LN-Derived Fibroblastic Reticular Cells: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Microbiology
Contributor:

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. 

  • 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. Year Host Interventions
Source Genotype Age (Weeks) Gender Type Time (Days)
Aparicio-Domingo et al. [18] 2020 Mice C57BL/6J IL-33gfp/gfp; IL-33gfp/+ 7–19 M LCMV clone 13 and WE virus; tamoxifen Single dose;
6 (3/week)
Dertschnig et al. [19] 2020 Mice C57BL/6 Female to male bone marrow transplant model (BMT), T cell-depleted, plus transgenic TCR-CD8 MataHari (Mh) NR M Dexamethasone; DT; Gy irradiation 3; 4
Eom et al. [20] 2020 Human Metastatic melanoma and surgery NA NA NA NA
Gonzalez et al. [21] 2020 Mice (NOD/ShiLtJ, NOR/LtJ, and NOD.CgTg); Human Type 1
diabetes
12 F NA NA
Knop et al. [22] 2020 Mice C57BL/6N and ROSA26RFP IL-7−/−, PGK-Cre, FLPO, RAG1−/−, Thy1.1+ OT-I NR NR NA NA
Perez-Shibayama et al. [23] 2020 Mice C57BL/6 CCL19-Cre IFNARfl/fl 8–10 NR LCMV Armstrong NR
Brown et al. [24] 2019 Mice C57BL/6 IL-6−/−; NOS2−/− 5–12 M PR8-GP33-41, LCMV, influenza, OT-1 T cells with OVA NR
Kasinath et al. [25] 2019 Mice CD-1 IGS or C57BL/6 or C57BL/6J CCL19-Cre iDTR 8–10 M Nephrotoxic serum (NTS); DT 3
Kelch et al. [26] 2019 Mice C57BL/6J NA 9–22 M NA NA
Majumder et al. [27] 2019 Mice C57BL/6 IL-17A−/−; IL-17RAfl/fl; OT-II, ACT1−/−; CCL19-Cre; IL23R−/−; Regnase1+/- 6–12 M-F MOG with Mycobacterium tuberculosis, pertussis toxin on/OT-II CD4+ T cells with OVA/DSS 2
Masters et al. [28] 2019 Mice C57BL/6 RAG−/−; CD45.1 2–4 m and 19–21 m M Influenza NR
Schaeuble et al. [29] 2019 Mice C57BL/6 NOS2−/−; OT-1; COX2−/−, COX2ΔCCL19Cre, and ROSA26-EYFPCCL19Cre ≥6 NR OVA and poly (I:C) 4
Dubrot et al. [30] 2018 Mice C57BL/6 CIITA−/−; pIV−/−; K14 TGP IVKO; RAG2−/− PROX-1-Cre MHC-IIfl >12m NR Tamoxifen; IFN-γ and FTY720 4 (Twice/day); 6
Knoblich et al. [31] 2018 Human Cadaveric donors NA NA NA NA
Maaraouf et al. [32] 2018 Mice C57BL/6 CCL19-Cre; iDTR; RAG1−/− NR NR DT; LTβr-Ig 1; 2
Chung et al. [33] 2017 Mice BALB/c or C57BL/6 TgMx1-Cre; DLL1fl/fl; DLL4fl/fl; NOTCH2fl/fl; RAG1−/− 6–10 or 8–12 M-F poly (I:C)/8.5-9 Gy;
poly (I:C)/6 Gy irradiation
0.16; 0.12
Gao et al. [34] 2017 Mice C57BL/6 and Human Colon cancer 6 F Lewis Long carcinoma cells NA
Pazstoi et al. [35] 2017 Mice BALB/c FOXP3hCD2xRAG2−/− xD011.10 NR M-F NA NA
Valencia et al. [36] 2017 Human Brain-dead organ donors NA M-F NA NA
Yu, M. et al. [37] 2017 Mice C57BL/6 and Human PTGS2Y385F/Y385F; OVA-specific CD8 (OT-I); CD4 (OT-II) 4–6 NR DC-vaccine 1.5
Gil-Cruz et al. [38] 2016 Mice C57BL/6N or C57BL/6N-Tg or R26R-EYFP Myd88−/−; TLR7−/−; CCL19-Cre 8–10 NR MHV A59; Citrobacter rodentium 12; 6
Novkovic et al. [39] 2016 Mice C57BL/6N or C57BL/6N-Tg CCL19-Cre; iDTR 6–9 NR DT 3 and 5
Royer et al. [40] 2016 Mice C57BL/6 or Gbt-1.1 CXCL10−/−; CXCR3−/−; STING−/−; CD18−/− 6–12 M-F HSV-1 NR
Takeda et al. [41] 2016 Mice C57BL/6J LPAR2−/−; ENPP2-flox, CCCL19-Cre, LPAR5−/−; LPAR6−/− 8–12 NR CD4+ T cells labeled with CMTMR; LTβR-Fc 0.6; 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.
 
 
 
 
 
Table 2. Characteristics of fibroblastic reticular cells isolation and their immunophenotype.
Ref. Lymph Node Region Digestion Type Digestion Solution FRC Culture Medium + Supplement FRC Immunophenotypic Characterization Technique for Cell Separation Purity
(%)
Aparicio-Domingo et al. [18] Axillary; brachial; inguinal Enzymatic Collagenase IV; DNase I; CaCl2 DMEM (2% FCS) CD45; CD31; PDPN Cell sorting >94
Dertschnig et al. [19] Peripheral; mesenteric Enzymatic DNase; Liberase NC CD45; CD31; PDPN Cell sorting NR
Eom et al. [20] Axillary; inguinal; cervical; mesenteric; mediastinum Enzymatic DNase I; Liberase DH RPMI-1640 CD45, CD31, PDPN NR NR
Gonzalez et al. [21] Skin-draining (brachial; axillary; inguinal); Pancreatic Enzymatic Collagenase P; DNase I; Dispase II NR CD45; CD31; PDPN Cell sorting NR
Knop et al. [22] Peripheral; mesenteric Enzymatic Collagenase P; Dispase II; DNase I; Latrunculin B RPMI-1640 CD45; CD31; PDPN Cell sorting >73.3
Perez-Shibayama et al. [23] Inguinal Enzymatic Collagenase F; DNase I RPMI NR NR NR
Brown et al. [24] NR Enzymatic Collagenase P; DNase I; Dispase α-MEM CD45; CD31; PDPN Cell sorting >95
Kasinath et al. [25] Kidney NA NA NA NA NA NA
Kelch et al. [26] Popliteal; mesenteric; Inguinal NA NA NA NA NA NA
Majumder et al. [27] Mesenteric; inguinal Enzymatic DNase I; Liberase; Dispase RPMI CD45; CD31; PDPN; Microbeads isolation >98
Masters et al. [28] Mesenteric;
popliteal
Enzymatic Liberase TL; Benzonuclease RPMI-1640 CD45; CD31; PDPN Microbeads isolation >90
Schaeuble et al. [29] Peripheral (axillary, brachial, inguinal) Enzymatic Collagenase IV; DNase I DMEM (2% FCS) CD45; CD31; PDPN Microbeads isolation ≥90
Dubrot et al. [30] Skin-draining Enzymatic Collagenase D; DNase I HBSS CD45; CD31; PDPN Cell sorting NR
Knoblich et al. [31] NR Enzymatic Collagenase P; DNase I; Dispase α-MEM (10% FBS) CD45; PDPN NR 99
Maaraouf et al. [32] Kidney Enzymatic Collagenase P; DNase I; Dispase II DMEM (10% FBS) CD45; CD31; PDPN NR NR
Chung et al. [33] Peripheral (cervical, axial, brachial, inguinal) Enzymatic Collagenase IV; DNase I DMEM (2% FBS) CD45; CD31; PDPN Cell sorting NR
Gao et al. [34] Inguinal Enzymatic Collagenase IV; DNase I RPMI-1640 (2% FBS) CD45; CD31; PDPN NA NA
Pazstoi et al. [35] Mesenteric Enzymatic Collagenase P; Dispase; DNase I RPMI-1640 CD45; CD31; PDPN Cell sorting 91–97
Valencia et al. [36] Mesenteric Mechanical disruption NR RPMI-1640 CD45, CD31, PDPN NR NR
Yu, M. et al. [37] Axillary; brachial; inguinal Enzymatic Collagenase P; Dispase; DNase I DMEM (10% FBS) CD45; CD31; PDPN Cell sorting >95
Gil-Cruz et al. [38] Mesenteric Enzymatic Collagenase D; DNase I RPMI-1640 (2% FCS) CD45; CD31; PDPN Cell sorting NR
Novkovic et al. [39] Inguinal Enzymatic Collagenase P; DNase I RPMI (2% FCS) PDPN NA NA
Royer et al. [40] Mandibular Mechanical disruption NR RPMI-1640 (10% FBS) NR NR NR
Takeda et al. [41] Mesenteric; peripheral; brachial Enzymatic Collagenase P; Dispase; DNase I RPMI-1640 CD45; CD31; PDPN Cell sorting NR
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 Cells Cell Type Separation Technique Immune Cell Preservation Solution and Supplementation Immune Cell Immunophenotypic Characterization
Aparicio-Domingo et al. [18] LN CD8+ T cells Non selection performed DMEM (2% FCS) CD45, CD8α, CD4, TRCαβ
Dertschnig et al. [19] LN T cells CD3 negative selection followed by
CD4 and CD8α
positive selection
(MicroBeads—Myltenyi)
NR CD45, CD45.1, CD3, CD4, CD8α, CD62L, CD44, CD69, CD127, Vα2, Vβ5
Eom et al. [20] LN NA NA NA CD45, CD3, CD8
Gonzalez et al. [21] Spleen CD8+ T cells CD8 isolation by negative selection (Microbeads—MojoSort) NR CD45, CD8, CD44, CD25
Knop et al. [22] LN; spleen T cells and NK CD8α positive selection (MicroBeads—Myltenyi) RPMI CD45, CD3, CD4, CD5, CD8α, CD62L, Bcl-2, CD127, Nk1.1, RORγt
Perez-Shibayama et al. [23] LN; spleen T cell subsets and exhaustion No selection performed RPMI CD45.1, CD45.2, CD45R, CD8α, CD8β, CD3e, CD44 CD62L, PD-1, PDL1
Brown et al. [24] LN CD8+ T cells CD8α positive selection (MicroBeads—Myltenyi) RPMI; α-MEM CD45.1, CD45.2, CD3, CD4, CD8, CD275, CD28, CD44
Kasinath et al. [25] LN; spleen CD4+ T cells No selection performed NR CD45, CD3, CD4, CD44, CD62L, IL-17A
Kelch et al. [26] LN NA NA NA NA
Majumder et al. [27] LN T and B cells NR NR CD45, CD45.2, CD4, B220, IL-17A, IL-17R
Masters et al. [28] LN; peripheral blood CD8+ T CD8 isolation by negative selection (Microbeads—MojoSort) NR CD45, CD45.1, CD45.2, CD69, CD8α
Schaeuble et al. [29] LN; spleen T cells No selection performed RPMI CD45, CD3, CD4, CD8α, CD44, CD62L, CD279, FoxP3, CD25
Dubrot et al. [30] LN; spleen T cells, B cells, Treg, and DC Pan T isolation by negative selection
(MicroBeads—Myltenyi)
NR CD45, CD44, CD3, CD4, CD8α, FOXp3, Ly5.1, CD11b, CD19, CD25, CD62L, PDCA-1, PD-1, IL-17, IFNγR
Knoblich et al. [31] LN; tonsils T cells Pan T isolation by negative selection
(MicroBeads—Myltenyi)
NR CD45, CD3, CD4, CD8, CD62L, CD27, CD45RO, CD25
Maaraouf et al. [32] Spleen T cells Pan T isolation by negative selection
(MicroBeads—Myltenyi)
NR CD45, CD4
Chung et al. [33] Spleen; peripheral blood T cells, B cells, FDCs, Treg, and DCs T cell Thy.1 selection
(Microbeads—StemCells Technologies)
NA CD45, CD3, CD4, CD8, FOXp3, CD157, CD19, B220, CD44, CD62L, CD11c, CD11b, CD169, CD21/35, F4/80, TCRβ
Gao et al. [34] LN T cells NR NR CD45, CD4, CD8
Pazstoi et al. [35] LN T cells CD4 positive selection
(Microbeads—Myltenyi)
EX VIVO CD45, CD45.2, CD4, CD2, CD9, CD24, CD25, CD63
Valencia et al. [36] LN CD4+ T cells CD4 naïve T cell negative selection
(Microbeads—Myltenyi)
RPMI (10% FCS) CD45, CD44, CD4
Yu, M. et al. [37] LN T cells Pan T cell negative selection
(Microbeads—StemCells Technologies)
RPMI (10% FBS) CD45, CD45.1, CD45.2, CD3, CD4, CD8α, CD25, CD69, CD44
Gil-Cruz et al. [38] PP; LN T cells, B cells, NK cells, Treg, and ILCs NR RPMI (10% FCS) CD45, CD3e, CD4, CD8α, EOMES, FoxP3, B220, CD19, CD127, CD62L, CD44, CD69, F4/80, IL-17A, IL-7Rα, GATA3, RORγt, IL-15RαIL-15Rβ, NKp46, NK1.1
Novkovic et al. [39] LN; Spleen DCs and T cells NR RPMI (2% FCS) CD45, CD3, CD8, CD4, CD11c, MHCII
Royer et al. [40] SLOs CD8+ T cells CD8 positive selection (Microbeads—Myltenyi) RPMI (10% FBS) CD45, CD3, CD4, CD8
Takeda et al. [41] LN; Spleen T cells, B cells CD4 naïve T cell negative selection
(Microbeads—Myltenyi)
RPMI 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 Types Study Target Time of Intervention Main Performed Evaluations Results FRC Role in Immune Response
Aparicio-Domingo et al. [18] IL-33-GFP reporter mice LCMV 3 days/w
for 2 weeks
FC and RNA sequencing FRC is one important IL-33 source in LNs, vital for driving acute and chronic antiviral T cell responses. Anti-viral response
Dertschnig et al. [19] FRC and DC ablation in vivo; identification of PTA regulatory genes; BTM model induction GvHD 2 weeks FC, RNA sequencing, confocal microscopy The loss of PTA presentation by FRCs during GVHD leads to permanent damage in their networks in lymphoid tissues. Control of peripheral tolerance
Eom et al. [20] Identification of distinctive subpopulations of CD90+ SCs present
in melanoma-infiltrated LNs
Melanoma NA FC, gene expression There 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] Tissue-engineered stromal reticula and FRC/T cell co-culture Type 1
diabetes
NA FC, immunofluorescence, imaging FRCs 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 T1D
Knop et al. [22] IL-7fl/fl mice and adoptive T cell transfer NA NA FC IL7, produced by LN FRCs-regulated T cell homeostasis, is crucial for TCM maintenance. IL7 produced by LN FRCs is crucial for TCM maintenance
Perez-Shibayama et al. [23] LCMV-infected mice, FRC ex vivo restimulation and cytokine production LCMV Armstrong 8 d FC IFNAR-dependent shift of FRC subsets toward an immunoregulatory state reduces exhaustive CD8+ T cell activation. IFN type 1 influences FRC peripheral tolerance
Brown et al. [24] FRC/T cell co-cultures Influenza and LCMV infection NR FC and RNA sequencing FRCs 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. FRCs influence the CD8 T cells fate
Kasinath et al. [25] Mouse FRC depletion and treatment with anti-PDPN antibody Crescentic Glomerulonephritis (GN) 3 d FC and gene expression Removal 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. Role of FRCs and PDPN expression in GN
Kelch et al. [26] 3D imaging and topological mapping NA NA EVIS imaging and confocal microscopy T 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 LNs
Majumder et al. [27] Metabolic assay Experimental autoimmune encephalomyelitis 7 d FC, immunoblotting, siRNA transfection During 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 assays Aging and influenza infection NR FC Age-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 LN
Schaeuble et al. [29] Nos2−/−, COX2−/− mice and FRC/T cell co-culture COX/Prostaglandin E2 pathway 4 d FC FRCs 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 proliferation
Dubrot et al. [30] Adoptive transfer T cells in RAG−/− mice and Treg suppression assay MHC II-induced expression by FRC and LEC and its impact on autoimmunity 5 d FC LNSCs inhibit autoreactive T-cell responses by directly presenting antigens through endogenous MHCII molecules. Control of peripheral tolerance in autoimmunity
Knoblich et al. [31] T cell and CAR T cell activation assay COX/Prostaglandin E2, iNOS, IDO and TGF-β pathways in FRCs, NA FC and RNA sequencing 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] FRC labeling and injection into mice Ischemic-reperfusion injury (IRI) NR FC, electron and confocal microscopy Depletion of FRCs reduced T cell activation in the kidney LNs and ameliorated renal injury in acute IRI. Role of FRCs in IRI
Chung et al. [33] FRC/T cell co-culture GvHD 4 h and 3 h FC FRCs 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 7 Tumor-draining LNs NA FC LN 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) NA FC and RNA sequencing Stromal 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-culture COX 2/Prostaglandin E2, iNOS, IDO and TGF-β pathways in FRCs 6 h FC COX2 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-culture COX 2/Prostaglandin E2 pathway in FRCs NA FC, WB Hyperactivity 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 regulation FRC secretion of IL-15 3 h FC FRC secretion of IL-15 regulates homeostatic ILC1 and NK cell maintenance. Role of FRCs in innate in immunity
Novkovic et al. [39] FRC network topological analysis FRC network NA Intravital 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 response
Royer et al. [40] Adoptive transfer of T cells and T cell response to herpesvirus-associated lymphadenitis HSV-1 4 h FC Dissemination 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
Takeda et al. [41] Lymphocyte migration Ablation of LPA-producing ectoenzyme autotaxin in FRCs NA FC, IMS, Intravital TPM LPA 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.

This entry is adapted from the peer-reviewed paper 10.3390/cells10051150

This entry is offline, you can click here to edit this entry!