LN-Derived Fibroblastic Reticular Cells: Comparison
Please note this is a comparison between Version 4 by Enzi Gong and Version 11 by Enzi Gong.

Lymph node structural organization is reported to be governed by the stromal cells. Fibroblastic reticular cells (FRCs), a subset of the stromal cells found inusually found and isolated from the T lymphocyte regioncell zone of lymph nodes (LNs) and other secondary lymphoid organs (SLOs), have , have recently been described as much more than structural cells.

FRCs are described to be organized in a conduit system called the “reticular fiber network”, responsible for transferring antigens from tissue to T cell zones in LNs and for controlling the conduit matrix deposition during lymph node expansionple structural cells. 

  • fibroblastic reticular cells
  • T cells
  • lymph nodes

1. Overview

Fibroblastic reticular cells (FRCs), usually found and isolated from the T cell zone of lymph nodes, have recently been described as much more than simple structural cells. Originally, these cells were described to form a conduit system called the “reticular fiber network” and for being responsible for transferring the lymph fluid drained from tissues through afferent lymphatic vessels to the T cell zone. However, nowadays, these cells are described as being capable of secreting several cytokines and chemokines and possessing the ability to interfere with the immune response, improving it, and also controlling lymphocyte proliferation. Here, we performed a systematic review of the several methods employed to investigate the mechanisms used by fibroblastic reticular cells to control the immune response, as well as their ability in determining the fate of T cells. We searched articles indexed and published in the last five years, between 2016 and 2020, in PubMed, Scopus, and Cochrane, following the PRISMA guidelines. We found 175 articles published in the literature using our searching strategies, but only 24 articles fulfilled our inclusion criteria and are discussed here. Other articles important in the built knowledge of FRCs were included in the introduction and discussion. The studies selected for this review used different strategies in order to access the contribution of FRCs to different mechanisms involved in the immune response: 21% evaluated viral infection in this context, 13% used a model of autoimmunity, 8% used a model of GvHD or cancer, 4% used a model of Ischemic-reperfusion injury (IRI). Another four studies just targeted a particular signaling pathway, such as MHC II expression, FRC microvesicles, FRC secretion of IL-15, FRC network, or ablation of the lysophosphatidic acid (LPA)-producing ectoenzyme autotaxin. In conclusion, our review shows the strategies used by several studies to isolate and culture fibroblastic reticular cells, the models chosen by each one, and dissects their main findings and implications in homeostasis and disease.  

2. Background

Lymph node structural organization is reported to be governed by the stromal cells [1]. Fibroblast reticular cells (FRCs), a subset of the stromal cells found in the T lymphocyte region of lymph nodes (LNs) and other secondary lymphoid organs (SLOs), have been described as much more than structural cells [2].
FRCs are described to be organized in a conduit system called the “reticular fiber network”, responsible for transferring antigens from tissue to T cell zones in LNs and for controlling the conduit matrix deposition during lymph node expansion [3][4].
Their ability for cytokine and chemokine production has been demonstrated in several studies [2][5][6], and their relevant multifunctional roles and multiple subsets have been previously defined [7]. In addition, mice and human lymph node-derived FRC’s ability to react to inflammatory stimuli has been described [8][9][10].
Moreover, a few studies have implicated FRC in peripheral tolerance. Certain stromal cells can express antigens from peripheral tissues (PTA) and mediate the maintenance of peripheral tolerance through the deletion of self-reactive T cells and other mechanisms [11][12][13][14][15][16]. In addition, other cells previously known as structural cells, such as epithelium, endothelium, and fibroblasts, have also been implicated as players in the immune response [17].
However, there are several obscure points in FRC biology that need elucidation, mainly that of their dual role augmenting and, thereby, controlling the immune response. In this sense, this systematic review lists several FRC mechanisms described as controlling mechanisms of the immune response [18][19][20][21][22][23][24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39][40][41].
The articles reviewed here report on using several animal models of disease and/or genetically modified mice as tools to investigate FRCs’ effect on T cells. These articles also approach and clarify the mechanisms involved in T cell proliferation or differentiation in subsets with regulatory, effector, or memory profiles [18][19][20][21][22][23][24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39][40][41]. In addition, these articles reported the markers used to identify and isolate FRCs, as well as the methods used for these cells’ cultivation.
Lately, FRCs’ ability for controlling the immune response and its role in several pathological conditions, such as viral infection, inflammation, metastatic cancer, and autoimmunity, are also included in this review. Consequently, we comprise here the latest updates in FRC biology, their impact on T cell fate, how they participate in diseases, and how they could be manipulated in order to ameliorate the course of certain conditions.

3. Conclusions

FRC is a specific subset of stromal cells present in the lymph node, and they are precisely located in the T cell zone. There are other stromal cell subsets in lymph nodes, described as double-negative cells, follicular dendritic cells, blood endothelial cells, lymphatic endothelial cells, and others that are not discussed in this review [42]. The results of this review firstly show the characteristics of the host type used for analyzing FRC function. In addition, strategies used by them in order to achieve their target objectives, including model characteristics, such as source, genotype, age, and gender, are described in Table 1. The main characteristics of FRCs, their origin, as well the lymph node (LN) digestion process, and techniques used for their isolation are described in Table 2. The immune cell sources, as well as their characteristics, are described in Table 3. All these variations between the models studied, cell origins, and characterization, sometimes lead to different conclusions, making the comparison between studies difficult or conflicting, such as the role of FRCs in T cell proliferation, sometimes described as stimulators and, at other times, as limiting. Next, we assembled the studies with the same subject (Table 4) and compared them, trying to show the differences and, more importantly, comparisons between the achieved results (Figure 1).
Table 1. Characteristics of hosts and the interventions that they received before in vitro analysis.
Ref.YearHostInterventions
SourceGenotypeAge (Weeks)GenderTypeTime (Days)
         
Table 2. Characteristics of fibroblastic reticular cells isolation and their immunophenotype.
Ref.Lymph Node RegionDigestion TypeDigestion SolutionFRC Culture Medium + SupplementFRC Immunophenotypic CharacterizationTechnique for Cell SeparationPurity

(%)
Aparicio-Domingo et al. [18]2020Mice C57BL/6JIL-33gfp/gfp; IL-33gfp/+7–19MLCMV clone 13 and WE virus; tamoxifenSingle dose;

6 (3/week)
Dertschnig et al. [19]2020Mice C57BL/6Female to male bone marrow transplant model (BMT), T cell-depleted, plus transgenic TCR-CD8 MataHari (Mh)NRM
Brown et al. [
24
Mice BALB/c
Aparicio-Domingo et al. [18]Axillary; brachial; inguinalEnzymaticCollagenase IV; DNase I; CaCl2DMEM (2% FCS)CD45; CD31; PDPNCell sortingDexamethasone; DT; Gy irradiation
CD4, CD8, B220, CD44
Abbreviations:—Ref.: reference; NR: not reported; NA: not applicable; FCS: fetal calf serum; PP: Peyer’s Patches; LN: lymph nodes; SLOs: secondary lymphoid organs; DCs: dendritic cells; NK: natural killer cells.
 
 
 
 
 
 
 
Table 4. Main characteristics of the studies used to assess the influence of fibroblastic reticular cells on the activation, expansion, or suppression of immune responses.
Ref.Trial TypesStudy TargetTime of InterventionMain Performed EvaluationsResultsFRC Role in Immune Response
3; 4
]LNCD8+ T cellsNon selection performedDMEM (2% FCS)CD45, CD8α, CD4, TRCαβ
Aparicio-Domingo et al. [18]IL-33-GFP reporter miceLCMV3 days/w

for 2 weeks
FC and RNA sequencingFRC is one important IL-33 source in LNs, vital for driving acute and chronic antiviral T cell responses.Anti-viral response>94Dertschnig et al. [19]LNT cellsCD3 negative selection followed by

CD4 and CD8α

positive selection

(MicroBeads—Myltenyi)
NR
Dertschnig et al. [CD45, CD45.1, CD3, CD4, CD8α, CD62L, CD44, CD69, CD127, Vα2, Vβ5
19]FRC and DC ablation in vivo; identification of PTA regulatory genes; BTM model inductionGvHD2 weeksFC, RNA sequencing, confocal microscopyThe loss of PTA presentation by FRCs during GVHD leads to permanent damage in their networks in lymphoid tissues.Control of peripheral toleranceEom et al. [20]2020HumanEom et al. [20]LNNAMetastatic melanoma and surgeryNANANANANA
NA
Eom et al. [20CD45, CD3, CD8
]Identification of distinctive subpopulations of CD90+ SCs present

in melanoma-infiltrated LNs
MelanomaNAFC, gene expressionThere are several distinct subsets of FRCs present in melanoma-infiltrated LNs. These FRCs may be related to cancer metastasis invasion and progression by avoiding T cells through secreted factors.Lymph node invasion metastasis and its correlation with FRC gene expression.Gonzalez et al. [21]Gonzalez et al. [21]SpleenCD8+ T cellsCD8 isolation by negative selection (Microbeads—MojoSort)
Gonzalez et al. [21]2020Mice (NOD/ShiLtJ, NOR/LtJ, and NOD.CgTg); HumanType 1

diabetes
12FNATissue-engineered stromal reticula and FRC/T cell co-cultureType 1

NA
NRdiabetesNACD45, CD8, CD44, CD25
FC, immunofluorescence, imagingFRCs modulate their interactions with autoreactive T cells by remodeling their reticular network in LNs. FRC with decreased contractility through gp38 downregulation, can loosen/relax their network, potentially decreasing FRC tolerogenic interactions with autoreactive T cells and promoting their escape from peripheral regulation in LNs.Role of FRCs on tolerance and T1DKnop et al. [22]2020Mice C57BL/6N and ROSA26RFPIL-7−/−, PGK-Cre, FLPO, RAG1−/−, Thy1.1+ OT-INRNRNANA
Knop et al. [22]LN; spleenT cells and NKCD8α positive selection (MicroBeads—Myltenyi)CD45, CD3, CD4, CD5, CD8α, CD62L, Bcl-2, CD127, Nk1.1, RORγt
Knop et al. [22]IL-7fl/fl mice and adoptive T cell transferNANAFCIL7, produced by LN FRCs-regulated T cell homeostasis, is crucial for TCM maintenance.IL7 produced by LN FRCs is crucial for TCMPerez-Shibayama et al. [23]2020Mice C57BL/6CCL19-Cre IFNARfl/fl8–10NRLCMV ArmstrongNR
maintenancePerez-Shibayama et al. [23]LN; spleenT cell subsets and exhaustionNo selection performedRPMICD45.1, CD45.2, CD45R, CD8α, CD8β, CD3e, CD44 CD62L, PD-1, PDL1Brown et al. [
Perez-Shibayama et al. [23]LCMV-infected mice, FRC ex vivo restimulation and cytokine productionLCMV Armstrong8 dFCIFNAR-dependent shift of FRC subsets toward an immunoregulatory state reduces exhaustive CD8+ T cell activation.IFN type 1 influences FRC peripheral tolerance24]]2019Mice C57BL/6IL-6−/−; NOS2−/−5–12MPR8-GP33-41, LCMV, influenza, OT-1 T cells with OVANR
NREnzymaticCollagenase P; DNase I; Dispaseα-MEMBrown et al. [CD45; CD31; PDPNCell sorting24>95]LNCD8+ T cellsCD8α positive selection (MicroBeads—Myltenyi)RPMI; α-MEMCD45.1, CD45.2, CD3, CD4, CD8, CD275, CD28, CD44Kasinath et al. [25]2019Mice CD-1 IGS or C57BL/6 or C57BL/6JCCL19-Cre iDTR8–10MNephrotoxic serum (NTS); DT3
Kelch et al. [26]2019Mice C57BL/6J
Brown et al. [24]NA9–22MNANA
FRC/T cell co-culturesInfluenza and LCMV infectionNRFC and RNA sequencingFRCs play a role over restricting T cell expansion—they can also outline the fate and function of CD8+ T cells through their IL-6 production.Majumder et al. [27]2019Mice C57BL/6IL-17A−/−; IL-17RAfl/fl; OT-II, ACT1−/−; CCL19-Cre; IL23R−/−; Regnase1+/-6–12M-FMOG with Mycobacterium tuberculosis, pertussis toxin on/OT-II CD4+ T cells with OVA/DSS2
Masters et al. [28]2019Mice C57BL/6RAG−/−; CD45.12–4 m and 19–21 mMInfluenzaNR
FRCs influence the CD8 T cells fateKasinath et al. [25]LN; spleenCD4+ T cellsNo selection performedNRCD45, CD3, CD4, CD44, CD62L, IL-17ASchaeuble et al. [29]2019Mice C57BL/6NOS2−/−; OT-1; COX2−/−, COX2ΔCCL19Cre, and ROSA26-EYFPCCL19Cre≥6NROVA and poly (I:C)4
Dubrot et al. [30]2018Mice C57BL/6CIITA−/−; pIV−/−; K14 TGP IVKO; RAG2−/− PROX-1-Cre MHC-IIfl>12mNRTamoxifen; IFN-γ and FTY7204 (Twice/day); 6
Knoblich et al. [31]2018
Kasinath et al. [25]Mouse FRC depletion and treatment with anti-PDPN antibodyCrescentic Glomerulonephritis (GN)3 dFC and gene expressionRemoval of kidney-draining lymph nodes, depletion of fibroblastic reticular cells, and treatment with anti-podoplanin antibodies each resulted in the reduction of kidney injury in GN.Kelch et al. [26]LNNANANANA
Role of FRCs and PDPN expression in GN
Kelch et al. [26]3D imaging and topological mappingNANAEVIS imaging and confocal microscopyT cell zones showed homogeneous branching, conduit density was significantly higher in the superficial T cell zone compared with the deep zone. Although the biological significance of this structural segregation is still unclear, independent reports have pointed to an asymmetry in cell positioning in both zones. Naive T cells tend to

occupy the deep TCZ, whereas memory T cells preferentially locate to the superficial zones,

and innate effector cells can often be found in the interfollicular regions.
FRC conduits and their distribution inside LNsMajumder et al. [27]LNT and B cellsNR
Majumder et al. [27NR]Metabolic assayExperimental autoimmune encephalomyelitis7 dFC, immunoblotting, siRNA transfectionDuring Th17 differentiation in LNs, IL-17 signals to FRCs and impacts LN stromal organization by promoting FRC activation through a switch on their phenotype from quiescence to highly metabolic.FRCs are impacted by metabolic alterations driven by IL-17
Masters et al. [28]FRC-mediated T cell proliferation inhibition and T cell survival assaysAging and influenza infectionNRFCAge-related changes in LN stromal cells may have the largest impact on the initiation of the immune response to influenza infection, and may be a factor contributing to delayed T cell responses to this virus.Aging impacts the adaptive anti-viral immune response initiation in LNHumanCadaveric donorsNANANAFOXP3hCD2xRAG2−/− xD011.10NRM-FNANA
Valencia et al. [36]2017HumanBrain-dead organ donorsNAM-FNANA
Yu, M. et al. [37]2017Mice C57BL/6 and HumanPTGS2Y385F/Y385F; OVA-specific CD8 (OT-I); CD4 (OT-II)4–6NRDC-vaccine1.5
Gil-Cruz et al. [38]2016Mice C57BL/6N or C57BL/6N-Tg or R26R-EYFPMyd88−/−; TLR7−/−; CCL19-Cre8–10NRMHV A59; Citrobacter rodentium12; 6
Novkovic et al. [39]2016Mice C57BL/6N or C57BL/6N-TgCCL19-Cre; iDTR6–9NRDT3 and 5
Dertschnig et al. [19]Peripheral; mesentericEnzymaticDNase; LiberaseNCCD45; CD31; PDPNCell sortingNR
Eom et al. [20]Axillary; inguinal; cervical; mesenteric; mediastinumEnzymaticDNase I; Liberase DHRPMI-1640CD45, CD31, PDPNNRNR
Gonzalez et al. [21]Skin-draining (brachial; axillary; inguinal); PancreaticEnzymaticCollagenase P; DNase I; Dispase IINRCD45; CD31; PDPNCell sortingNR
Knop et al. [22]Peripheral; mesentericEnzymaticCollagenase P; Dispase II; DNase I; Latrunculin BRPMI-1640CD45; CD31; PDPNCell sorting>73.3
Perez-Shibayama et al. [23]InguinalEnzymaticCollagenase F; DNase IRPMINRNRNRRoyer et al. [40]2016Mice C57BL/6 or Gbt-1.1CXCL10−/−; CXCR3−/−; STING−/−; CD18−/−6–12M-FHSV-1NR
Takeda et al. [41]2016Mice C57BL/6JLPAR2−/−; ENPP2-flox, CCCL19-Cre, LPAR5−/−
Kasinath et al. [25]KidneyNANANANANANA
Kelch et al. [26]Popliteal; mesenteric; InguinalNANANANANANA
Majumder et al. [27]Mesenteric; inguinalEnzymaticDNase I; Liberase; DispaseRPMICD45; CD31; PDPN;Microbeads isolation>98
Masters et al. [28]Mesenteric;

popliteal
EnzymaticLiberase TL; BenzonucleaseRPMI-1640CD45; CD31; PDPNMicrobeads isolation>90
Schaeuble et al. [29]
Schaeuble et al. [29Peripheral (axillary, brachial, inguinal)]Nos2Enzymatic−/−Collagenase IV; DNase IDMEM (2% FCS), COX2−/−CD45; CD31; PDPN mice and FRC/T cell co-cultureMicrobeads isolation≥90
COX/Prostaglandin E2 pathway4 dFCFRCs constitutively express high levels of COX2 and its product PGE2, thereby identified as a mechanism of T cell proliferation control.PGE2 and COX2 pathways in FRCs are implicated in the control of T cell proliferationDubrot et al. [30]Skin-drainingEnzymaticCollagenase D; DNase IHBSSCD45; CD31; PDPNCell sortingNRNA
Dubrot et al. [30]Adoptive transfer T cells in RAG−/− mice and Treg suppression assayMHC II-induced expression by FRC and LEC and its impact on autoimmunity5 dFCLNSCs inhibit autoreactive T-cell responses by directly presenting antigens through endogenous MHCII molecules.Control of peripheral tolerance in autoimmunityKnoblich et al. [31]NREnzymaticCollagenase P; DNase I; Dispaseα-MEM (10% FBS)CD45; PDPNNR99
Knoblich et al. [31]T cell and CAR T cell activation assayCOX/Prostaglandin E2, iNOS, IDO and TGF-β pathways in FRCs,NAFC and RNA sequencingMaaraouf et al. [32]
Maaraouf et al. [322018]KidneyMice C57BL/6CCL19-Cre; iDTR; RAG1−/−NRNRDT; LTβr-Ig1; 2
FRCs block proliferation and modulate differentiation of newly activated naïve human T cells, without requiring T cell feedback.FRCs used several pathways to control T cell proliferation
Maaraouf et al. [32]EnzymaticCollagenase P; DNase I; Dispase IIDMEM (10% FBS)FRC labeling and injection into miceIschemic-reperfusion injury (IRI)CD45; CD31; PDPNNRNRNRFC, electron and confocal microscopyDepletion of FRCs reduced T cell activation in the kidney LNs and ameliorated renal injury in acute IRI.Role of FRCs in IRIChung et al. [33]2017Mice BALB/c or C57BL/6
Chung et al. [33TgMx1-Cre; DLL1
Chung et al. [33]fl/fl; DLL4fl/fl; NOTCH2fl/fl; RAG1−/−6–10 or 8–12M-FFRC/T cell co-culturepoly (I:C)/8.5-9 Gy;

poly (I:C)/6 Gy irradiation
0.16; 0.12
]Peripheral (cervical, axial, brachial, inguinal)GvHD4 h and 3 hFCFRCs delivered NOTCH signals to donor alloreactive T cells at early stages after allo-BMT to program the pathogenicity of these T cells.Role of FRC NOTCH-signaling in activating alloreactive T cells
Gao et al. [34]FRC expression and secretion of Interleukin 7Tumor-draining LNsNAFCLN tumor-infiltrating cells decreased the FRC population and IL-7 secretion, leading to declined numbers of T cells in TDLNs. This may partly explain the weakened ability of immune surveillance in TDLNs.Role of IL-7 secretion by FRCs and its impact on tumor-draining LNs
Pazstoi et al. [35]Treg induction in presence of FRC microvesicles.FRC microvesicles (MVEs)NAFC and RNA sequencingStromal cells originating from LNs contributed to peripheral tolerance by fostering de novo Treg induction by MVEs carrying high levels of TGF-β.Role of FRC MVEs in inducing peripheral tolerance
Valencia et al. [36]FRC/T cell co-cultureCOX 2/Prostaglandin E2, iNOS, IDO and TGF-β pathways in FRCs6 hFCCOX2 expression was detected in human FRCs but was not considerably upregulated after inflammatory stimulation, concluding that human and murine FRCs would regulate T lymphocytes responses using different mechanisms.Role of FRCs integrating innate and adaptive immune responses and balancing tolerance and immunogenicity
Yu, M. et al. [37]FRC/T cell co-cultureCOX 2/Prostaglandin E2 pathway in FRCsNAFC, WBHyperactivity of COX-2/PGE2 pathways in FRCs is a mechanism that maintains peripheral T cell tolerance during homeostasis.PGE2 and COX2 pathways in FRCs are implicated in the control of T cell proliferation.
Gil-Cruz et al. [38]ILC1 and NK cells regulationFRC secretion of IL-153 hFCFRC secretion of IL-15 regulates homeostatic ILC1 and NK cell maintenance.Role of FRCs in innate in immunity
Novkovic et al. [39]Royer et al. [40]MandibularMechanical disruptionNRRPMI-1640 (10% FBS)NRNRNR
Takeda et al. [41]Mesenteric; peripheral; brachialEnzymaticCollagenase P; Dispase; DNase IRPMI-1640CD45; CD31; PDPNCell sortingNR
Abbreviations—Ref.: reference; NR: not reported; NA: not applicable; FBS: fetal bovine serum; FCS: fetal calf serum; PDPN or gp38: podoplanin; NC: not cultivated.
 
 
 
Table 3. Characteristics of the main type of immune cells used for analysis with fibroblastic reticular cells.
Ref.Source of CellsCell TypeSeparation TechniqueImmune Cell Preservation Solution and SupplementationImmune Cell Immunophenotypic Characterization
Aparicio-Domingo et al. [18
RPMI
CD45, CD45.2, CD4, B220, IL-17A, IL-17R
Masters et al. [
28
]LN; peripheral bloodCD8+ TCD8 isolation by negative selection (Microbeads—MojoSort)NRCD45, CD45.1, CD45.2, CD69, CD8α
Schaeuble et al. [29]LN; spleenT cellsNo selection performedRPMICD45, CD3, CD4, CD8α, CD44, CD62L, CD279, FoxP3, CD25
Dubrot et al. [30]LN; spleenT cells, B cells, Treg, and DCPan T isolation by negative selection

(MicroBeads—Myltenyi)
NRCD45, CD44, CD3, CD4, CD8α, FOXp3, Ly5.1, CD11b, CD19, CD25, CD62L, PDCA-1, PD-1, IL-17, IFNγR
Knoblich et al. [31]LN; tonsilsT cellsPan T isolation by negative selection

(MicroBeads—Myltenyi)
NRCD45, CD3, CD4, CD8, CD62L, CD27, CD45RO, CD25
Maaraouf et al. [32]SpleenT cellsPan T isolation by negative selection

(MicroBeads—Myltenyi)
NRCD45, CD4EnzymaticCollagenase IV; DNase IDMEM (2% FBS)CD45; CD31; PDPNCell sorting
Chung et al. [NR33]Spleen; peripheral bloodT cells, B cells, FDCs, Treg, and DCsT cell Thy.1 selection

(Microbeads—StemCells Technologies)
NACD45, CD3, CD4, CD8, FOXp3, CD157, CD19, B220, CD44, CD62L, CD11c, CD11b, CD169, CD21/35, F4/80, TCRβGao et al. [34]2017Mice C57BL/6 and HumanColon cancer6FLewis Long carcinoma cellsNA
Gao et al. [34]InguinalEnzymaticCollagenase IV; DNase IRPMI-1640 (2% FBS)
Gao et al. [CD45; CD31; PDPN34]LNT cellsNRNANRNACD45, CD4, CD8Pazstoi et al. [35]
Pazstoi et al. [35]2017MesentericEnzymaticCollagenase P; Dispase; DNase IRPMI-1640CD45; CD31; PDPNCell sorting91–97
Pazstoi et al. [35]LNT cellsCD4 positive selection

(Microbeads—Myltenyi)
EX VIVOCD45, CD45.2, CD4, CD2, CD9, CD24, CD25, CD63Valencia et al. [36]MesentericMechanical disruptionNRRPMI-1640CD45, CD31, PDPNNR
Valencia et al. [NR
36]LNCD4+ T cellsCD4 naïve T cell negative selection

(Microbeads—Myltenyi)
RPMI (10% FCS)CD45, CD44, CD4Yu, M. et al. [37]Axillary; brachial; inguinal
Yu, M. et al. [37Enzymatic]LNCollagenase P; Dispase; DNase IDMEM (10% FBS)T cellsPan T cell negative selection

(Microbeads—StemCells Technologies)CD45; CD31; PDPN
Cell sorting>95
RPMI (10% FBS)CD45, CD45.1, CD45.2, CD3, CD4, CD8α, CD25, CD69, CD44Gil-Cruz et al. [38]MesentericEnzymatic
Gil-Cruz et al. [38]PP; LNT cells, B cells, NK cells, Treg, and ILCsCollagenase D; DNase IRPMI-1640 (2% FCS)CD45; CD31; PDPNNRRPMI (10% FCS)Cell sortingCD45, CD3e, CD4, CD8α, EOMES, FoxP3, B220, CD19, CD127, CD62L, CD44, CD69, F4/80, IL-17A, IL-7Rα, GATA3, RORγt, IL-15RαIL-15Rβ, NKp46, NK1.1NR
Novkovic et al. [39]InguinalEnzymaticCollagenase P; DNase IRPMI (2% FCS)PDPNNA
Novkovic et al. [39]LN; SpleenDCs and T cellsNRNARPMI (2% FCS)CD45, CD3, CD8, CD4, CD11c, MHCII
FRC network topological analysisFRC networkNAIntravital TPM with morphometric 3D reconstitution analysis.Physical scaffold of LNs formed by the FRC network is critical for the maintenance of LN functionality.FRC network disruption impacts the immune responseRoyer et al. [40]SLOsCD8+ T cellsCD8 positive selection (Microbeads—Myltenyi)RPMI (10% FBS)CD45, CD3, CD4, CD8
Royer et al. [40]Adoptive transfer of T cells and T cell response to herpesvirus-associated lymphadenitisHSV-14 hFCDissemination of the virus to secondary lymphoid organs impairs HSV-specific CD8+ T cell responses by driving pathological alterations to the FRCs conduit system, resulting in fewer HSV-specific CD8+ T cells in circulation.Role of FRC in virus-specific T CD8 response; LPAR6−/−8–12NRCD4+ T cells labeled with CMTMR; LTβR-Fc0.6; 1.04; 28
Abbreviations—Ref.: reference; NR: not reported; NA: not applicable; M: male; F: female; DT: diphtheria toxin; NTS: nephrotoxic serum; DC: dendritic cells; LCMV: lymphocytic choriomeningitis virus; WE: lymphocytic choriomeningitis virus strain WE; MOG: myelin oligodendrocyte glycoprotein; OVA: ovalbumin; HSV-1: herpes simplex virus 1; DSS: dextran sodium sulfate colitis; MHV: mouse hepatitis virus; CMTMR: cell tracker; LTβR: lymphotoxin-β receptor; FTY720: immunomodulator, IL: interleukin, TCR: T cell receptor, RAG1: recombination activating gene 1; IFNAR: interferon-α/β receptor; NOS2: nitric oxide synthase 2; CCL19: chemokine (C-C motif) ligand 19; PGK: phosphoglycerate kinase 1; FLPO: is an artificial derivative of the recombinase encoded by the saccharomyces cerevisiae 2μ plasmid; Thy1.1: thymus cell antigen 1.1; OT-I: ovalbumin TCR-I; OT-II: ovalbumin TCR-II; iDTR: inducible diphtheria toxin receptor; ACT1: adaptor for IL-17 receptors; COX2: cyclooxygenase-2; EYFP: enhanced yellow fluorescent protein; CIITA: class II transactivator factor; pIV-promoter IV, MHC-II: major histocompatibility complex class II; PROX1- prospero homeobox 1; DLL: delta; FOXP3: forkhead box P3; PTGS2: prostaglandin endoperoxide synthase 2; Myd88: myeloid differentiation primary response 88; TLR7: toll-like receptor 7; CXCL10: C-X-C motif chemokine ligand 10; CXCR3: C-X-C motif chemokine receptor 3: STING: stimulator of interferon response; LPAR: lysophosphatidic acid receptors; ENPP2: ectonucleotide pyrophosphatase/phosphodiesterase 2; IFN-γ: interferon gamma; Gy: gray.
Takeda et al. [
41
]
LN; Spleen
T cells, B cells
Takeda et al. [
41
CD4 naïve T cell negative selection


(Microbeads—Myltenyi)
RPMI
]
Lymphocyte migration
Ablation of LPA-producing ectoenzyme autotaxin in FRCsNAFC, IMS, Intravital TPMLPA produced by LN FRCs acts locally to LPA2 to induce T cell motility.Role of FRCs in T cell local migration
Abbreviations—Ref.: reference; NR: not reported; NA: not applicable; FC: flow cytometry; WB: Western blotting; IMS: imaging mass spectrometry; Intravital TPM: intravital two-photon microscopy.
 
Figure 1. Schematic illustration of lymph nodes, FRC localization, and their role on lymphocytes in different scenarios of the immune response: (A) Viral infection, (B) Inflammation, (C) Autoimmunity, (D) Metastatic cancer, (E) Homeostasis, (F) GvHD.
The first scenario discussed was on viral infection (Figure 1A). Aparicio-Domingo et al., in an LCMV study, concluded that FRCs displayed a stimulatory role, being a main source of IL-33 in the lymph node and crucial for leading to acute and chronic antiviral T cell responses. They also showed that FRCs mainly act on CD8 T lymphocytes by signaling via ST2 expressed by these T cells [18]. Severino et al. demonstrated previously, in 2017, the increased IL-33 gene expression in human FRCs after treatment with IFN-γ or IL-1β and TNF-α. These cytokines are usually released during a course of an immune response, supporting the Aparicio-Domingo et al. findings that FRCs are the main source for IL-33 [9]. Perez-Shibayama et al., using the LCMV model like Aparicio-Domingo et al., commented that FRCs contributed to an immunostimulatory state to prevent virus replication and spread. However, they also found a regulatory role of FRCs, showing an IFN-α-signaling dependent shift of FRCs toward an immunoregulatory state, reducing exhaustive CD8 T lymphocyte activation. They claim that type 1 IFN-mediated control of LCMV replication in FRCs is one of the major factors that determine the quality of the antiviral CD8+ T cell response [23]. In agreement, Talemi and Hofer sustain the idea that interferons delay the viral spread in infection, acting as sentinels, warning uninfected cells, and also are negative feedback regulators acting at a single-cell level [43]. Regarding the anti-viral response for influenza and LCMV, Brown et al. [24] showed that FRCs function is more than controlling T cell expansion. FRCs also outline the fate and function of CD8 T lymphocytes through their IL-6 production, and CD8 T cells exposed to both FCRs and IL-6 are driven to a memory phenotype. In addition, CD8 T cells cultivated in the presence of FRCs are more persistent during a viral infection than CD8 T cells stimulated without FRC presence [24]. Moreover, the pleiotropic function and the importance of IL-6 were reported before, supporting that this cytokine, in certain environments, could be an important player for guiding the immune response [44]. Next, Masters et al., reported that after aging-related changes, FRCs have an altered impact on the beginning of the immune response to influenza infection, consequently contributing to delayed T lymphocytes responses to this virus [28]. Moreover, their findings on the importance of homeostatic chemokines for the success of the anti-viral response are also supported by Chai et al., who previously reported on the importance of these chemokines secreted by FRCs to the immune response against virus infection, and by Thompson et al., who also reported on the role of the lymph node in aging mice and its negative impact on T cells [45][46]. Lastly, Royer et al. proposed that HSV-1 in lymph nodes can cause pathological alterations in the FRC conduit system, resulting in fewer HSV-specific CD8 T lymphocytes in circulation, and a diminished anti-viral response to this virus. In addition, they claim that immunodeficiency can occur as a secondary outcome of FRC alterations to SLOs [40]. Their results are supported by other models that impair T cell responses due to virus-associated damage to FRCs [47][48]. Concerning inflammation (Figure 1B) and the COX/PGE2 pathway, which converts arachidonic acid in several prostanoids via the enzymes COX1 and COX2, FRCs have been proposed to play dual roles by either promoting or inhibiting adaptive immunity [49][50], similar to myeloid and T cells. Schaeuble et al.’s experiments revealed that FRCs can control T cell responses, independently of other cells, by two pathways that lead to NO release, clarifying that one pathway is activated via the sensing of IFN-y by FRCs, which is activated only by strong T cell responses, and another pathway is mediated by COX2-dependent synthesis of PGE2, which signals via EP1 and EP2 during both weak and strong T cells responses [29]. Knoblich et al. also demonstrated that FRCs control T cell proliferation and modulate their differentiation [31]. Knoblich et al. included even more mechanisms that control T cell proliferation besides IFN-y and PGE2, which, in human cells, do not release NO, but instead activate IDO; they point to TGF-β and the adenosine 2A receptor (A2AR) as other signaling pathways affecting T cell proliferation. They also demonstrated that human FRCs affect the fate of naïve T cells, diminishing their differentiation into central memory while enhancing effector and effector memory phenotypes [31]. Yu, M. et al. support these findings with their previous study on the animal model and in vitro assays, confirming that hyperactive COX-2/PGE2 pathways in FRCs are a mechanism that maintains peripheral T cell tolerance [37]. In addition, Valencia et al. demonstrated the differences between mice and humans regarding COX inflammatory pathways, and concluded that human and murine FRCs would regulate T lymphocytes responses using different mechanisms, and arguing that, in humans, IDO would play a more important role than iNOS/NO [36]. Further, in autoimmunity, the FRC network seems to play an important role (Figure 1C). Gonzalez et al., using a type 1 diabetes (T1D) model and a 3D system of culture, found that in T1D FRCs, the reticular network organization was altered, displayed larger pores, and had a lower expression of podoplanin compared to a control animal or control culture system. They also demonstrated a reduced expression of PTAs and T1D antigens in T1D FRCs. Consequently, FRCs modulated their interactions with autoreactive T lymphocytes by remodeling their reticular network in LNs; PTAs and podoplanin played a central role and their alterations may favor T1D [21]. These findings are supported by a previous study from the same group that investigated alterations in pancreatic lymph nodes from humans and mice [51]. Kasinath et al. studied crescentic glomerulonephritis (GN), an autoimmune inflammatory condition characterized by the rapid deterioration of kidney function. They investigated the role of fibroblastic reticular cells residing in the stromal compartment of the kidney lymph node in this model. They observed that FRCs are fundamental to the propagation of the immune response in nephrotoxic serum nephritis. Following GN development, they observed an increase in effector memory and Th17 cells in the kidney LN. In addition, they observed that the removal of the kidney lymph node, a depletion of fibroblastic reticular cells, and treatment with anti-podoplanin antibodies each resulted in a reduction of kidney injury [25]. Majumder et al. studied the EAE model, and they also showed Th17 differentiation in LNs and that the signaling in the receptor for IL-17 in FRCs is related to collagen deposition in LNs. This work suggests that Th17 cells promote ECM deposition in inflamed LNs through FRCs-IL-17 signaling, independently of LN size or hypercellularity. As a consequence of Th17 in LN, the released IL-17 signals in FRCs impact LN stromal organization, leading to FRC activation by changing their phenotype from quiescence to highly metabolic. Moreover, the absence of IL-17 signaling in FRCs does not lead to immune failings but does cause impaired B cell responses, due to the reduced availability of BAFF, which is critical for the germinal center formation and maintenance [27]. In metastatic cancer (Figure 1D), FRCs appear to be regulated by the tumor cells. Eom et al. showed in human melanoma that FRCs in tumor-infiltrated LNs may favor cancer invasion and progression through secretion of soluble factors, alterations in the lymph node structure, and by promoting pathological conditions such as fibrosis [20]. Gao et al. also showed in tumor-infiltrated LNs a decrease in FRCs and IL-7 secretion, leading to a declined number and diminished function of T cells in LNs [34]. In homeostasis, as displayed in Figure 1E, FRCs played an important role in secreting homeostatic chemokines, promoting the meeting between T cells and dendritic cells on the T cell zone, and also by secreting IL-7, an essential cytokine involved in T cell effector memory differentiation [2][14][28]. Furthermore, in graft versus host disease (GvHD) FRCs’ ability for peripheral tissue antigen (PTA) presentation and NOTCH signaling have been shown to be important features in the aggravation and maintenance of the GvHD state (Figure 1F). Dertschnig et al. showed that the loss of PTA presentation by FRCs during GVHD leads to permanent damage in their networks in lymphoid tissues, compromising peripheral tolerance. In addition, they demonstrated that not only the disruption of FCRs occurs during GvHD but also the capacity for the regeneration of this network is impaired, different to what was found for viral infection, where the damage occurs, but after viral clearance, the network is restored [19]. Chung et al. showed that FRC-delivered NOTCH signals through the ligands DLL1 and DLL4 to donor alloreactive T cells help to program the pathogenicity of these T cells. Moreover, they demonstrated that the early use of neutralizing antibodies against DDL1 and DDL4 abrogated GvHD [33]. As seen by Kasinath et al. in GN [25], Maaraouf et al., using ischemic reperfusion injury (IRI) with multiple IRI [32], reported that kidney LNs (KLNs) cause excessive deposition of ECM fibers containing fibronectin and collagen, which leads to local fibrosis, similar to kidney fibrosis. They confirmed that depletion of FRCs reduced T cell activation in the KLNs and ameliorated renal injury in acute IRI [25][32]. Regarding pathway investigation, Knop et al. demonstrated that FRC-derived IL-7 plays an essential role in maintaining central memory T cells, but is dispensable for naïve T cell survival [22]. Dubrot et al. showed a mechanism of T lymphocyte proliferation inhibition by the IFN-y-induced expression of MHC II [30]. In addition, they demonstrated that the deletion of MHC II in LN stromal cells in vivo leads to diminished Treg frequency and functions, and, at the same time, enhances effector cell differentiation, further leading to T cell tissue infiltration and the subsequent development of T cell-mediated autoimmunity [30]. Pazstoi et al. used the stromal compartment of gut-draining LNs to demonstrate that FRCs own the tolerogenic capacity that controls T cells. They also demonstrated that mesenteric LNs (mLNs) are more capable of inducing [35] Treg profiles than the peripheral ones. Likewise, they demonstrated that FRCs contribute to peripheral tolerance by developing de novo Treg by releasing microvesicles (MVEs), which carry high levels of TGF-β [35]. Gil-Cruz et al. also used mLNs and Peyer’s patches (PP) as the targets of their study and identified that an antiviral response driven by ILC1 and NK was regulated by the FRCs’ limiting provision of IL-15 [38]. This mechanism control seems to be activated by TLR7 and/or IL-1β, and its control is designated by the MyD88-dependent pathway [38]. Novkovic confirmed that the physical scaffold of LNs formed by the FRC network is critical for the maintenance and functionality of LNs [39], and Takeda et al. demonstrated the role of LPA derived from FRCs in T cell motility [41]. Kelch et al. demonstrated, by imaging, the conduit density in the deep and superficial T cell zone, concluding that although T cells within the superficial zone stay in constant contact with FRCs, and in the deep T cell zone, there is a gap that does not guarantee simultaneous contact for all T cells in this region [26]. In summary, FRCs in homeostasis plays an important role in secreting homeostatic chemokines and IL-7, which are essential for the immune response initiation and for T cell effector memory phenotype differentiation. In a viral setting, FRCs are the main source of IL-33, playing a regulatory role by diminishing the T cell exhaustion, and acting on T cell fate through IL-6 secretion. In this same setting, aging FRCs have a negative impact on T cells. In inflammation, FRCs have been proposed to play a dual role by either promoting or inhibiting adaptive immunity. The main mechanisms behind inflammation are related to IFN-y and PGE2-signaling that, in murine cells, release NO and, in humans, activate IDO. In autoimmunity, the reticular network organization was altered, displayed lower expression of PTAs and podoplanin, and, in this context, IL-17 signaling impacted LN stromal organization, leading to highly metabolically activated FRCs. In metastatic cancer, FRCs appear to be regulated by the tumor cells decreasing IL-7 secretion and enhancing other soluble factors, causing alterations in the lymph node structure, such as fibrosis. In GvHD, the loss of PTA presentation by FRCs leads to permanent damage in their networks, compromising peripheral tolerance.

References

  1. Denton, A.E.; Carr, E.J.; Magiera, L.P.; Watts, A.J.B.; Fearon, D.T. Embryonic FAP+ lymphoid tissue organizer cells generate the reticular network of adult lymph nodes. J. Exp. Med. 2019, 216, 2242–2252.
  2. Link, A.; Vogt, T.K.; Favre, S.; Britschgi, M.R.; Acha-Orbea, H.; Hinz, B.; Cyster, J.G.; Luther, S.A. Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells. Nat. Immunol. 2007, 8, 1255–1265.
  3. Sixt, M.; Kanazawa, N.; Selg, M.; Samson, T.; Roos, G.; Reinhardt, D.P.; Pabst, R.; Lutz, M.B.; Sorokin, L. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity 2005, 22, 19–29.
  4. Martinez, V.G.; Pankova, V.; Krasny, L.; Singh, T.; Makris, S.; White, I.J.; Benjamin, A.C.; Dertschnig, S.; Horsnell, H.L.; Kriston-Vizi, J.; et al. Fibroblastic Reticular Cells Control Conduit Matrix Deposition during Lymph Node Expansion. Cell Rep. 2019, 29, 2810–2822.
  5. Luther, S.A.; Vogt, T.K.; Siegert, S. Guiding blind T cells and dendritic cells: A closer look at fibroblastic reticular cells found within lymph node T zones. Immunol. Lett. 2011, 138, 9–11.
  6. Mueller, S.N.; Ahmed, R. Lymphoid stroma in the initiation and control of immune responses. Immunol. Rev. 2008, 224, 284–294.
  7. Alvarenga, H.G.; Marti, L. Multifunctional Roles of Reticular Fibroblastic Cells: More Than Meets the Eye? J. Immunol. Res. 2014, 2014, 402038.
  8. Fletcher, A.L.; Acton, S.E.; Knoblich, K. Lymph node fibroblastic reticular cells in health and disease. Nat. Rev. Immunol. 2015, 15, 350–361.
  9. Severino, P.; Palomino, D.T.; Alvarenga, H.; Almeida, C.B.; Pasqualim, D.C.; Cury, A.; Salvalaggio, P.R.; De Vasconcelos Macedo, A.L.; Andrade, M.C.; Aloia, T.; et al. Human Lymph Node-Derived Fibroblastic and Double-Negative Reticular Cells Alter Their Chemokines and Cytokines Expression Profile Following Inflammatory Stimuli. Front. Immunol. 2017, 8, 141.
  10. Vega, F.; Coombes, K.R.; Thomazy, V.A.; Patel, K.; Lang, W.; Jones, D. Tissue-specific function of lymph node fibroblastic reticulum cells. Pathobiol. J. Immunopathol. Mol. Cell. Biol. 2006, 73, 71–81.
  11. Frontera, V.; Arcangeli, M.L.; Zimmerli, C.; Bardin, F.; Obrados, E.; Audebert, S.; Bajenoff, M.; Borg, J.P.; Aurrand-Lions, M. Cutting edge: JAM-C controls homeostatic chemokine secretion in lymph node fibroblastic reticular cells expressing thrombomodulin. J. Immunol. 2011, 187, 603–607.
  12. Kaldjian, E.P.; Gretz, J.E.; Anderson, A.O.; Shi, Y.; Shaw, S. Spatial and molecular organization of lymph node T cell cortex: A labyrinthine cavity bounded by an epithelium-like monolayer of fibroblastic reticular cells anchored to basement membrane-like extracellular matrix. Int. Immunol. 2001, 13, 1243–1253.
  13. Lukacs-Kornek, V.; Malhotra, D.; Fletcher, A.L.; Acton, S.E.; Elpek, K.G.; Tayalia, P.; Collier, A.-r.; Turley, S.J. Regulated release of nitric oxide by nonhematopoietic stroma controls expansion of the activated T cell pool in lymph nodes. Nat. Immunol. 2011, 12, 1096–1104.
  14. Fletcher, A.L.; Malhotra, D.; Turley, S.J. Lymph node stroma broaden the peripheral tolerance paradigm. Trends Immunol. 2011, 32, 12–18.
  15. Gardner, J.M.; Devoss, J.J.; Friedman, R.S.; Wong, D.J.; Tan, Y.X.; Zhou, X.; Johannes, K.P.; Su, M.A.; Chang, H.Y.; Krummel, M.F.; et al. Deletional tolerance mediated by extrathymic Aire-expressing cells. Science 2008, 321, 843–847.
  16. Nadafi, R.; Gago de Graça, C.; Keuning, E.D.; Koning, J.J.; de Kivit, S.; Konijn, T.; Henri, S.; Borst, J.; Reijmers, R.M.; van Baarsen, L.G.M.; et al. Lymph Node Stromal Cells Generate Antigen-Specific Regulatory T Cells and Control Autoreactive T and B Cell Responses. Cell Rep. 2020, 30, 4110–4123.
  17. Krausgruber, T.; Fortelny, N.; Fife-Gernedl, V.; Senekowitsch, M.; Schuster, L.C.; Lercher, A.; Nemc, A.; Schmidl, C.; Rendeiro, A.F.; Bergthaler, A.; et al. Structural cells are key regulators of organ-specific immune responses. Nature 2020, 583, 296–302.
  18. Aparicio-Domingo, P.; Cannelle, H.; Buechler, M.B.; Nguyen, S.; Kallert, S.M.; Favre, S.; Alouche, N.; Papazian, N.; Ludewig, B.; Cupedo, T.; et al. Fibroblast-derived IL-33 is dispensable for lymph node homeostasis but critical for CD8 T-cell responses to acute and chronic viral infection. Eur. J. Immunol. 2020, 51, 76–90.
  19. Dertschnig, S.; Evans, P.; Santos, E.S.P.; Manzo, T.; Ferrer, I.R.; Stauss, H.J.; Bennett, C.L.; Chakraverty, R. Graft-versus-host disease reduces lymph node display of tissue-restricted self-antigens and promotes autoimmunity. J. Clin. Investig. 2020, 130, 1896–1911.
  20. Eom, J.; Park, S.M.; Feisst, V.; Chen, C.J.J.; Mathy, J.E.; McIntosh, J.D.; Angel, C.E.; Bartlett, A.; Martin, R.; Mathy, J.A.; et al. Distinctive Subpopulations of Stromal Cells Are Present in Human Lymph Nodes Infiltrated with Melanoma. Cancer Immunol. Res. 2020, 8, 990–1003.
  21. Gonzalez Badillo, F.; Zisi Tegou, F.; Masina, R.; Wright, S.; Scully, M.; Harwell, L.; Lupp, M.; Postigo-Fernandez, J.; Creusot, R.J.; Tomei, A.A. Tissue-Engineered Stromal Reticula to Study Lymph Node Fibroblastic Reticular Cells in Type I Diabetes. Cell. Mol. Bioeng. 2020, 13, 419–434.
  22. Knop, L.; Deiser, K.; Bank, U.; Witte, A.; Mohr, J.; Philipsen, L.; Fehling, H.J.; Müller, A.J.; Kalinke, U.; Schüler, T. IL-7 derived from lymph node fibroblastic reticular cells is dispensable for naive T cell homeostasis but crucial for central memory T cell survival. Eur. J. Immunol. 2020, 50, 846–857.
  23. Perez-Shibayama, C.; Islander, U.; Lütge, M.; Cheng, H.W.; Onder, L.; Ring, S.S.; de Martin, A.; Novkovic, M.; Colston, J.; Gil-Cruz, C.; et al. Type I interferon signaling in fibroblastic reticular cells prevents exhaustive activation of antiviral CD8+ T cells. Sci. Immunol. 2020, 5.
  24. Brown, F.D.; Sen, D.R.; LaFleur, M.W.; Godec, J.; Lukacs-Kornek, V.; Schildberg, F.A.; Kim, H.J.; Yates, K.B.; Ricoult, S.J.H.; Bi, K.; et al. Fibroblastic reticular cells enhance T cell metabolism and survival via epigenetic remodeling. Nat. Immunol. 2019, 20, 1668–1680.
  25. Kasinath, V.; Yilmam, O.A.; Uehara, M.; Jiang, L.; Ordikhani, F.; Li, X.; Salant, D.J.; Abdi, R. Activation of fibroblastic reticular cells in kidney lymph node during crescentic glomerulonephritis. Kidney Int. 2019, 95, 310–320.
  26. Kelch, I.D.; Bogle, G.; Sands, G.B.; Phillips, A.R.J.; LeGrice, I.J.; Dunbar, P.R. High-resolution 3D imaging and topological mapping of the lymph node conduit system. PLoS Biol. 2019, 17, e3000486.
  27. Majumder, S.; Amatya, N.; Revu, S.; Jawale, C.V.; Wu, D.; Rittenhouse, N.; Menk, A.; Kupul, S.; Du, F.; Raphael, I.; et al. IL-17 metabolically reprograms activated fibroblastic reticular cells for proliferation and survival. Nat. Immunol. 2019, 20, 534–545.
  28. Masters, A.R.; Hall, A.; Bartley, J.M.; Keilich, S.R.; Lorenzo, E.C.; Jellison, E.R.; Puddington, L.; Haynes, L. Assessment of Lymph Node Stromal Cells as an Underlying Factor in Age-Related Immune Impairment. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2019, 74, 1734–1743.
  29. Schaeuble, K.; Cannelle, H.; Favre, S.; Huang, H.Y.; Oberle, S.G.; Speiser, D.E.; Zehn, D.; Luther, S.A. Attenuation of chronic antiviral T-cell responses through constitutive COX2-dependent prostanoid synthesis by lymph node fibroblasts. PLoS Biol. 2019, 17, e3000072.
  30. Dubrot, J.; Duraes, F.V.; Harlé, G.; Schlaeppi, A.; Brighouse, D.; Madelon, N.; Göpfert, C.; Stokar-Regenscheit, N.; Acha-Orbea, H.; Reith, W.; et al. Absence of MHC-II expression by lymph node stromal cells results in autoimmunity. Life Sci. Alliance 2018, 1, e201800164.
  31. Knoblich, K.; Cruz Migoni, S.; Siew, S.M.; Jinks, E.; Kaul, B.; Jeffery, H.C.; Baker, A.T.; Suliman, M.; Vrzalikova, K.; Mehenna, H.; et al. The human lymph node microenvironment unilaterally regulates T-cell activation and differentiation. PLoS Biol. 2018, 16, e2005046.
  32. Maarouf, O.H.; Uehara, M.; Kasinath, V.; Solhjou, Z.; Banouni, N.; Bahmani, B.; Jiang, L.; Yilmam, O.A.; Guleria, I.; Lovitch, S.B.; et al. Repetitive ischemic injuries to the kidneys result in lymph node fibrosis and impaired healing. JCI Insight 2018, 3.
  33. Chung, J.; Ebens, C.L.; Perkey, E.; Radojcic, V.; Koch, U.; Scarpellino, L.; Tong, A.; Allen, F.; Wood, S.; Feng, J.; et al. Fibroblastic niches prime T cell alloimmunity through Delta-like Notch ligands. J. Clin. Investig. 2017, 127, 1574–1588.
  34. Gao, J.; Zhao, L.; Liu, L.; Yang, Y.; Guo, B.; Zhu, B. Disrupted fibroblastic reticular cells and interleukin-7 expression in tumor draining lymph nodes. Oncol. Lett. 2017, 14, 2954–2960.
  35. Pasztoi, M.; Pezoldt, J.; Beckstette, M.; Lipps, C.; Wirth, D.; Rohde, M.; Paloczi, K.; Buzas, E.I.; Huehn, J. Mesenteric lymph node stromal cell-derived extracellular vesicles contribute to peripheral de novo induction of Foxp3(+) regulatory T cells. Eur. J. Immunol. 2017, 47, 2142–2152.
  36. Valencia, J.; Jiménez, E.; Martínez, V.G.; Del Amo, B.G.; Hidalgo, L.; Entrena, A.; Fernández-Sevilla, L.M.; Del Río, F.; Varas, A.; Vicente, Á.; et al. Characterization of human fibroblastic reticular cells as potential immunotherapeutic tools. Cytotherapy 2017, 19, 640–653.
  37. Yu, M.; Guo, G.; Zhang, X.; Li, L.; Yang, W.; Bollag, R.; Cui, Y. Fibroblastic reticular cells of the lymphoid tissues modulate T cell activation threshold during homeostasis via hyperactive cyclooxygenase-2/prostaglandin E(2) axis. Sci. Rep. 2017, 7, 3350.
  38. Gil-Cruz, C.; Perez-Shibayama, C.; Onder, L.; Chai, Q.; Cupovic, J.; Cheng, H.W.; Novkovic, M.; Lang, P.A.; Geuking, M.B.; McCoy, K.D.; et al. Fibroblastic reticular cells regulate intestinal inflammation via IL-15-mediated control of group 1 ILCs. Nat. Immunol. 2016, 17, 1388–1396.
  39. Novkovic, M.; Onder, L.; Cupovic, J.; Abe, J.; Bomze, D.; Cremasco, V.; Scandella, E.; Stein, J.V.; Bocharov, G.; Turley, S.J.; et al. Topological Small-World Organization of the Fibroblastic Reticular Cell Network Determines Lymph Node Functionality. PLoS Biol. 2016, 14, e1002515.
  40. Royer, D.J.; Conrady, C.D.; Carr, D.J. Herpesvirus-Associated Lymphadenitis Distorts Fibroblastic Reticular Cell Microarchitecture and Attenuates CD8 T Cell Responses to Neurotropic Infection in Mice Lacking the STING-IFNα/β Defense Pathways. J. Immunol. 2016, 197, 2338–2352.
  41. Takeda, A.; Kobayashi, D.; Aoi, K.; Sasaki, N.; Sugiura, Y.; Igarashi, H.; Tohya, K.; Inoue, A.; Hata, E.; Akahoshi, N.; et al. Fibroblastic reticular cell-derived lysophosphatidic acid regulates confined intranodal T-cell motility. eLife 2016, 5, e10561.
  42. Krishnamurty, A.T.; Turley, S.J. Lymph node stromal cells: Cartographers of the immune system. Nat. Immunol. 2020, 21, 369–380.
  43. Talemi, S.R.; Höfer, T. Antiviral interferon response at single-cell resolution. Immunol. Rev. 2018, 285, 72–80.
  44. Hunter, C.A.; Jones, S.A. IL-6 as a keystone cytokine in health and disease. Nat. Immunol. 2015, 16, 448–457.
  45. Chai, Q.; Onder, L.; Scandella, E.; Gil-Cruz, C.; Perez-Shibayama, C.; Cupovic, J.; Danuser, R.; Sparwasser, T.; Luther, S.A.; Thiel, V.; et al. Maturation of lymph node fibroblastic reticular cells from myofibroblastic precursors is critical for antiviral immunity. Immunity 2013, 38, 1013–1024.
  46. Thompson, H.L.; Smithey, M.J.; Uhrlaub, J.L.; Jeftić, I.; Jergović, M.; White, S.E.; Currier, N.; Lang, A.M.; Okoye, A.; Park, B.; et al. Lymph nodes as barriers to T-cell rejuvenation in aging mice and nonhuman primates. Aging Cell 2019, 18, e12865.
  47. Mueller, S.N.; Matloubian, M.; Clemens, D.M.; Sharpe, A.H.; Freeman, G.J.; Gangappa, S.; Larsen, C.P.; Ahmed, R. Viral targeting of fibroblastic reticular cells contributes to immunosuppression and persistence during chronic infection. Proc. Natl. Acad. Sci. USA 2007, 104, 15430–15435.
  48. Zeng, M.; Smith, A.J.; Wietgrefe, S.W.; Southern, P.J.; Schacker, T.W.; Reilly, C.S.; Estes, J.D.; Burton, G.F.; Silvestri, G.; Lifson, J.D.; et al. Cumulative mechanisms of lymphoid tissue fibrosis and T cell depletion in HIV-1 and SIV infections. J. Clin. Investig. 2011, 121, 998–1008.
  49. Malhotra, D.; Fletcher, A.L.; Turley, S.J. Stromal and hematopoietic cells in secondary lymphoid organs: Partners in immunity. Immunol. Rev. 2013, 251, 160–176.
  50. Siegert, S.; Luther, S.A. Positive and negative regulation of T cell responses by fibroblastic reticular cells within paracortical regions of lymph nodes. Front. Immunol. 2012, 3, 285.
  51. Postigo-Fernandez, J.; Farber, D.L.; Creusot, R.J. Phenotypic alterations in pancreatic lymph node stromal cells from human donors with type 1 diabetes and NOD mice. Diabetologia 2019, 62, 2040–2051.
More