanding of immune cell functions and interactions. We discuss the intricate relationships between the cellular FRC and the fibrillar conduit networks, which together form the basis for efficient communication between immune cells and the tissues they survey.


Subject(s)
Cell Communication , Fibroblasts , Stromal Cells , Extracellular Matrix , Lymph Nodes
3.
Sci Immunol ; 5(51)2020 09 11.
Article in English | MEDLINE | ID: mdl-32917792

ABSTRACT

Fibroblastic reticular cells (FRCs) are stromal cells that actively promote the induction of immune responses by coordinating the interaction of innate and adaptive immune cells. However, whether and to which extent immune cell activation is determined by lymph node FRC reprogramming during acute viral infection has remained unexplored. Here, we genetically ablated expression of the type I interferon-α receptor (Ifnar) in Ccl19-Cre+ cells and found that sensing of type I interferon imprints an antiviral state in FRCs and thereby preserves myeloid cell composition in lymph nodes of naive mice. During localized lymphocytic choriomeningitis virus infection, IFNAR signaling precipitated profound phenotypic adaptation of all FRC subsets enhancing antigen presentation, chemokine-driven immune cell recruitment, and immune regulation. The IFNAR-dependent shift of all FRC subsets toward an immunostimulatory state reduced exhaustive CD8+ T cell activation. In sum, these results unveil intricate circuits underlying type I IFN sensing in lymph node FRCs that enable protective antiviral immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Fibroblasts/immunology , Interferon Type I/immunology , Lymphocytic Choriomeningitis/immunology , Stromal Cells/immunology , Animals , Cell Line , Interferon-gamma/immunology , Lymphocytic choriomeningitis virus , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/immunology
4.
Nat Commun ; 11(1): 3677, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32699279

ABSTRACT

Through the formation of concentration gradients, morphogens drive graded responses to extracellular signals, thereby fine-tuning cell behaviors in complex tissues. Here we show that the chemokine CXCL13 forms both soluble and immobilized gradients. Specifically, CXCL13+ follicular reticular cells form a small-world network of guidance structures, with computer simulations and optimization analysis predicting that immobilized gradients created by this network promote B cell trafficking. Consistent with this prediction, imaging analysis show that CXCL13 binds to extracellular matrix components in situ, constraining its diffusion. CXCL13 solubilization requires the protease cathepsin B that cleaves CXCL13 into a stable product. Mice lacking cathepsin B display aberrant follicular architecture, a phenotype associated with effective B cell homing to but not within lymph nodes. Our data thus suggest that reticular cells of the B cell zone generate microenvironments that shape both immobilized and soluble CXCL13 gradients.


Subject(s)
B-Lymphocytes/immunology , Cellular Microenvironment/immunology , Chemokine CXCL13/metabolism , Dendritic Cells, Follicular/immunology , Adaptive Immunity , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cathepsin B/genetics , Cathepsin B/metabolism , Cell Line , Chemokine CXCL13/immunology , Computer Simulation , Dendritic Cells, Follicular/cytology , Dendritic Cells, Follicular/metabolism , Extracellular Matrix/metabolism , Humans , Mice , Mice, Knockout , Microscopy, Fluorescence , Models, Biological , Palatine Tonsil/cytology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Stromal Cells/immunology , Stromal Cells/metabolism
5.
Nat Immunol ; 21(6): 649-659, 2020 06.
Article in English | MEDLINE | ID: mdl-32424359

ABSTRACT

Efficient generation of germinal center (GC) responses requires directed movement of B cells between distinct microenvironments underpinned by specialized B cell-interacting reticular cells (BRCs). How BRCs are reprogrammed to cater to the developing GC remains unclear, and studying this process is largely hindered by incomplete resolution of the cellular composition of the B cell follicle. Here we used genetic targeting of Cxcl13-expressing cells to define the molecular identity of the BRC landscape. Single-cell transcriptomic analysis revealed that BRC subset specification was predetermined in the primary B cell follicle. Further topological remodeling of light and dark zone follicular dendritic cells required CXCL12-dependent crosstalk with B cells and dictated GC output by retaining B cells in the follicle and steering their interaction with follicular helper T cells. Together, our results reveal that poised BRC-defined microenvironments establish a feed-forward system that determines the efficacy of the GC reaction.


Subject(s)
Darkness , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Immunomodulation/radiation effects , Light , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Cell Communication , Chemokine CXCL12/metabolism , Mice , Mice, Transgenic , Phenotype , Single-Cell Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
6.
Cell Rep ; 30(3): 893-904.e6, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31968261

ABSTRACT

Fibroblastic reticular cells (FRCs) form a road-like cellular network in lymph nodes (LNs) that provides essential chemotactic, survival, and regulatory signals for immune cells. While the topological characteristics of the FRC network have been elaborated, the network properties of the micro-tubular conduit system generated by FRCs, which drains lymph fluid through a pipeline-like system to distribute small molecules and antigens, has remained unexplored. Here, we quantify the crucial 3D morphometric parameters and determine the topological properties governing the structural organization of the intertwined networks. We find that the conduit system exhibits lesser small-worldness and lower resilience to perturbation compared to the FRC network, while the robust topological organization of both networks is maintained in a lymphotoxin-ß-receptor-independent manner. Overall, the high-resolution topological analysis of the "roads-and-pipes" networks highlights essential parameters underlying the functional organization of LN micro-environments and will, hence, advance the development of multi-scale LN models.


Subject(s)
Lymph Nodes/anatomy & histology , Lymph Nodes/physiology , Animals , Fibroblasts/cytology , Kinetics , Lymphotoxin beta Receptor/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Signal Transduction
7.
Science ; 366(6467): 881-886, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31727837

ABSTRACT

Myocarditis can develop into inflammatory cardiomyopathy through chronic stimulation of myosin heavy chain 6-specific T helper (TH)1 and TH17 cells. However, mechanisms governing the cardiotoxicity programming of heart-specific T cells have remained elusive. Using a mouse model of spontaneous autoimmune myocarditis, we show that progression of myocarditis to lethal heart disease depends on cardiac myosin-specific TH17 cells imprinted in the intestine by a commensal Bacteroides species peptide mimic. Both the successful prevention of lethal disease in mice by antibiotic therapy and the significantly elevated Bacteroides-specific CD4+ T cell and B cell responses observed in human myocarditis patients suggest that mimic peptides from commensal bacteria can promote inflammatory cardiomyopathy in genetically susceptible individuals. The ability to restrain cardiotoxic T cells through manipulation of the microbiome thereby transforms inflammatory cardiomyopathy into a targetable disease.


Subject(s)
Autoimmune Diseases/complications , Bacteroides/immunology , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/microbiology , Gastrointestinal Microbiome/immunology , Myocarditis/complications , Peptides/immunology , beta-Galactosidase/immunology , Animals , Autoimmune Diseases/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Humans , Intestines/microbiology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myocarditis/immunology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/immunology , Th17 Cells/immunology
8.
Nat Commun ; 10(1): 1739, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988302

ABSTRACT

The splenic white pulp is underpinned by poorly characterized stromal cells that demarcate distinct immune cell microenvironments. Here we establish fibroblastic reticular cell (FRC)-specific fate-mapping in mice to define their embryonic origin and differentiation trajectories. Our data show that all reticular cell subsets descend from multipotent progenitors emerging at embryonic day 19.5 from periarterial progenitors. Commitment of FRC progenitors is concluded during the first week of postnatal life through occupation of niches along developing central arterioles. Single cell transcriptomic analysis facilitated deconvolution of FRC differentiation trajectories and indicated that perivascular reticular cells function both as adult lymphoid organizer cells and mural cell progenitors. The lymphotoxin-ß receptor-independent sustenance of postnatal progenitor stemness unveils that systemic immune surveillance in the splenic white pulp is governed through subset specification of reticular cells from a multipotent periarterial progenitor cell. In sum, the finding that discrete signaling events in perivascular niches determine the differentiation trajectories of reticular cell networks explains the development of distinct microenvironmental niches in secondary and tertiary lymphoid tissues that are crucial for the induction and regulation of innate and adaptive immune processes.


Subject(s)
Cell Lineage , Cellular Microenvironment , Fibroblasts/physiology , Animals , Cell Differentiation , Gene Expression Profiling , Immunologic Surveillance , Lymphocytes , Mice , Spleen
9.
Front Immunol ; 9: 2428, 2018.
Article in English | MEDLINE | ID: mdl-30405623

ABSTRACT

Adaptive immune responses develop in secondary lymphoid organs such as lymph nodes (LNs) in a well-coordinated series of interactions between migrating immune cells and resident stromal cells. Although many processes that occur in LNs are well understood from an immunological point of view, our understanding of the fundamental organization and mechanisms that drive these processes is still incomplete. The aim of systems biology approaches is to unravel the complexity of biological systems and describe emergent properties that arise from interactions between individual constituents of the system. The immune system is greater than the sum of its parts, as is the case with any sufficiently complex system. Here, we review recent work and developments of computational LN models with focus on the structure and organization of the stromal cells. We explore various mathematical studies of intranodal T cell motility and migration, their interactions with the LN-resident stromal cells, and computational models of functional chemokine gradient fields and lymph flow dynamics. Lastly, we discuss briefly the importance of hybrid and multi-scale modeling approaches in immunology and the technical challenges involved.


Subject(s)
Computational Biology/methods , Computer Simulation , Lymph Nodes/immunology , Stromal Cells/physiology , T-Lymphocytes/physiology , Animals , Chemotaxis , Humans , Models, Theoretical , Systems Biology
10.
Sci Immunol ; 3(26)2018 08 10.
Article in English | MEDLINE | ID: mdl-30097537

ABSTRACT

Immune protection of the body cavities depends on the swift activation of innate and adaptive immune responses in nonclassical secondary lymphoid organs known as fat-associated lymphoid clusters (FALCs). Compared with classical secondary lymphoid organs such as lymph nodes and Peyer's patches, FALCs develop along distinct differentiation trajectories and display a reduced structural complexity. Although it is well established that fibroblastic reticular cells (FRCs) are an integral component of the immune-stimulating infrastructure of classical secondary lymphoid organs, the role of FRCs in FALC-dependent peritoneal immunity remains unclear. Using FRC-specific gene targeting, we found that FRCs play an essential role in FALC-driven immune responses. Specifically, we report that initiation of peritoneal immunity was governed through FRC activation in a myeloid differentiation primary response 88 (MYD88)-dependent manner. FRC-specific ablation of MYD88 blocked recruitment of inflammatory monocytes into FALCs and subsequent CD4+ T cell-dependent B-cell activation and IgG class switching. Moreover, containment of Salmonella infection was compromised in mice lacking MYD88 expression in FRCs, indicating that FRCs in FALCs function as an initial checkpoint in the orchestration of protective immune responses in the peritoneal cavity.


Subject(s)
Fibroblasts/cytology , Fibroblasts/immunology , Intra-Abdominal Fat/immunology , Peritoneal Cavity/physiology , Animals , Chemokine CCL2/immunology , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/immunology , Myeloid Differentiation Factor 88/immunology , Salmonella Infections/immunology , Salmonella typhimurium , Tumor Necrosis Factor-alpha/immunology
11.
Oncoimmunology ; 7(4): e1414129, 2018.
Article in English | MEDLINE | ID: mdl-29632733

ABSTRACT

The tumor microenvironment harbors cancer-associated fibroblasts that function as major modulators of cancer progression. Here, we assessed to which extent distinct cancer-associated fibroblast subsets impact mammary carcinoma growth and cancer cell stemness in an orthotopic murine model. We found that fibroblasts expressing the Cre recombinase under the control of the interleukin 7 promoter occupied mainly the tumor margin where they physically interacted with tumor cells. Intratumoral ablation of interleukin 7-expressing fibroblasts impaired breast tumor growth and reduced the clonogenic potential of cancer cells. Moreover, cDNA expression profiling revealed a distinct oncogenic signature of interleukin 7-producing fibroblasts. In particular, Cxcl12 expression was strongly enhanced in interleukin 7-producing fibroblasts and cell type-specific genetic ablation and systemic pharmacological inhibition revealed that the CXCL12/CXCR4 axis impacts breast tumor cell stemness. Elevated expression of CXCL12 and other stem cell factors in primary human breast cancer-associated fibroblasts indicates that certain fibroblast populations support tumor cell stemness and thereby promote breast cancer growth.

12.
J Allergy Clin Immunol ; 142(4): 1257-1271.e4, 2018 10.
Article in English | MEDLINE | ID: mdl-29391257

ABSTRACT

BACKGROUND: A particular characteristic of non-small cell lung cancer is the composition of the tumor microenvironment with a very high proportion of fibroblastic stromal cells (FSCs). OBJECTIVE: Lapses in our basic knowledge of fibroblast phenotype and function in the tumor microenvironment make it difficult to define whether FSC subsets exist that exhibit either tumor-promoting or tumor-suppressive properties. METHODS: We used gene expression profiling of lung versus tumor FSCs from patients with non-small cell lung cancer. Moreover, CCL19-expressing FSCs were studied in transgenic mouse models by using a lung cancer metastasis model. RESULTS: CCL19 mRNA expression in human tumor FSCs correlates with immune cell infiltration and intratumoral accumulation of CD8+ T cells. Mechanistic dissection in murine lung carcinoma models revealed that CCL19-expressing FSCs form perivascular niches to promote accumulation of CD8+ T cells in the tumor. Targeted ablation of CCL19-expressing tumor FSCs reduced immune cell recruitment and resulted in unleashed tumor growth. CONCLUSION: These data suggest that a distinct population of CCL19-producing FSCs fosters the development of an immune-stimulating intratumoral niche for immune cells to control cancer growth.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Chemokine CCL19/immunology , Fibroblasts/immunology , Lung Neoplasms/immunology , Stromal Cells/immunology , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Chemokine CCL19/genetics , Humans , Lung Neoplasms/genetics , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/transplantation , Transcriptome , Tumor Microenvironment/immunology
13.
Immunity ; 47(1): 80-92.e4, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28709801

ABSTRACT

Lymph nodes (LNs) are strategically situated throughout the body at junctures of the blood vascular and lymphatic systems to direct immune responses against antigens draining from peripheral tissues. The current paradigm describes LN development as a programmed process that is governed through the interaction between mesenchymal lymphoid tissue organizer (LTo) cells and hematopoietic lymphoid tissue inducer (LTi) cells. Using cell-type-specific ablation of key molecules involved in lymphoid organogenesis, we found that initiation of LN development is dependent on LTi-cell-mediated activation of lymphatic endothelial cells (LECs) and that engagement of mesenchymal stromal cells is a succeeding event. LEC activation was mediated mainly by signaling through receptor activator of NF-κB (RANK) and the non-canonical NF-κB pathway and was steered by sphingosine-1-phosphate-receptor-dependent retention of LTi cells in the LN anlage. Finally, the finding that pharmacologically enforced interaction between LTi cells and LECs promotes ectopic LN formation underscores the central LTo function of LECs.


Subject(s)
Endothelial Cells/physiology , Lymph Nodes/physiology , Mesenchymal Stem Cells/physiology , Organogenesis , Animals , Cell Differentiation , Cells, Cultured , Choristoma , Embryo, Mammalian , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction
14.
Methods Mol Biol ; 1591: 43-57, 2017.
Article in English | MEDLINE | ID: mdl-28349474

ABSTRACT

Secondary lymphoid organs have developed segregated niches that are able to initiate and maintain effective immune responses. Such global organization requires tight control of diverse cellular components, specifically those that regulate lymphocyte trafficking. Fibroblastic reticular cells (FRCs) form a densely interconnected network in lymph nodes and provide key factors necessary for T cell migration and retention, and foster subsequent interactions between T cells and dendritic cells. Development of integrative systems biology approaches has made it possible to elucidate this multilevel complexity of the immune system. Here, we present a graph theory-based analysis of the FRC network in murine lymph nodes, where generation of the network topology is performed using high-resolution confocal microscopy and 3D reconstruction. This approach facilitates the analysis of physical cell-to-cell connectivity, and estimation of topological robustness and global behavior of the network when it is subjected to perturbation in silico.


Subject(s)
Cell Communication/immunology , Fibroblasts/immunology , Lymph Nodes/immunology , Animals , Cell Movement/immunology , Dendritic Cells/immunology , Mice , T-Lymphocytes/immunology
15.
Nat Immunol ; 17(12): 1388-1396, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27798617

ABSTRACT

Fibroblastic reticular cells (FRCs) of secondary lymphoid organs form distinct niches for interaction with hematopoietic cells. We found here that production of the cytokine IL-15 by FRCs was essential for the maintenance of group 1 innate lymphoid cells (ILCs) in Peyer's patches and mesenteric lymph nodes. Moreover, FRC-specific ablation of the innate immunological sensing adaptor MyD88 unleashed IL-15 production by FRCs during infection with an enteropathogenic virus, which led to hyperactivation of group 1 ILCs and substantially altered the differentiation of helper T cells. Accelerated clearance of virus by group 1 ILCs precipitated severe intestinal inflammatory disease with commensal dysbiosis, loss of intestinal barrier function and diminished resistance to colonization. In sum, FRCs act as an 'on-demand' immunological 'rheostat' by restraining activation of group 1 ILCs and thereby preventing immunopathological damage in the intestine.


Subject(s)
Citrobacter rodentium/immunology , Coronavirus Infections/immunology , Enterobacteriaceae Infections/immunology , Fibroblasts/immunology , Interleukin-15/metabolism , Lymphocytes/immunology , Murine hepatitis virus/immunology , Animals , Cells, Cultured , Immunity, Innate , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Peyer's Patches/pathology , Th1 Cells/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism
16.
PLoS Biol ; 14(7): e1002515, 2016 07.
Article in English | MEDLINE | ID: mdl-27415420

ABSTRACT

Fibroblastic reticular cells (FRCs) form the cellular scaffold of lymph nodes (LNs) and establish distinct microenvironmental niches to provide key molecules that drive innate and adaptive immune responses and control immune regulatory processes. Here, we have used a graph theory-based systems biology approach to determine topological properties and robustness of the LN FRC network in mice. We found that the FRC network exhibits an imprinted small-world topology that is fully regenerated within 4 wk after complete FRC ablation. Moreover, in silico perturbation analysis and in vivo validation revealed that LNs can tolerate a loss of approximately 50% of their FRCs without substantial impairment of immune cell recruitment, intranodal T cell migration, and dendritic cell-mediated activation of antiviral CD8+ T cells. Overall, our study reveals the high topological robustness of the FRC network and the critical role of the network integrity for the activation of adaptive immune responses.


Subject(s)
Cell Communication/immunology , Cell Movement/immunology , Fibroblasts/immunology , Lymph Nodes/immunology , T-Lymphocytes/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Count , Cell Movement/genetics , Chemokine CCL19/genetics , Chemokine CCL19/immunology , Chemokine CCL19/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Fibroblasts/cytology , Fibroblasts/metabolism , Lymph Nodes/cytology , Lymph Nodes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Models, Immunological , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
17.
Eur J Immunol ; 45(8): 2218-31, 2015 08.
Article in English | MEDLINE | ID: mdl-25973789

ABSTRACT

The thymic epithelium forms specialized niches to enable thymocyte differentiation. While the common epithelial progenitor of medullary and cortical thymic epithelial cells (mTECs and cTECs) is well defined, early stages of mTEC lineage specification have remained elusive. Here, we utilized in vivo targeting of mTECs to resolve their differentiation pathways and to determine whether mTEC progenitors participate in thymocyte education. We found that mTECs descend from a lineage committed, podoplanin (PDPN)-expressing progenitor located at the cortico-medullary junction. PDPN(+) junctional TECs (jTECs) represent a distinct TEC population that builds the thymic medulla, but only partially supports negative selection and thymocyte differentiation. Moreover, conditional gene targeting revealed that abrogation of alternative NF-κB pathway signaling in the jTEC stage completely blocked mTEC development. Taken together, this study identifies jTECs as lineage-committed mTEC progenitors and shows that NF-κB-dependent progression of jTECs to mTECs is critical to secure central tolerance.


Subject(s)
Cell Differentiation/immunology , Epithelial Cells/immunology , Membrane Glycoproteins/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Stem Cells/immunology , Thymus Gland/immunology , Animals , Cell Differentiation/genetics , Epithelial Cells/cytology , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , NF-kappa B/genetics , Signal Transduction/genetics , Stem Cells/cytology , Thymus Gland/cytology
18.
Biochim Biophys Acta ; 1828(3): 1004-12, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23196344

ABSTRACT

A challenge when designing membrane-active peptide antibiotics with therapeutic potential is how to ensure a useful antibacterial activity whilst avoiding unacceptable cytotoxicity for host cells. Understanding their mode of interaction with membranes and the reasons underlying their ability to distinguish between bacterial and eukaryotic cytoplasmic cells is crucial for any rational attempt to improve this selectivity. We have approached this problem by analysing natural helical antimicrobial peptides of anuran origin, using a structure-activity database to determine an antimicrobial selectivity index (SI) relating the minimal inhibitory concentration against Escherichia coli to the haemolytic activity (SI=HC(50)/MIC). A parameter that correlated strongly with SI, derived from the lengthwise asymmetry of the peptides' hydrophobicity (sequence moment), was then used in the "Designer" algorithm to propose novel, highly selective peptides. Amongst these are the 'adepantins', peptides rich in glycines and lysines that are highly selective for Gram-negative bacteria, have an exceptionally low haemolytic activity, and are less than 50% homologous to any other natural or synthetic antimicrobial peptide. In particular, they showed a very high SI for E. coli (up to 400) whilst maintaining an antimicrobial activity in the 0.5-4µM range. Experiments with monomeric, dimeric and fluorescently labelled versions of the adepantins, using different bacterial strains, host cells and model membrane systems provided insight into their mechanism of action.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Animals , Anura , Base Sequence , Circular Dichroism , Dimerization , Dose-Response Relationship, Drug , Drug Design , Erythrocytes/drug effects , Erythrocytes/microbiology , Escherichia coli/drug effects , Escherichia coli/metabolism , Glycine/chemistry , Humans , Liposomes/chemistry , Microbial Sensitivity Tests , Molecular Sequence Data , Peptides/chemistry , Structure-Activity Relationship , Surface Properties , Time Factors
19.
Bioinformatics ; 28(10): 1406-7, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22467909

ABSTRACT

SUMMARY: Anuran tissues, and especially skin, are a rich source of bioactive peptides and their precursors. We here present a manually curated database of antimicrobial and other defense peptides with a total of 2571 entries, most of them in the precursor form with demarcated signal peptide (SP), acidic proregion(s) and bioactive moiety(s) corresponding to 1923 non-identical bioactive sequences. Search functions on the corresponding web server facilitate the extraction of six distinct SP classes. The more conserved of these can be used for searching cDNA and UniProtKB databases for potential bioactive peptides, for creating PROSITE search patterns, and for phylogenetic analysis.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Anura/immunology , Databases, Protein , Animals , Antimicrobial Cationic Peptides/analysis , Antimicrobial Cationic Peptides/immunology , Databases, Factual , Deoxyadenine Nucleotides , Peptides , Phylogeny , Skin/chemistry , Skin/immunology
20.
Eur Biophys J ; 40(4): 371-85, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21274708

ABSTRACT

We describe computational approaches for identifying promising lead candidates for the development of peptide antibiotics, in the context of quantitative structure-activity relationships (QSAR) studies for this type of molecule. A first approach deals with predicting the selectivity properties of generated antimicrobial peptide sequences in terms of measured therapeutic indices (TI) for known antimicrobial peptides (AMPs). Based on a training set of anuran AMPs, the concept of sequence moments was used to construct algorithms that could predict TIs for a second test set of natural AMPs and could also predict the effect of point mutations on TI values. This approach was then used to design peptide antibiotics (adepantins) not homologous to known natural or synthetic AMPs. In a second approach, many novel putative AMPs were identified from DNA sequences in EST databases, using the observation that, as a rule, specific subclasses of highly conserved signal peptides are associated exclusively with AMPs. Both anuran and teleost sequences were used to elucidate this observation and its implications. The predicted therapeutic indices of identified sequences could then be used to identify new types of selective putative AMPs for future experimental verification.


Subject(s)
Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Computational Biology/methods , Anti-Infective Agents/chemistry , Anti-Infective Agents/therapeutic use , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/therapeutic use , Drug Design , Knowledge Bases , Quantitative Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...