key: cord-0803235-sktdrd8q authors: Lütge, Mechthild; Pikor, Natalia B.; Ludewig, Burkhard title: Differentiation and activation of fibroblastic reticular cells date: 2021-05-27 journal: Immunol Rev DOI: 10.1111/imr.12981 sha: cef2aae7f190d3a8995e5f081a531504943917c7 doc_id: 803235 cord_uid: sktdrd8q Secondary lymphoid organs (SLO) are underpinned by fibroblastic reticular cells (FRC) that form dedicated microenvironmental niches to secure induction and regulation of innate and adaptive immunity. Distinct FRC subsets are strategically positioned in SLOs to provide niche factors and govern efficient immune cell interaction. In recent years, the use of specialized mouse models in combination with single‐cell transcriptomics has facilitated the elaboration of the molecular FRC landscape at an unprecedented resolution. While single‐cell RNA‐sequencing has advanced the resolution of FRC subset characterization and function, the high dimensionality of the generated data necessitates careful analysis and validation. Here, we reviewed novel findings from high‐resolution transcriptomic analyses that refine our understanding of FRC differentiation and activation processes in the context of infection and inflammation. We further discuss concepts, strategies, and limitations for the analysis of single‐cell transcriptome data from FRCs and the wide‐ranging implications for our understanding of stromal cell biology. Innate and adaptive immune responses are initiated and coordinated in secondary lymphoid organs (SLOs) that are strategically positioned throughout the body to sample antigens from pathogens, commensal organisms, tumors, or any other environmental sources. In addition, innate immunological signals from inflamed tissues or the body surfaces are relayed to SLOs to amplify or atone immune reactions. Diligent processes in SLOs guide the cellular interactions that generate protective immunity while minimizing immunopathological damage in the tissues. The decision-making processes that determine the strength, breadth, and specificity of adaptive immune responses occur in dedicated niches within SLOs. Specialized lymphoid organ fibroblasts, commonly referred to as fibroblastic reticular cells (FRCs), form the scaffold of SLOs and determine the microenvironmental conditions for lymphocyte activation and differentiation. 1 signals from a dedicated sampling site, (b) sample and distribute antigen to lymphocytes and antigen-presenting cells, and (c) promote the efficient activation and interaction of cognate T and B lymphocytes. 5 To this end, all SLOs adopt a similar structural patterning with an antigen-sampling zone filled with specialized myeloid cell subsets, and dedicated B cell follicles and adjoining T cell regions to initiate and direct lymphocyte priming and differentiation. Activated lymphocytes must then converge within the B cell follicle or the T cell zone to achieve, for example, the generation of high-affinity antibody responses or effector CD8 + T cells, respectively. This series of "combinatorial decision processes" guiding myeloid cell and lymphocyte homing and compartmentalization is orchestrated by chemokines secreted by FRCs. 6,7 CXCL13-expressing FRCs govern B cell clustering and follicle formation, 8, 9 while CCL19/CCL21-expressing FRC orchestrate dendritic cell and T cell homing and homeostasis. [10] [11] [12] Within each zone, specialized FRC subsets coordinate the directed movement of myeloid cells and activated lymphocytes or the display of antigens to harmoniously secure efficient adaptive immunity. Several terms have been used to describe the fibroblastic stromal cells underpinning SLOs, including myofibroblasts, pericytes, and FRCs. 13 In 1968, the term reticular cells was first used to describe the elongated fibroblastic cells surrounding thin filamentous processes, a reticulum, traversing the B cell follicle of the lymph node, 14 and the spleen. 15 This topological property is also conserved by FRCs in Peyer's patches, 16 and the formation of a reticulum is a feature not only of fibroblastic cells in the B cell follicle but also in the T cell zone. 13, 17 More recently, FRCs have been characterized by the expression of podoplanin (PDPN, also referred to as gp38) and lack of the endothelial marker CD31 (platelet endothelial cell adhesion molecule-1, PECAM-1). 11 Although PDPN + CD31 − FRCs have often been associated with the T cell zone, 11, 18, 19 the term FRC is now well-accepted to broadly demarcate immune-interacting fibroblastic stromal cells within SLOs. Additional topological and functional attributes characterize distinct FRC subsets, such as those in the B cell follicle or the T cell zone. While CCL19-secreting FRCs, as well as a subset of CXCL13secreting, antigen-presenting cells termed follicular dendritic cells (FDCs), have prevailed in our knowledge of FRC subsets, in recent years, major advances in genetic models paired with the advent of single-cell transcriptomics have created novel means to resolve FRC heterogeneity and function. New, highly specialized FRC subsets have been defined and implicated in the fine-tuned coordination of lymphocyte migration and priming. In this review, we will discuss recent findings delineating FRC differentiation and activation obtained by genetic targeting in mouse models and discuss important strategies and limitations of high dimensional data analysis for the understanding of FRC biology. The most basic and still frequently used approach to identify FRCs has been based on the expression of PDPN and an extracellular matrix protein that is recognized by the antibody ER-TR7. 11, 20 However, PDPN-negative FRCs populate several sites in lymph nodes, 21 and PDPN expression in the spleen is largely restricted to the T cell zone. 22, 23 Thus, tracking changes in the relative abundance, phenotype, or transcriptional profile of non-hematopoietic CD31 -PDPN + cells only offers a very limited insight into the nature of FRCs in the classical SLOs (lymph nodes, splenic white pulp, or Peyer's patches) or non-classical SLOs such as fat-associated lymphoid clusters (FALCs). Genetic targeting of particular cell types in vivo using specific promoters to drive the expression of real-time reporters or the Cre recombinase has offered novel means to elaborate origin, phenotype, and function of FRCs. In contrast to T and B cells, FRC-secreted factors are not readily or sensitively assayed by traditional methods such as flow cytometry or histology. Over the last 10 years, several strains of reporter mice and lineage tracing models expressing Cre recombinase and/or fluorescent proteins (eg, the enhanced yellow fluorescent protein, EYFP) under the promoter of key FRC signature genes have been developed, revolutionizing FRC immunobiology (Table 1 ). Two such transgenic mouse models that take advantage of FRC chemokine expression are the Ccl19-Cre 24 spleen at E19.5 23 ) thereby targeting FRCs in lymph nodes, 21, 24, 28 the splenic white pulp, 29 and Peyer's patches. 30 Similarly, in lymph node anlagen, the Cxcl13-Cre transgene is expressed at E14 in mesenchymal stromal cells, marking all major lymph node FRC subsets in adult mice. 25 In contrast, Cxcl13-expressing, TdTomato + FRCs are largely confined to the B cell follicles. 25, 31 Targeting of this transgene in the splenic white pulp and Peyer's patches remains to be studied in detail. In addition to these signature chemokine-based FRC-targeting models, a number of transgenic strains permit the lineage tracing of a particular FRC repertoire. The DM2 BAC transgenic model harbors a multiple reporter construct containing the diphtheria toxin receptor (DTR), firefly luciferase, and the fluorescent reporter mCherry under the control of the fibroblast activation proteinα (Fap) promoter. 32 The absence of a Cre recombinase-driven labeling of transgene-targeted cells does not make this particular model suitable for lineage tracing but rather acts as a direct reporter of current FAP expression. The BAC-encoded DTR permits the inducible deletion of FAP-expressing cells following DT administration. In lymph nodes and Peyer's patches, FAP expression is primarily restricted to FRCs in the T cell zone but is detected in a low frequency of cells in the splenic white pulp. 33 The Col6a1-Cre crossed to the R26 mT/mG reporter allows for targeting and lineage tracing of cells expressing, or having expressed Col6a1. 34 This model predominantly targets FDCs and marginal zone reticular cells (MRCs) in Peyer's patches but does not readily target FRCs in the splenic white pulp or lymph nodes. 35 The Cd21-Cre R26R-tdRFP mouse model was designed as a reporter and lineage tracing model of FDCs, based on the high expression of CD21 (murine complement receptor 2) by these cells. 36 Ccl19-iEYFP additionally encodes for a TdTomato reporter under the control of the Ccl19-promoter to distinguish current from past promoter activity. These models have been used to identify a common FAP-expressing progenitor of lymph node FRCs 39 and to delineate differentiation trajectories of splenic white pulp FRCs that were found to originate from multipotent periarterial, CCL19-expressing FRC progenitors. 23 Additional tools to study clonal relationships include multicolor tagging systems based on Brainbow models. 40, 41 The combinatorial expression of multiple fluorescent proteins under the control of the Rosa26 promoter permits the Cre-induced lineage tracing or fate mapping of individual clones within a targeted cell population. This system has been used to study the developmental origin and differentiation dynamics of FDCs in lymph nodes, 42 and FRCs in the splenic white pulp 23 and Peyer's patches. 27 Collectively, these genetic models have aided in elucidating patterns of FRC differentiation across SLOs by marking progenitorprogeny relationships, enriching known or rare cell populations for single-cell RNA-sequencing, and demonstrably establishing the function of FRC subsets as will be discussed in the following sections. In addition, the broad and constitutive targeting of fibroblasts also makes many of these Cre recombinase-driven lineage tracing models suitable for targeting stromal cells in inflammatory lesions or the tumor microenvironment, 32,43-45 making it possible to study immune-interacting stromal cells outside of lymphoid tissues. In recent years, single-cell profiling has become a fast-growing field with a vast number of technological advances accompanied by a flood of new computational tools and algorithms for single-cell data analysis. 46 This rapid progress opens a multitude of new opportunities, but at the same time poses challenges for standardization, and consequently for data interpretation and reproducibility. 47, 48 Key variables that ultimately determine the quality and reproducibility of the analysis include sequencing depth, total cell number, and clustering strategy. 49, 50 However, these parameters themselves depend on whether heterogeneous cell types are being analyzed, such as in whole organ analyses, or whether the heterogeneity across subsets or activation states of a single-cell type is being performed. 51 Thus, a robust and meaningful clustering depth, assignment, and validation strongly determine the biological interpretation of cellular heterogeneity using single-cell transcriptomics. The FRC landscape consists of specialized subsets that form distinct niches within SLOs corresponding to the T cell zone, B cell follicle, antigen-sampling regions, and sites of lymphocyte entry ( Figure 1 ). 1, 5 As such, FRCs can be broadly categorized as T cell zone reticular cell Instead, it is often favorable to run a number of algorithms with different parameters to gain confidence about cluster robustness and potential "over-" or "under-clustering". 47 As a general rule of thumb, reducing technical noise and resolving the heterogeneity of highly similar or rare cell populations requires a sufficiently high cell input number, and in the case of FRCs, can be facilitated by enriching for desired subsets using available reporter models 51 (see Table 1 ). Once cluster robustness is tested, the list of each cluster's marker genes should reflect the spatial positioning and immune cell-interacting partners of FRC subsets. Notably, the final choice of clustering depth can vary and should be adapted to the research question and a resolution that can be validated. [56] [57] [58] For validation, overlapping expression of unbiased, computed cluster markers should be verified against known signature genes and spatial orientation by confocal microscopy 59,60 ( Figure 2A ). While histological cluster validation can be challenging and requires highly expressed marker genes and good antibodies, it achieves a reliable description of FRC subset heterogeneity as shown in a number of recent studies. 23, 27, 31, 53, 54 The molecular properties of FRC subsets as revealed by single-cell transcriptomics will be discussed in more detail in section 4. PRC Cd34, Ly6a phenotype of FRC subsets in order to secure niche-specific, optimal "catering" conditions. TRCs are involved in various crucial processes to establish special- PRCs, as their name suggests, are perivascular fibroblasts that fulfill reticular cell functions such as chemokine expression and conduit formation. 13 As perivascular cells ensheath the length of the vascular tree, single-cell transcriptomic analyses demarcate PRCs as CD34-and LY6A (stem cell antigen 1)-expressing cells surrounding blood vessels in each SLO. 23, 27, 53, 54 Further detailed studies are needed to resolve the molecular identity of PRCs along the vascular tree as has been done for PRCs in the CNS 90 or for lymph node blood endothelial cells. 91 Moreover, while the precursor potential of CD34 + PRCs has been demonstrated during development, 23 it remains unclear whether only a subset of PRCs exhibits precursor potential during SLO differentiation and whether the same or different PRC subsets maintain self-renewing properties in already formed SLOs, especially in the context of inflammation-induced remodeling. In lymph nodes, the medulla is a site of lymphocyte exit and myeloid cell accumulation, penetrated by large blood vessels and lymphatic sinuses. The lymph node vascular tree is rooted in the medulla with large arteries and veins traversing this region, 91 is required for FRC lineage commitment, 24,25 and a further "signal 2" drives subset differentiation 80, 106 (Figure 3 ). During embryonic development, splenic precursors emerge as mesenchymal condensation within the dorsal mesogastrium at Figure 3A ). The timed, conditional deletion of LTβR from Ccl19-iEYFP-targeted FRCs revealed a block in fate-mapped FRC subset differentiation, whereas mural cell specification and sustenance of a multipotent periarterial progenitor was found to be LTβR-independent. Based on these findings a two-signal program was proposed as model for splenic FRC subset specification. In this model, LTβR-dependent activation of mLTo cells serves as "signal 1" to commit to an FRC lineage, while further FRC subset specialization depends on secondary signals that likely reflect the immune microenvironment and extrinsic imprints from interacting cellular partners ( Figure 3B ). As the sustenance of adult reticular progenitors is independent of LTβR signaling, a yet undefined preceding signal is required to establish and maintain the periarteriolar progenitor niche. 23 Further cell-targeted genetic perturbation studies are required to delineate the nature of this initiating signal in the splenic white pulp. Depending on the bodily region, lymph node organogenesis in mice is initiated between E12 and E17 and is driven by sequential acti- 25, 111 and adipocyte precursor cells. 112 In line with this finding, it was shown that the genetic abrogation of the canonical NFκB pathway in Ccl19-Cre-expressing cells inhibits FRC differentiation, 113 and constant LTβR signaling throughout postnatal life is required for FRC differentiation. 114 Together these studies reinforce the notion that LTβR and NFκB signaling act as major switch for FRC subset differentiation, equivalent to the "signal 1" suggested for splenic FRCs. A recent study described YAP and TAZ, effectors of Hippo signaling, as additional factors that determine FRC lineage commitment prior to LTβR engagement 115 identifying this pathway as an initial signal preceding "signal 1" in FRC differentiation ( Figure 3B ). Since G-protein-coupled receptors, mechanical forces and Wnt signaling can feed into the YAP/TAZ signaling pathway, 116 further studies are warranted to explore the cellular and molecular triggers of the Hippo pathway in lymph node FRC precursors. Peyer's patch organogenesis is initiated at E16.5 when LTi cells ac- FDCs using a multicolor fate-mapping system in lymph nodes. 42 Although pharmacological studies suggest that the continued engagement of key pathways such as the LTβR and TNFR pathway are important to maintaining postnatal FRC subset identity, 98, 114 to what extent these progenitor-progeny relationships are maintained or reshaped in established SLOs over a lifetime of pathogen surveillance remains to be determined. In the course of the encounter with pathogens, lymphoid microenvironments increase in size through the recruitment and proliferation of immune cells leading to profound changes in the reticular cell network. 2 In addition to FRC proliferation, 74,120 topological changes in FRC network organization occur to support germinal center responses, 31 lymphangiogenesis, 121 Concomitantly, FRCs are involved in modulating adaptive immune responses via the expression of peripheral tissue antigens, 3 inhibitory molecules such as programmed death-ligand 1 (PD-L1), 54, 125 or metabolites such as nitric oxide or prostaglandin E2/cyclooxygenase-2 (PGE 2 /COX-2) in mice, [126] [127] [128] Although there are some inconsistencies in FRC subset characterization across studies (owing to differences in clustering resolution or cell input number), tremendous progress has been made in defining the molecular and functional FRC landscape in SLOs in murine models. It will be important to further elaborate FRC-driven regulation of lymphocyte activation and regulation as pioneered by Fletcher and colleagues who identified molecular mechanisms underlying FRC-restricted T cell activation as potential therapeutic strategies for CAR-T cells. 129 Clearly, future technological advances such as spatial transcriptomics 132 will facilitate the study of human FRCs, bridging knowledge and insights from the murine setting. As SLO FRCs can be regarded as the prototypical immune-interacting fibroblasts, a profound understanding of FRC activation and differentiation will meaningfully guide research on therapeutically targeting stromal cell-underpinned niches in autoimmunity and cancer. This study received financial support from the Swiss National Science Foundation/Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (grant 180011 to NBP and grants 177208 and 182583 to BL) and Novartis Foundation for Biomedical Research to (20C217 to NBP). The funders had no role in preparation of the manuscript. Therapeutics AG, St. Gallen, Switzerland. Burkhard Ludewig https://orcid.org/0000-0002-7685-573X Lymph node stromal cells: cartographers of the immune system Development and immunological function of lymph node stromal cells The fibroblastic T cell niche in lymphoid tissues Evolution of lymphoid tissues Form follows function: lymphoid tissue microarchitecture in antimicrobial immune defence Lymphocyte homing and homeostasis Chemokines and cell migration in secondary lymphoid organs A B-cell-homing chemokine made in lymphoid follicles activates Burkitt's lymphoma receptor-1 A chemokine-driven positive feedback loop organizes lymphoid follicles Molecular cloning of a novel human CC chemokine EBI1-ligand chemokine that is a specific functional ligand for EBI1, CCR7 Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells Podoplanin-rich stromal networks induce dendritic cell motility via activation of the C-type lectin receptor CLEC-2 Cords, channels, corridors and conduits: critical architectural elements facilitating cell interactions in the lymph node cortex Antigens in immunity. XV. Ultrastructural features of antigen capture in primary and secondary lymphoid follicles Localization of 125I-labeled antigen in germinal centers of mouse spleen: histologic and ultrastructural autoradiographic studies of the secondary immune reaction Ontogeny of reticular cells in the ileal Peyer's patch of sheep and goats Sophisticated strategies for information encounter in the lymph node: the reticular network as a conduit of soluble information and a highway for cell traffic Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes Fibroblastic reticular cells: organization and regulation of the T lymphocyte life cycle Reticular fibroblasts in peripheral lymphoid organs identified by a monoclonal antibody A distinct subset of fibroblastic stromal cells constitutes the cortex-medulla boundary subcompartment of the lymph node Restoration of lymphoid organ integrity through the interaction of lymphoid tissue-inducer cells with stroma of the T cell zone Origin and differentiation trajectories of fibroblastic reticular cells in the splenic white pulp Maturation of lymph node fibroblastic reticular cells from myofibroblastic precursors is critical for antiviral immunity Lymphatic endothelial cells control initiation of lymph node organogenesis Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus Fibroblastic reticular cell lineage convergence in Peyer's patches governs intestinal immunity Phenotypic and morphological properties of germinal center dark zone Cxcl12-expressing reticular cells Perivascular fibroblasts of the developing spleen act as LTalpha1beta2-dependent precursors of both T and B zone organizer cells Fibroblastic reticular cells regulate intestinal inflammation via IL-15-mediated control of group 1 ILCs Remodeling of light and dark zone follicular dendritic cells governs germinal center responses Depletion of stromal cells expressing fibroblast activation protein-alpha from skeletal muscle and bone marrow results in cachexia and anemia Fibroblastic reticular cells of the lymph node are required for retention of resting but not activated CD8+ T cells Mesenchymal cell targeting by TNF as a common pathogenic principle in chronic inflammatory joint and intestinal diseases CollagenVI-Cre mice: a new tool to target stromal cells in secondary lymphoid organs Follicular dendritic cell-specific prion protein (PrP) expression alone is sufficient to sustain prion infection in the spleen Identification of a new stromal cell type involved in the regulation of inflamed B cell follicles Stringent doxycycline dependent control of CRE recombinase in vivo Embryonic FAP(+) lymphoid tissue organizer cells generate the reticular network of adult lymph nodes Multicolor fate mapping of langerhans cell homeostasis Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system Fate mapping reveals origin and dynamics of lymph node follicular dendritic cells Type I interferon induces CXCL13 to support ectopic germinal center formation CCL19-producing fibroblastic stromal cells restrain lung carcinoma growth by promoting local antitumor T-cell responses Central nervous system stromal cells control local CD8(+) T cell responses during virus-induced neuroinflammation Exploring the single-cell RNA-seq analysis landscape with the scRNA-tools database Challenges in unsupervised clustering of single-cell RNA-seq data Computational and analytical challenges in single-cell transcriptomics Identifying cell populations with scRNA-Seq Sequencing depth and coverage: key considerations in genomic analyses Single cell RNA sequencing of rare immune cell populations Fibroblastic reticular cells at the nexus of innate and adaptive immune responses Single-cell RNA sequencing of lymph node stromal cells reveals niche-associated heterogeneity Type I interferon signaling in fibroblastic reticular cells prevents exhaustive activation of antiviral CD8(+) T cells A systematic performance evaluation of clustering methods for single-cell RNA-seq data Tutorial: guidelines for the computational analysis of single-cell RNA sequencing data Current best practices in single-cell RNAseq analysis: a tutorial Single-cell RNA sequencing technologies and bioinformatics pipelines The technology and biology of single-cell RNA sequencing The evolving concept of cell identity in the single cell era Scaling single-cell genomics from phenomenology to mechanism Defining cell types and states with single-cell genomics Identifying gene expression programs of cell-type identity and cellular activity with single-cell RNA-Seq Computational analysis of cell-to-cell heterogeneity in single-cell RNA-sequencing data reveals hidden subpopulations of cells Computational assignment of cell-cycle stage from single-cell transcriptome data Validation of noise models for single-cell transcriptomics Cell MixS: quantifying and visualizing batch effects in single-cell RNAseq data Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors muscat detects subpopulation-specific state transitions from multi-sample multi-condition single-cell transcriptomics data B cell homeostasis and follicle confines are governed by fibroblastic reticular cells Lymph node fibroblastic reticular cells directly present peripheral tissue antigen under steady-state and inflammatory conditions Fibroblastic reticular cells enhance T cell metabolism and survival via epigenetic remodeling Neonatally imprinted stromal cell subsets induce tolerogenic dendritic cells in mesenteric lymph nodes Topological small-world organization of the fibroblastic reticular cell network determines lymph node functionality Topological structure and robustness of the lymph node conduit system Plasma cell output from germinal centers is regulated by signals from Tfh and stromal cells Class-switch recombination occurs infrequently in germinal centers Development and migration of plasma cells in the mouse lymph node Long-term antigen retention by dendritic cells in the popliteal lymph node of immunized mice Follicular dendritic cells emerge from ubiquitous perivascular precursors Germinal center dark and light zone organization is mediated by CXCR4 and CXCR5 Organizer-like reticular stromal cell layer common to adult secondary lymphoid organs Marginal reticular cells: a stromal subset directly descended from the lymphoid tissue organizer Structure and function of the immune system in the spleen CD169+ macrophages at the crossroads of antigen presentation Activated Peyer's patch B cells sample antigen directly from M cells in the subepithelial dome Structure and function of the spleen Blood vessels of the Peyer's patch in the mouse: I. Topographic studies Context-dependent development of lymphoid stroma from adult CD34(+) adventitial progenitors A molecular atlas of cell types and zonation in the brain vasculature A molecular map of murine lymph node blood vascular endothelium at single cell resolution Lymphatic endothelial cells of the lymph node Lymph node stromal CCL2 limits antibody responses Identification of a new subset of lymph node stromal cells involved in regulating plasma cell homeostasis Development and maturation of secondary lymphoid tissues Development of secondary lymphoid organs The lymphotoxin beta receptor controls organogenesis and affinity maturation in peripheral lymphoid tissues Turning off follicular dendritic cells Abnormal development of peripheral lymphoid organs in mice deficient in lymphotoxin Distinct roles in lymphoid organogenesis for lymphotoxins alpha and beta revealed in lymphotoxin beta-deficient mice Dissecting the role of lymphotoxin in lymphoid organs by conditional targeting A fresh view on lymph node organogenesis Distinct role of surface lymphotoxin expressed by B cells in the organization of secondary lymphoid tissues B cells are crucial for both development and maintenance of the splenic marginal zone Lymphoid tissue inducer cells and the evolution of CD4 dependent high-affinity antibody responses Cellular source and molecular form of TNF specify its distinct functions in organization of secondary lymphoid organs The dynamics of spleen morphogenesis Development and function of the mammalian spleen Lymph node mesenchymal and endothelial stromal cells cooperate via the RANK-RANKL cytokine axis to shape the sinusoidal macrophage niche Ontogeny of stromal organizer cells during lymph node development Endothelial cell-specific lymphotoxin-beta receptor signaling is critical for lymph node and high endothelial venule formation Lymphotoxin-beta receptor signaling through NF-kappaB2-RelB pathway reprograms adipocyte precursors as lymph node stromal cells Essential role of canonical NF-kappaB activity in the development of stromal cell subsets in secondary lymphoid organs Early-life programming of mesenteric lymph node stromal cell identity by the lymphotoxin pathway regulates adult mucosal immunity YAP/TAZ direct commitment and maturation of lymph node fibroblastic reticular cells Hippo-YAP/TAZ signalling in organ regeneration and regenerative medicine Peyer's patch inducer cells play a leading role in the formation of B and T cell zone architecture IL-7 receptor alpha+ CD3(-) cells in the embryonic intestine induces the organizing center of Peyer's patches Effect of mature lymphocytes and lymphotoxin on the development of the follicleassociated epithelium and M cells in mouse Peyer's patches Infection programs sustained lymphoid stromal cell responses and shapes lymph node remodeling upon secondary challenge Interactions between fibroblastic reticular cells and B cells promote mesenteric lymph node lymphangiogenesis Lymphotoxin-dependent B cell-FRC crosstalk promotes de novo follicle formation and antibody production following intestinal helminth infection Replicating viral vector platform exploits alarmin signals for potent CD8(+) T cell-mediated tumour immunotherapy Fibroblastic reticular cells initiate immune responses in visceral adipose tissues and secure peritoneal immunity Viral targeting of fibroblastic reticular cells contributes to immunosuppression and persistence during chronic infection Attenuation of chronic antiviral T-cell responses through constitutive COX2-dependent prostanoid synthesis by lymph node fibroblasts Regulated release of nitric oxide by nonhematopoietic stroma controls expansion of the activated T cell pool in lymph nodes Fibroblastic reticular cells of the lymphoid tissues modulate T cell activation threshold during homeostasis via hyperactive cyclooxygenase-2/prostaglandin E2 axis The human lymph node microenvironment unilaterally regulates T-cell activation and differentiation Functional role of type I and type II interferons in antiviral defense Recombinant vesicular stomatitis (Indiana) virus expressing New Jersey and Indiana glycoproteins induces neutralizing antibodies to each serotype in swine, a natural host Computational methods for single-cell omics across modalities FDC-specific functions of p55TNFR and IKK2 in the development of FDC networks and of antibody responses Differentiation and activation of fibroblastic reticular cells