Next Article in Journal
A Robust Assay to Monitor Ataxin-3 Amyloid Fibril Assembly
Previous Article in Journal
When Electrospun Fiber Support Matters: In Vitro Ovine Long-Term Folliculogenesis on Poly (Epsilon Caprolactone) (PCL)-Patterned Fibers
Previous Article in Special Issue
The Roles of TNFR2 Signaling in Cancer Cells and the Tumor Microenvironment and the Potency of TNFR2 Targeted Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

LTα, TNF, and ILC3 in Peyer’s Patch Organogenesis

by
Violetta S. Gogoleva
1,
Dmitry V. Kuprash
1,
Sergei I. Grivennikov
1,†,
Alexei V. Tumanov
2,
Andrey A. Kruglov
1,3,4 and
Sergei A. Nedospasov
1,3,5,6,*
1
Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
2
Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229, USA
3
Belozersky Institute of Physico-Chemical Biology and Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
4
AG Chronic Inflammation, Deutsches Rheuma-Forschungszentrum (DRFZ), A Leibniz Institute, 10117 Berlin, Germany
5
Institute of Cell Biology and Neurobiology, Charite-Universitatsmedizin, 10117 Berlin, Germany
6
Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, 35340 Federal Territory Sirius, Russia
*
Author to whom correspondence should be addressed.
Current address: Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
Cells 2022, 11(12), 1970; https://doi.org/10.3390/cells11121970
Submission received: 8 May 2022 / Revised: 11 June 2022 / Accepted: 17 June 2022 / Published: 19 June 2022
(This article belongs to the Special Issue The TNF Receptor Superfamily in Health and Disease)

Abstract

:
TNF and LTα are structurally related cytokines of the TNF superfamily. Their genes are located in close proximity to each other and to the Ltb gene within the TNF/LT locus inside MHC. Unlike Ltb, transcription of Tnf and of Lta is tightly controlled, with the Tnf gene being an immediate early gene that is rapidly induced in response to various inflammatory stimuli. Genes of the TNF/LT locus play a crucial role in lymphoid tissue organogenesis, although some aspects of their specific contribution remain controversial. Here, we present new findings and discuss the distinct contribution of TNF produced by ILC3 cells to Peyer’s patch organogenesis.

1. Introduction

Tumor necrosis factor (TNF) and lymphotoxin alpha (LTα) are the first molecularly cloned cytokines of the TNF superfamily with similar cytotoxic activities of their recombinant gene products [1]. Both TNF and LTα can form soluble homotrimers, sTNF and sLTα3, respectively, that bind to and signal through both TNFR1 and TNFR2 [2], although some studies question the physiological relevance of LTα3-TNFR2 interaction [3,4]. Renaming of lymphotoxin as TNFβ caused some confusion, which was resolved later with the discovery of LTβ [5], uncovering a constellation of LTα functions that are not shared by TNF [6].
Although LTα can be a soluble TNF-like factor in vivo [7], its major forms are membrane-bound heterotrimeric complexes with LTβ, such as LTα1β2 and LTα2β1. While LTα1β2 signals through unique LTβR [8], LTα2β1 can interact both with TNFR1 and TNFR2 [9]. A further complication in the TNF/LT ligand–receptor system is the fact that LIGHT, another member of TNF superfamily, being a ligand for HVEM, may also serve as a ligand for LTβR [10].
Since biologics targeting TNF (or both TNF and soluble LTα) are used for treatment of autoimmune diseases, it is of great importance to understand all functions mediated by the ligands of TNF/LT system in normal physiology. As models for such an analysis, multiple engineered mouse strains with complete or conditional inactivation of corresponding genes were generated (reviewed in [11,12]). These studies unexpectedly uncovered that both TNF and LT are involved in the development and the structure-functional organization of secondary lymphoid organs, including lymph nodes and Peyer’s patches (PP) in the small intestine. PP development during gestation relies on the activation of the LTα1β2/LTβR axis, as PP are completely absent in all independently generated Lta-, Ltb- and Ltbr-deficient mice [13,14,15,16,17]. As it was earlier demonstrated for lymph node development, PP formation requires expression of LTα1β2 on the lymphoid-tissue inducer (LTi) cell, a specialized type 3 innate lymphoid cell [18]. However, uniquely for PP, LTα1β2 expression on LTi is induced after the activation of tyrosine kinase receptor RET signaling in CD11c+LTα1β2+ haematopoietic cells [19]. LTα1β2-expressing LTi then activate LTβR+ mesenchymal cells, leading to the upregulation of cell adhesion molecules and chemokines, such as CXCL13, CCL19 and CCL21, that attract and retain lymphocytes at the site of the developing PP [20].
Although we have a detailed knowledge of how lymph node development is orchestrated by TNF superfamily molecules, some issues concerning PP development remain unresolved. First, the physiological thresholds of TNF/LTα signals required for proper PP development remain debatable. For example, double heterozygosity of both Lta and Ltb results in the complete lack of PP, implicating the importance of gene dosage during the process of PP formation [21]. Second, the contribution of TNF to PP development also remains controversial. Several independently generated Tnfr1-deficient mouse strains display a range of defects in PP organogenesis [22,23,24]. In line with this, PP formation in several independently designed Tnf-deficient mouse strains was either reduced [23,25] or completely absent [26]. To address the role of TNF and LTα in the process of PP formation in this study, we relied on several neo-free knockout strains generated using LoxP-Cre technology [14,16,26]. Our findings with these mice do not support earlier conclusions on the threshold requirements for LTα/LTβ signal involvement and they implicate a role for TNF produced by ILC3 in PP development.

2. Materials and Methods

2.1. Mice

LtaΔ/Δ [14], LtbΔ/Δ [16], TnfΔ/Δ [26], Tnffl/fl, TnfΔLysM, TnfΔT [27] mice were described previously. Tnfrsf1a−/− (Tnfr1−/−) were kindly provided by Prof. K. Pfeffer [28]. TnfΔILC3,T mice were generated by crossing Tnffl/fl and Rorc(γt)-CreTg mice [11]. TnfΔCD11c were generated by crossing Tnffl/fl and CD11c-Cre mice [29]. Experimental mice were either generated on or extensively backcrossed onto a C57BL/6 genetic background. Heterozygous (Lta+/Δ) and littermate (Tnffl/fl) controls were used as wild-type controls. All mice analyzed were sex- and age-matched (8–12 weeks old). Mice were bred and housed under the standard conditions of the Animal Breeding Facility, BIBCh, RAS (the Unique Research Unit Bio-Model of the IBCh, RAS; the Bioresource Collection—Collection of SPF-Laboratory Rodents for Fundamental, Biomedical and Pharmacological Studies under contract #075-15-2021-1067 with the Ministry of Science and Higher Education of the Russian Federation) accredited at the international level by AAALAC. All animal experiments were approved by the local authorities IACUC committee of BIBCh RAS (protocol no. 125, 29 December 2020) and performed according to institutional guidelines and Bioethics Committee of the Engelhardt Institute of Molecular Biology, Russian Academy of Sciences.

2.2. Visualization of Peyer’s Patches

The small intestine was removed and placed in 10% acetic acid for 5–10 min, then Peyer’s patches were counted. Images of intestines were taken with a consumer digital camera. Whole-mount immunostaining was carried out as described in [26]. For staining of the PP anlagen, rat anti-MAdCAM-1 mAb MECA-367 (BD Biosciences, San Jose, CA, USA) was used with horseradish peroxidase conjugated anti-rat Ig (Jackson ImmunoResearch, West Grove, PA, USA) as a secondary antibody, and SIGMA FAST DAB with a metal enhancer (Sigma-Aldrich, St. Louis, MO, USA) was used for the enzymatic reaction.

2.3. Cell Isolation from the Intestinal Lamina Propria

The small intestines were extracted and placed into the ice-cold PBS. Peyer’s patches and fat were removed. The small intestine was cut longitudinally and cleaned of the feces. Subsequently, the tissue was twice incubated in HBSS supplemented with 10 mM HEPES and 5 mM EDTA at 37 °C for 20 min. Samples were twice washed, minced, and incubated in HBSS supplemented with 2% FBS, 0.5 mg/mL Collagenase D (Roche, Mannheim, Germany), 0.5 mg/mL DNase I (Sigma Aldrich), 0.5 U/mL Dispase (Gibco, Waltham, MA, USA) at 37 °C for 20 min. After filtering through a 70 μm strainer, cells were centrifuged in a 40/80 Percoll gradient [30].

2.4. Flow Cytometry and Cell Sorting

Cells were stained with Fixable Viability Dye (eBioscience, San Diego, CA, USA) and the following antibodies: anti-CD45 (30-F11, BioLegend, London, UK), anti-CD4 (RM4-5, BioLegend), anti-CCR6 (29-2L17, BioLegend), anti-IL-7Rα (A7R34, Invitrogen, Waltham, MA, USA), Mouse Hematopoietic Lineage Cocktail (eBioscience): anti-CD3 (17A2), anti-B220 (RA3-6B2), anti-CD11b (M1/70), anti-TER-119 (TER-119), anti-Gr-1 (RB6-8C5). Intracellular staining was performed using an eBioscience Foxp3/Transcription Factor Staining Buffer Set and the following antibodies: anti-RORγt (B2D, eBioscience), anti-GATA3 (16E10A23, BioLegend). Flow cytometric analysis was performed using FACSAria III flow cytometer (BD Bioscience) and FlowJo software (Tree Star). Cell sorting of splenic ILC3 and CD4+ T cells was performed using anti-CD4 (GK1.5; eBioscience) and Hematopoietic Lineage Cocktail (anti-CD3, anti-CD19, anti-CD11c) on FACSAria (BD Bioscience). ILC3 were identified as LineageCD4+ cells, CD4+ T cells were identified as Lineage+CD4+ cells. Purity was more than 90%.

2.5. Quantitative PCR Analysis

Total RNA was isolated using TRI reagent (Sigma-Aldrich), according to the manufacturer’s protocol. RNA was treated with DNase I (Thermo Fischer Scientific, Waltham, MA, USA; Cat #: EN0525), according to the manufacturer’s instructions. To transcribe RNA to cDNA, random hexamer primers (TIB Molbiol, Berlin, Germany) and RevertAid H Minus reverse transcriptase (Thermo Fischer Scientific; Cat #: EP0451) were used, according to the manufacturer’s protocol. The generated single-stranded cDNA was directly used for quantitative real-time PCR. cDNA transcript expression of indicated genes was analyzed with Maxima SYBR Green/ROX qPCR master mix (Thermo Fischer Scientific; Cat #: K0222), according to the manufacturer’s protocol by using a StepOne Plus Real Time PCR system (Applied Biosystems, Foster City, CA, USA) or Mx3000P qPCR system (Stratagene, La Jolla, CA, USA).

2.6. Statistical Analysis

All statistical analyses were performed using Prism 9 software (GraphPad, San Diego, CA, USA). Unpaired t test and one-way ANOVA tests were used. p values of <0.05 were considered significant.

3. Results

3.1. Physiological Thresholds for LTα and TNF Signaling in Peyer’s Patch Development

The differential contribution of LTα and LTβ to the development of mesenteric lymph nodes implicated distinct physiological thresholds for TNF- and lymphotoxin-dependent activation of mesenchymal cells. Interestingly, Koni et al. reported that unlike single heterozygotes, the double heterozygous Lta+/−Ltb+/− mice completely lacked PP [21]. However, this phenotype may be explained by the possible cooperation of TNFR1 and LTβR downstream pathways in PP formation [21]. Additionally, both Lta−/− [13] and Ltb−/− [15] knockout mice retained the neo cassette that could possibly affect the expression of neighboring genes, for example, Tnf (Figure S1A,B). Importantly, the presence of the neo cassette may affect the expression of neighboring genes [31,32]. Therefore, we reexamined the effects of the LTα/LTβ gene dosage in PP organogenesis using neo-free LtaΔ/Δ and LtbΔ/Δ mice [14,16] on a pure C57BL/6 background (Figure S1A,B). Surprisingly, these “alternative” Lta+/Δ/Ltb+/Δ double heterozygotes developed PP normally, as did single heterozygous Lta+/Δ (Figure 1A) and Ltbr+/− mice (data not shown).
We then crossed the conventional Lta−/− mice [13] with neo-free LtbΔ/Δ mice and observed an intermediate phenotype with a significantly reduced numbers of PP (Figure 1A). As reported earlier, Lta−/− mice, unlike LtaΔ/Δ mice, showed a diminished ability of myeloid cells to produce TNF due to the interference of the neo cassette with the far upstream elements of TNF promoter/enhancer region [14,33]. These results suggest that impaired TNF production is responsible for defective PP development in LTα haploinsufficient mice. Of note, the PP development was not affected in conventional single heterozygous Lta+/− mice [21], suggesting that Ltb gene dosage may also be important at certain gestational steps in PP formation.

3.2. TNF Produced by ILC3 Is Critical for Peyer’s Patches Development

Since it is known that soluble LTα3 and TNF share the TNFR1 but not the LTβR receptor, we next re-addressed the requirement for TNF and TNFR1 during PP development. As reported earlier, neither neo-free TnfΔ/Δ (Figure S1C) nor Tnfr1-deficient mice— both on a C57BL/6 background—developed PP (Figure 1B), excluding the effects of genetic modifiers affecting the phenotype [28].
Analysis of TnfΔ/Δ E16.5 embryos showed that they lacked PP anlagen [26]. This finding indicates that TNF may be required during the early stages of PP development, i.e., at the induction phase of LTα1β2 expression or clustering of LTβR+ mesenchymal cells and LTi cells. As RET+CD11c+ hematopoietic progenitor cells contribute to the early steps of PP formation [18] we therefore analyzed PP development in mice with the inactivation of TNF in LysM+ cells [34] and CD11c+ cells (Figure 2A).
We found that TNF derived from LysM+ myeloid cells and CD11c+ cells is dispensable to PP formation (Figure 2A).
To define the contribution of ILC3-derived TNF, we generated mice with conditional inactivation of TNF in ILC3 and T cells by crossing Tnffl/fl and Rorc(γt)-CreTg mice [11]. Since ILC3 cells are known to express high levels of LTα, LTβ, and TNF mRNA [35], we first analyzed the expression of TNF/LT locus genes in purified ILC3 and CD4+ T cells isolated from the spleens of adult TnfΔILC3,T and Tnffl/fl mice. We found no difference in LTα and LTβ mRNA levels between wild-type and TnfΔILC3,T mice (Figure 2B) and no detectable TNF mRNA in either ILC3 or CD4+ T cells (Figure 2B). Importantly, TnfΔILC3,T mice completely lacked PP, whereas TnfΔT displayed normal PP numbers (Figure 2A). These results reveal that ILC3-derived TNF is critical for PP organogenesis. To sum up, these results suggest that activation of the TNF/TNFR1 axis is important for PP formation, with the specific contribution of ILC3-derived TNF.

3.3. ILC3 in PP Development and in Adult Lamina Propria

Peyer’s patches represent secondary lymphoid organs in the small intestine implicated in both the induction of immune tolerance and in defense against certain microorganisms [36]. Although 8 week old TnfΔ/Δ and TnfΔILC3,T mice showed a complete absence of PP, they both developed a single lymphoid patch at the cecum of the large intestine (Figure 3A).
These results indicate that TNF is dispensable for cecal patch development. Further immunohistochemical staining for MAdCAM-1 revealed the absence of PP anlagen in newborn TnfΔ/Δ and TnfΔILC3,T mice (Figure 3A).
Since ILC3 subsets are involved in both lymphoid organ formation and in regulation of gut immune homeostasis, we next analyzed ILC populations in the lamina propria of adult TNF/LT deficient animals (Figure S2). We found that the frequency of RORγt+ ILC3 in small intestine of Lta-deficient mice increased (Figure 3B). Similarly, the frequency of RORγt+ ILC3 in mice with either Tnf or Tnfr1 deficiency increased compared to the Tnffl/fl control mice (Figure 3C). Thus, we suggest that the absence of PP is not due to reduced numbers of ILC3. Moreover, these results indicate that TNF/LT ligands not only regulate PP development during embryogenesis but also control the homeostasis of ILC3 in the intestines of adult animals.

4. Discussion

TNF and LTα are structurally and functionally related cytokines of the TNF superfamily. They mediate multiple functions, including organogenesis and structural maintenance of lymphoid tissue [37,38]. However, the requirements of TNF and LTα during development may vary for different secondary lymphoid organs [39]. For example, lymph node development (except for mesenteric and sacral LNs) strictly depends on the activation of LTα1β2 signaling [18], while TNF seems to be dispensable in this process, as lymph nodes are present in mice with Tnf [23,26], Tnfr1 [22,23], or Tnfr2 deletion [40]. However, TNF clearly contributes to the functional microarchitecture of the spleen and the lymph nodes [41,42].
The significance of functional allele numbers, a so-called gene dosage effect, in genetically modified mice was demonstrated for both TNF and LTα. For example, mice heterozygous for both Tnf and Lta deficiency produced decreased plasma TNF levels in response to LPS stimulation as compared to wild-type mice [43]. Our demonstration of normal PP development in Lta+/Δ/Ltb+/Δ double heterozygous mice challenges the interpretation of Koni et al. [21] and calls for an alternative explanation of that apparent paradox. First of all, the requirements of TNF and TNFR1 in PP organogenesis remain controversial, partly due to the close linkage of Tnf with Lta and Ltb and of Tnfr1 with Ltbr. Possibly, the differences in targeting designs for Tnf- and Tnfr1-deficient mice led to inconsistent phenotypes, particularly concerning PP development. In the current study, we confirmed the complete absence of PP in neo-free TnfΔ/Δ and Tnfr1-deficient mice on a C57BL/6 background. We speculate that the presence of the neo cassette in conventional Tnf−/− mice and/or the possible contribution of genetic modifiers in Tnfr1−/− mice generated on a mixed 129Sv x C57BL/6 background with reduced PP number [23] may have caused a compensatory upregulation of lymphotoxin genes.
With regard to the cellular source of TNF, we identified RORγt+ ILC3 but not CD11c+ cells as the critical contributor to PP development. ILC3-derived TNF specifically regulates formation of PP in the small intestine but not of the cecal patch in the large intestine. In line with our study, previous work demonstrated that the mechanism of development and maintenance of the cecal patch is distinct from PP and colonic solitary intestinal lymphoid tissues [44,45].
Moreover, ILC3-derived TNF is critical for the gestational development of PP anlagen. Interestingly, in the adult organism TNF clearly contributes to the maintenance of PP. Specifically, B cell-derived TNF controls the development of follicular dendritic cells and B-cell follicles in PP [42,46]. Based on our data, it appears that TNF and LTα regulate the frequency of ILC3 in the intestinal lamina propria. We also do not exclude a possibility that increased numbers of ILC3 in the intestines of PP-deficient neo-free TnfΔ/Δ and LtaΔ/Δ mice is a result of compensatory feedback loop mechanism.
As a result, we propose an updated model of a stepwise contribution of TNF and LTα in PP development (Figure 4).
The first step (E15.5) is the appearance of VCAM-1+ stromal cells clusters, also known as lymphoid tissue organizer cells (LTo) [47]. This step starts from the interaction of VCAM-1+ stromal cells producing artemin (ARTN) with RET+c-Kit+CD11c+CD4CD3IL-7Ra hematopoietic progenitor cells, also known as lymphoid tissue initiator cells (LTin) [19,48]. Initial adhesion and clustering of these cells is dependent on ART-RET interactions and is LTi-independent [48]. These CD11c+ hematopoietic progenitors start to express LTα1β2 at higher levels than the LTi cells at E15.5, possibly due to the activation of RET signaling [19]. However, RET stimulation of explanted cultures from intestines with ART and other RET ligands failed to induce LTβ expression by LTin cells [48]. It is possible that increased LTα1β2 levels by LTin cells could be due to LTα upregulation, as transcription of LTA gene in various cell types is tightly controlled as compared to LTβ. This is consistent with data regarding the functional activity of an LTα1β2 complex and that the LTα subunit is a limiting component for the assembly of a functional heteromeric LTα1β2 complex [49]. LTα1β2 on CD11c+ cells then enables their interaction with LTβR on stromal cells suggesting that this may be a first checkpoint dependent on TNF superfamily ligands signaling.
At the second step (E17.5), more LTi cells are recruited to the PP anlagen. Stromal cells produce IL-7 which promotes the survival of LTα1β2-expressing LTi cells, leading to their interaction with LTβR+ stromal cells [50,51]. Here, we propose that in addition to LTα1β2, LTi cells may produce TNF, because deletion of Tnf in ILC3 results in a complete block of PP development. Interestingly, membrane-bound TNF on LTi cells appears to be involved, as mice expressing only this form of TNF develop PP with relatively normal microarchitecture [52]. Although we cannot draw a definite conclusion as to which step of PP development is controlled by TNF, data of TnfΔ/Δ embryos that lacked PP anlagen at E16.5 [26] suggest that TNF may be critical during the first two steps of PP formation. Hypothetically, LTin and LTi cells may produce TNF and sLTα3 that interact with TNFR1 on an unknown cell, since Tnfr1- but not Tnfr2-deficient mice lack PP. Recent analysis of mice with conditional inactivation of TNFR1 in CCL19-Cre and Col6A1-Cre expressing cells revealed reduced B cell follicles and the impaired stromal microarchitecture of PP [53]. Additionally, single cell RNA seq analysis and fate mapping experiments identified two distinct fibroblastic reticular cell lineages in PP anlagen at E18.5: perivascular and subepithelial, which synergistically direct PP development in both LTβR- and TNFR1-dependent manner [53].
Starting from E17.5, the third step includes production of chemokines attracting hematopoietic cells to the PP, such as CXCL13, CCL19, and CCL21 [18]. These chemokines are produced by stromal cells via activation of non-canonical NF-κB [54]. Engagement of TNFR1 by TNF or LTα is known to activate the canonical NF-κB pathway, leading to the transcription of different target genes, including adhesion molecules [37,54]. Although the role of the non-canonical NF-κB pathway in the development of PP is well established [55,56], mice deficient in p50, one of the canonical NF-κB subunits, possess a significantly reduced number of PP [55]. Moreover, simultaneous deletion of TNFR1 and another NF-κB subunit, RelA, results in the absence of PP, which is explained by the dependence of stromal cells on RelA [57]. We propose that TNFR1-dependent activation of canonical NF-κB is not only crucial for the activation of various proinflammatory genes but also for some homeostatic functions, including the late steps of PP formation. The possible contribution of LTα3/TNFR1 to PP development should also be considered since its specific role in the organization of mucosal lymph nodes was demonstrated in double Ltb/Tnf-deficient mice [41]. This model, with distinct contributions of TNF and LTα molecules into PP formation, resembles the sequence of events described for mature isolated lymphoid follicles [58].

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cells11121970/s1, Figure S1: Comparison of different LTα, LTβ and TNF genes targeting strategies. Schematic representation of wild-type TNF/LT locus and (A) neo-free and conventional Lta-deficient mice, (B) neo-free and conventional Ltb-deficient mice, (C) neo-free Tnf-deficient mice. Figure S2: Flow cytometry gating strategy for identification of ILC subsets in the lamina propria of the small intestine. ILC3 were identified as ViaDyeCD45+Lin IL-7Rα+RORγt+ cells.

Author Contributions

D.V.K., A.A.K. and S.A.N. designed the research; V.S.G., D.V.K., S.I.G., A.V.T. and A.A.K. performed research and analyzed data; V.S.G., D.V.K., A.V.T., A.A.K. and S.A.N. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

The research was supported by the Russian Science Foundation (grant #19-75-30032). Breeding and genotyping of multiple mouse lines were supported by the Ministry of Science and Higher Education of the Russian Federation (grant #075-15-2019-1660). This research has been conducted in part in the frame of the Interdisciplinary Scientific and Educational School of Moscow University “Molecular Technologies of the Living Systems and Synthetic Biology”.

Institutional Review Board Statement

Animal manipulations were carried out in accordance with recommendations in the Guide for the Care and Use of Laboratory Animals (NRC 2011); the European Convention for the Protection of Vertebrate Animals Used for Experimental and Other Scientific Purposes, Council of Europe (ETS 123); and “The Guidelines for Manipulations with Experimental Animals” (the decree of the Presidium of the Russian Academy of Sciences of 2 April 1980, no. 12000–496). The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Scientific Council of the Engelhardt Institute of Molecular Biology RAS. Breeding and use of mice was approved by the Animal Breeding Facility of BIBCh RAS (AAALACi No. 001903) and the IACUC committee of the Branch of Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (protocol no. 125 and date 29 December 2020).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data supporting the reported results are available on request from the corresponding author.

Acknowledgments

The authors thank A. Dygay, M. Bondareva, E. Gubernatorova, E. Gorshkova, M. Drutskaya, S. Kamalyan, and R. Zvartsev for their assistance and A. Newman for useful discussion. We are also indebted to Alexander Chernov for his help with maintenance of the mouse colony.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gray, P.W.; Aggarwal, B.B.; Benton, C.V.; Bringman, T.S.; Henzel, W.J.; Jarrett, J.A.; Leung, D.W.; Moffat, B.; Ng, P.; Svedersky, L.P.; et al. Cloning and expression of cDNA for human lymphotoxin, a lymphokine with tumour necrosis activity. Nature 1984, 312, 721–724. [Google Scholar] [CrossRef] [PubMed]
  2. Etemadi, N.; Holien, J.K.; Chau, D.; Dewson, G.; Murphy, J.M.; Alexander, W.S.; Parker, M.W.; Silke, J.; Nachbur, U. Lymphotoxin alpha induces apoptosis, necroptosis and inflammatory signals with the same potency as tumour necrosis factor. FEBS J. 2013, 280, 5283–5297. [Google Scholar] [CrossRef] [PubMed]
  3. Medvedev, A.E.; Espevik, T.; Ranges, G.; Sundan, A. Distinct roles of the two tumor necrosis factor (TNF) receptors in modulating TNF and lymphotoxin alpha effects. J. Biol. Chem. 1996, 271, 9778–9784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Bossen, C.; Ingold, K.; Tardivel, A.; Bodmer, J.L.; Gaide, O.; Hertig, S.; Ambrose, C.; Tschopp, J.; Schneider, P. Interactions of tumor necrosis factor (TNF) and TNF receptor family members in the mouse and human. J. Biol. Chem. 2006, 281, 13964–13971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Browning, J.L.; Ngam-ek, A.; Lawton, P.; DeMarinis, J.; Tizard, R.; Chow, E.P.; Hession, C.; O’Brine-Greco, B.; Foley, S.F.; Ware, C.F. Lymphotoxin beta, a novel member of the TNF family that forms a heteromeric complex with lymphotoxin on the cell surface. Cell 1993, 72, 847–856. [Google Scholar] [CrossRef]
  6. Nedospasov, S.A. There exists only one tumor necrosis factor. Med. Immunol. 2020, 22, 1221–1224. [Google Scholar] [CrossRef]
  7. Kruglov, A.A.; Grivennikov, S.I.; Kuprash, D.V.; Winsauer, C.; Prepens, S.; Seleznik, G.M.; Eberl, G.; Littman, D.R.; Heikenwalder, M.; Tumanov, A.V.; et al. Nonredundant function of soluble LTalpha3 produced by innate lymphoid cells in intestinal homeostasis. Science 2013, 342, 1243–1246. [Google Scholar] [CrossRef] [Green Version]
  8. Crowe, P.D.; VanArsdale, T.L.; Walter, B.N.; Ware, C.F.; Hession, C.; Ehrenfels, B.; Browning, J.L.; Din, W.S.; Goodwin, R.G.; Smith, C.A. A lymphotoxin-beta-specific receptor. Science 1994, 264, 707–710. [Google Scholar] [CrossRef] [PubMed]
  9. Kucka, K.; Lang, I.; Zhang, T.; Siegmund, D.; Medler, J.; Wajant, H. Membrane lymphotoxin-alpha2beta is a novel tumor necrosis factor (TNF) receptor 2 (TNFR2) agonist. Cell Death Dis. 2021, 12, 360. [Google Scholar] [CrossRef]
  10. Mauri, D.N.; Ebner, R.; Montgomery, R.I.; Kochel, K.D.; Cheung, T.C.; Yu, G.L.; Ruben, S.; Murphy, M.; Eisenberg, R.J.; Cohen, G.H.; et al. LIGHT, a new member of the TNF superfamily, and lymphotoxin alpha are ligands for herpesvirus entry mediator. Immunity 1998, 8, 21–30. [Google Scholar] [CrossRef] [Green Version]
  11. Kruglov, A.A.; Kuchmiy, A.; Grivennikov, S.I.; Tumanov, A.V.; Kuprash, D.V.; Nedospasov, S.A. Physiological functions of tumor necrosis factor and the consequences of its pathologic overexpression or blockade: Mouse models. Cytokine Growth Factor Rev. 2008, 19, 231–244. [Google Scholar] [CrossRef] [PubMed]
  12. Koroleva, E.P.; Fu, Y.X.; Tumanov, A.V. Lymphotoxin in physiology of lymphoid tissues—Implication for antiviral defense. Cytokine 2018, 101, 39–47. [Google Scholar] [CrossRef] [PubMed]
  13. De Togni, P.; Goellner, J.; Ruddle, N.H.; Streeter, P.R.; Fick, A.; Mariathasan, S.; Smith, S.C.; Carlson, R.; Shornick, L.P.; Strauss-Schoenberger, J.; et al. Abnormal development of peripheral lymphoid organs in mice deficient in lymphotoxin. Science 1994, 264, 703–707. [Google Scholar] [CrossRef] [PubMed]
  14. Liepinsh, D.J.; Grivennikov, S.I.; Klarmann, K.D.; Lagarkova, M.A.; Drutskaya, M.S.; Lockett, S.J.; Tessarollo, L.; McAuliffe, M.; Keller, J.R.; Kuprash, D.V.; et al. Novel lymphotoxin alpha (LTalpha) knockout mice with unperturbed tumor necrosis factor expression: Reassessing LTalpha biological functions. Mol. Cell. Biol. 2006, 26, 4214–4225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Koni, P.A.; Sacca, R.; Lawton, P.; Browning, J.L.; Ruddle, N.H.; Flavell, R.A. Distinct roles in lymphoid organogenesis for lymphotoxins alpha and beta revealed in lymphotoxin beta-deficient mice. Immunity 1997, 6, 491–500. [Google Scholar] [CrossRef] [Green Version]
  16. Alimzhanov, M.B.; Kuprash, D.V.; Kosco-Vilbois, M.H.; Luz, A.; Turetskaya, R.L.; Tarakhovsky, A.; Rajewsky, K.; Nedospasov, S.A.; Pfeffer, K. Abnormal development of secondary lymphoid tissues in lymphotoxin beta-deficient mice. Proc. Natl. Acad. Sci. USA 1997, 94, 9302–9307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Futterer, A.; Mink, K.; Luz, A.; Kosco-Vilbois, M.H.; Pfeffer, K. The lymphotoxin beta receptor controls organogenesis and affinity maturation in peripheral lymphoid tissues. Immunity 1998, 9, 59–70. [Google Scholar] [CrossRef] [Green Version]
  18. van de Pavert, S.A.; Mebius, R.E. New insights into the development of lymphoid tissues. Nat. Rev. Immunol. 2010, 10, 664–674. [Google Scholar] [CrossRef]
  19. Veiga-Fernandes, H.; Coles, M.C.; Foster, K.E.; Patel, A.; Williams, A.; Natarajan, D.; Barlow, A.; Pachnis, V.; Kioussis, D. Tyrosine kinase receptor RET is a key regulator of Peyer’s patch organogenesis. Nature 2007, 446, 547–551. [Google Scholar] [CrossRef]
  20. Honda, K.; Nakano, H.; Yoshida, H.; Nishikawa, S.; Rennert, P.; Ikuta, K.; Tamechika, M.; Yamaguchi, K.; Fukumoto, T.; Chiba, T.; et al. Molecular basis for hematopoietic/mesenchymal interaction during initiation of Peyer’s patch organogenesis. J. Exp. Med. 2001, 193, 621–630. [Google Scholar] [CrossRef]
  21. Koni, P.A.; Flavell, R.A. A role for tumor necrosis factor receptor type 1 in gut-associated lymphoid tissue development: Genetic evidence of synergism with lymphotoxin beta. J. Exp. Med. 1998, 187, 1977–1983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Neumann, B.; Luz, A.; Pfeffer, K.; Holzmann, B. Defective Peyer’s patch organogenesis in mice lacking the 55-kD receptor for tumor necrosis factor. J. Exp. Med. 1996, 184, 259–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Pasparakis, M.; Alexopoulou, L.; Grell, M.; Pfizenmaier, K.; Bluethmann, H.; Kollias, G. Peyer’s patch organogenesis is intact yet formation of B lymphocyte follicles is defective in peripheral lymphoid organs of mice deficient for tumor necrosis factor and its 55-kDa receptor. Proc. Natl. Acad. Sci. USA 1997, 94, 6319–6323. [Google Scholar] [CrossRef] [Green Version]
  24. Peschon, J.J.; Torrance, D.S.; Stocking, K.L.; Glaccum, M.B.; Otten, C.; Willis, C.R.; Charrier, K.; Morrissey, P.J.; Ware, C.B.; Mohler, K.M. TNF receptor-deficient mice reveal divergent roles for p55 and p75 in several models of inflammation. J. Immunol. 1998, 160, 943–952. [Google Scholar]
  25. Korner, H.; Cook, M.; Riminton, D.S.; Lemckert, F.A.; Hoek, R.M.; Ledermann, B.; Kontgen, F.; de St Groth, B.F.; Sedgwick, J.D. Distinct roles for lymphotoxin-alpha and tumor necrosis factor in organogenesis and spatial organization of lymphoid tissue. Eur. J. Immunol. 1997, 27, 2600–2609. [Google Scholar] [CrossRef] [PubMed]
  26. Kuprash, D.V.; Tumanov, A.V.; Liepinsh, D.J.; Koroleva, E.P.; Drutskaya, M.S.; Kruglov, A.A.; Shakhov, A.N.; Southon, E.; Murphy, W.J.; Tessarollo, L.; et al. Novel tumor necrosis factor-knockout mice that lack Peyer’s patches. Eur. J. Immunol. 2005, 35, 1592–1600. [Google Scholar] [CrossRef] [PubMed]
  27. Grivennikov, S.I.; Tumanov, A.V.; Liepinsh, D.J.; Kruglov, A.A.; Marakusha, B.I.; Shakhov, A.N.; Murakami, T.; Drutskaya, L.N.; Forster, I.; Clausen, B.E.; et al. Distinct and nonredundant in vivo functions of TNF produced by t cells and macrophages/neutrophils: Protective and deleterious effects. Immunity 2005, 22, 93–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Pfeffer, K.; Matsuyama, T.; Kundig, T.M.; Wakeham, A.; Kishihara, K.; Shahinian, A.; Wiegmann, K.; Ohashi, P.S.; Kronke, M.; Mak, T.W. Mice deficient for the 55 kd tumor necrosis factor receptor are resistant to endotoxic shock, yet succumb to L. monocytogenes infection. Cell 1993, 73, 457–467. [Google Scholar] [CrossRef]
  29. Caton, M.L.; Smith-Raska, M.R.; Reizis, B. Notch-RBP-J signaling controls the homeostasis of CD8- dendritic cells in the spleen. J. Exp. Med. 2007, 204, 1653–1664. [Google Scholar] [CrossRef] [Green Version]
  30. Gronke, K.; Kofoed-Nielsen, M.; Diefenbach, A. Isolation and Flow Cytometry Analysis of Innate Lymphoid Cells from the Intestinal Lamina Propria. Methods Mol. Biol. 2017, 1559, 255–265. [Google Scholar] [CrossRef]
  31. Pham, C.T.; MacIvor, D.M.; Hug, B.A.; Heusel, J.W.; Ley, T.J. Long-range disruption of gene expression by a selectable marker cassette. Proc. Natl. Acad. Sci. USA 1996, 93, 13090–13095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. West, D.B.; Engelhard, E.K.; Adkisson, M.; Nava, A.J.; Kirov, J.V.; Cipollone, A.; Willis, B.; Rapp, J.; de Jong, P.J.; Lloyd, K.C. Transcriptome Analysis of Targeted Mouse Mutations Reveals the Topography of Local Changes in Gene Expression. PLoS Genet 2016, 12, e1005691. [Google Scholar] [CrossRef]
  33. Shakhov, A.N.; Collart, M.A.; Vassalli, P.; Nedospasov, S.A.; Jongeneel, C.V. Kappa B-type enhancers are involved in lipopolysaccharide-mediated transcriptional activation of the tumor necrosis factor alpha gene in primary macrophages. J. Exp. Med. 1990, 171, 35–47. [Google Scholar] [CrossRef] [PubMed]
  34. Shi, J.; Hua, L.; Harmer, D.; Li, P.; Ren, G. Cre Driver Mice Targeting Macrophages. Methods Mol. Biol. 2018, 1784, 263–275. [Google Scholar] [CrossRef] [PubMed]
  35. Gury-BenAri, M.; Thaiss, C.A.; Serafini, N.; Winter, D.R.; Giladi, A.; Lara-Astiaso, D.; Levy, M.; Salame, T.M.; Weiner, A.; David, E.; et al. The Spectrum and Regulatory Landscape of Intestinal Innate Lymphoid Cells Are Shaped by the Microbiome. Cell 2016, 166, 1231–1246 e1213. [Google Scholar] [CrossRef]
  36. Jung, C.; Hugot, J.P.; Barreau, F. Peyer’s Patches: The Immune Sensors of the Intestine. Int. J. Inflam. 2010, 2010, 823710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Atretkhany, K.N.; Gogoleva, V.S.; Drutskaya, M.S.; Nedospasov, S.A. Distinct modes of TNF signaling through its two receptors in health and disease. J. Leukoc. Biol. 2020, 107, 893–905. [Google Scholar] [CrossRef] [PubMed]
  38. Fu, Y.X.; Chaplin, D.D. Development and maturation of secondary lymphoid tissues. Annu. Rev. Immunol. 1999, 17, 399–433. [Google Scholar] [CrossRef] [PubMed]
  39. Randall, T.D.; Carragher, D.M.; Rangel-Moreno, J. Development of secondary lymphoid organs. Annu. Rev. Immunol. 2008, 26, 627–650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Erickson, S.L.; de Sauvage, F.J.; Kikly, K.; Carver-Moore, K.; Pitts-Meek, S.; Gillett, N.; Sheehan, K.C.; Schreiber, R.D.; Goeddel, D.V.; Moore, M.W. Decreased sensitivity to tumour-necrosis factor but normal T-cell development in TNF receptor-2-deficient mice. Nature 1994, 372, 560–563. [Google Scholar] [CrossRef]
  41. Kuprash, D.V.; Alimzhanov, M.B.; Tumanov, A.V.; Anderson, A.O.; Pfeffer, K.; Nedospasov, S.A. TNF and lymphotoxin beta cooperate in the maintenance of secondary lymphoid tissue microarchitecture but not in the development of lymph nodes. J. Immunol. 1999, 163, 6575–6580. [Google Scholar] [PubMed]
  42. Tumanov, A.V.; Grivennikov, S.I.; Kruglov, A.A.; Shebzukhov, Y.V.; Koroleva, E.P.; Piao, Y.; Cui, C.Y.; Kuprash, D.V.; Nedospasov, S.A. Cellular source and molecular form of TNF specify its distinct functions in organization of secondary lymphoid organs. Blood 2010, 116, 3456–3464. [Google Scholar] [CrossRef] [PubMed]
  43. Amiot, F.; Boussadia, O.; Cases, S.; Fitting, C.; Lebastard, M.; Cavaillon, J.M.; Milon, G.; Dautry, F. Mice heterozygous for a deletion of the tumor necrosis factor-alpha and lymphotoxin-alpha genes: Biological importance of a nonlinear response of tumor necrosis factor-alpha to gene dosage. Eur. J. Immunol. 1997, 27, 1035–1042. [Google Scholar] [CrossRef]
  44. Baptista, A.P.; Olivier, B.J.; Goverse, G.; Greuter, M.; Knippenberg, M.; Kusser, K.; Domingues, R.G.; Veiga-Fernandes, H.; Luster, A.D.; Lugering, A.; et al. Colonic patch and colonic SILT development are independent and differentially regulated events. Mucosal. Immunol. 2013, 6, 511–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Masahata, K.; Umemoto, E.; Kayama, H.; Kotani, M.; Nakamura, S.; Kurakawa, T.; Kikuta, J.; Gotoh, K.; Motooka, D.; Sato, S.; et al. Generation of colonic IgA-secreting cells in the caecal patch. Nat. Commun. 2014, 5, 3704. [Google Scholar] [CrossRef] [Green Version]
  46. Tumanov, A.V.; Kuprash, D.V.; Mach, J.A.; Nedospasov, S.A.; Chervonsky, A.V. Lymphotoxin and TNF produced by B cells are dispensable for maintenance of the follicle-associated epithelium but are required for development of lymphoid follicles in the Peyer’s patches. J. Immunol. 2004, 173, 86–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Adachi, S.; Yoshida, H.; Kataoka, H.; Nishikawa, S. Three distinctive steps in Peyer’s patch formation of murine embryo. Int. Immunol. 1997, 9, 507–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Patel, A.; Harker, N.; Moreira-Santos, L.; Ferreira, M.; Alden, K.; Timmis, J.; Foster, K.; Garefalaki, A.; Pachnis, P.; Andrews, P.; et al. Differential RET signaling pathways drive development of the enteric lymphoid and nervous systems. Sci. Signal. 2012, 5, ra55. [Google Scholar] [CrossRef]
  49. Williams-Abbott, L.; Walter, B.N.; Cheung, T.C.; Goh, C.R.; Porter, A.G.; Ware, C.F. The lymphotoxin-alpha (LTalpha) subunit is essential for the assembly, but not for the receptor specificity, of the membrane-anchored LTalpha1beta2 heterotrimeric ligand. J. Biol. Chem. 1997, 272, 19451–19456. [Google Scholar] [CrossRef] [Green Version]
  50. Yoshida, H.; Naito, A.; Inoue, J.; Satoh, M.; Santee-Cooper, S.M.; Ware, C.F.; Togawa, A.; Nishikawa, S.; Nishikawa, S. Different cytokines induce surface lymphotoxin-alphabeta on IL-7 receptor-alpha cells that differentially engender lymph nodes and Peyer’s patches. Immunity 2002, 17, 823–833. [Google Scholar] [CrossRef] [Green Version]
  51. Meier, D.; Bornmann, C.; Chappaz, S.; Schmutz, S.; Otten, L.A.; Ceredig, R.; Acha-Orbea, H.; Finke, D. Ectopic lymphoid-organ development occurs through interleukin 7-mediated enhanced survival of lymphoid-tissue-inducer cells. Immunity 2007, 26, 643–654. [Google Scholar] [CrossRef]
  52. Ruuls, S.R.; Hoek, R.M.; Ngo, V.N.; McNeil, T.; Lucian, L.A.; Janatpour, M.J.; Korner, H.; Scheerens, H.; Hessel, E.M.; Cyster, J.G.; et al. Membrane-bound TNF supports secondary lymphoid organ structure but is subservient to secreted TNF in driving autoimmune inflammation. Immunity 2001, 15, 533–543. [Google Scholar] [CrossRef] [Green Version]
  53. Prados, A.; Onder, L.; Cheng, H.W.; Morbe, U.; Lutge, M.; Gil-Cruz, C.; Perez-Shibayama, C.; Koliaraki, V.; Ludewig, B.; Kollias, G. Fibroblastic reticular cell lineage convergence in Peyer’s patches governs intestinal immunity. Nat. Immunol. 2021, 22, 510–519. [Google Scholar] [CrossRef]
  54. Dejardin, E.; Droin, N.M.; Delhase, M.; Haas, E.; Cao, Y.; Makris, C.; Li, Z.W.; Karin, M.; Ware, C.F.; Green, D.R. The lymphotoxin-beta receptor induces different patterns of gene expression via two NF-kappaB pathways. Immunity 2002, 17, 525–535. [Google Scholar] [CrossRef] [Green Version]
  55. Paxian, S.; Merkle, H.; Riemann, M.; Wilda, M.; Adler, G.; Hameister, H.; Liptay, S.; Pfeffer, K.; Schmid, R.M. Abnormal organogenesis of Peyer’s patches in mice deficient for NF-kappaB1, NF-kappaB2, and Bcl-3. Gastroenterology 2002, 122, 1853–1868. [Google Scholar] [CrossRef]
  56. Yilmaz, Z.B.; Weih, D.S.; Sivakumar, V.; Weih, F. RelB is required for Peyer’s patch development: Differential regulation of p52-RelB by lymphotoxin and TNF. EMBO J. 2003, 22, 121–130. [Google Scholar] [CrossRef] [Green Version]
  57. Alcamo, E.; Hacohen, N.; Schulte, L.C.; Rennert, P.D.; Hynes, R.O.; Baltimore, D. Requirement for the NF-kappaB family member RelA in the development of secondary lymphoid organs. J. Exp. Med. 2002, 195, 233–244. [Google Scholar] [CrossRef]
  58. Lorenz, R.G.; Chaplin, D.D.; McDonald, K.G.; McDonough, J.S.; Newberry, R.D. Isolated lymphoid follicle formation is inducible and dependent upon lymphotoxin-sufficient B lymphocytes, lymphotoxin beta receptor, and TNF receptor I function. J. Immunol. 2003, 170, 5475–5482. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Distinct contribution of various genetic deficiencies involving TNF/LT ligand–receptor system to Peyer’s patch development. (A) Peyer’s patches numbers in adult Ltafl/fl (n = 4), doubly heterozygous Lta+/ΔLtb+/Δ (n = 8), doubly heterozygous Lta+/−Ltb+/Δ (n = 3), single heterozygous Lta+/Δ (n = 9), and homozygous LtaΔ/Δ mice (n = 13); (B) Peyer’s patches numbers in adult Tnffl/fl (n = 11), Tnfr1−/− (n = 14), and TnfΔ/Δ (n = 5) mice. Each symbol represents an individual mouse; mean ± SEM. ** p < 0.01, **** p < 0.0001 (one-way ANOVA).
Figure 1. Distinct contribution of various genetic deficiencies involving TNF/LT ligand–receptor system to Peyer’s patch development. (A) Peyer’s patches numbers in adult Ltafl/fl (n = 4), doubly heterozygous Lta+/ΔLtb+/Δ (n = 8), doubly heterozygous Lta+/−Ltb+/Δ (n = 3), single heterozygous Lta+/Δ (n = 9), and homozygous LtaΔ/Δ mice (n = 13); (B) Peyer’s patches numbers in adult Tnffl/fl (n = 11), Tnfr1−/− (n = 14), and TnfΔ/Δ (n = 5) mice. Each symbol represents an individual mouse; mean ± SEM. ** p < 0.01, **** p < 0.0001 (one-way ANOVA).
Cells 11 01970 g001
Figure 2. ILC3-derived TNF is critical for Peyer’s patches development. (A) Peyer’s patches numbers in adult Tnffl/fl (n = 11), TnfΔLysM (n = 13), TnfΔCD11c (n = 4), TnfΔT (n = 4), and TnfΔILC3,T (n = 5) mice; (B) mRNA expression of LTα, LTβ, and TNF in purified ILC3 and CD4+ T cells isolated from the spleens of Tnffl/fl and TnfΔILC3,T mice. Each symbol represents an individual mouse; mean ± SEM. **** p < 0.0001 (one-way ANOVA).
Figure 2. ILC3-derived TNF is critical for Peyer’s patches development. (A) Peyer’s patches numbers in adult Tnffl/fl (n = 11), TnfΔLysM (n = 13), TnfΔCD11c (n = 4), TnfΔT (n = 4), and TnfΔILC3,T (n = 5) mice; (B) mRNA expression of LTα, LTβ, and TNF in purified ILC3 and CD4+ T cells isolated from the spleens of Tnffl/fl and TnfΔILC3,T mice. Each symbol represents an individual mouse; mean ± SEM. **** p < 0.0001 (one-way ANOVA).
Cells 11 01970 g002
Figure 3. LTα and TNF regulate ILC3 frequency in lamina propria in an adult organism. (A) Photographs of mouse intestine, from representative animals, showing the absence of Peyer’s patches and the presence of a cecal patch (left) and immunohistochemical staining of intestine from newborn mice (right). Arrows indicate Peyer’s patches (left) and areas of specific MAdCAM-1 staining (right), Scale bars, 4 mm; (B,C) Frequency of RORγt+ ILC3 cells and representative FACS plots of RORγt+ and GATA3+ cells among LinIL-7Rα+ cells gated on live CD45+ cells isolated from the lamina propria (right). Each symbol represents an individual mouse; mean ± SEM. * p < 0.05 (two-tailed unpaired Student’s t-test), ** p < 0.01 (one-way ANOVA).
Figure 3. LTα and TNF regulate ILC3 frequency in lamina propria in an adult organism. (A) Photographs of mouse intestine, from representative animals, showing the absence of Peyer’s patches and the presence of a cecal patch (left) and immunohistochemical staining of intestine from newborn mice (right). Arrows indicate Peyer’s patches (left) and areas of specific MAdCAM-1 staining (right), Scale bars, 4 mm; (B,C) Frequency of RORγt+ ILC3 cells and representative FACS plots of RORγt+ and GATA3+ cells among LinIL-7Rα+ cells gated on live CD45+ cells isolated from the lamina propria (right). Each symbol represents an individual mouse; mean ± SEM. * p < 0.05 (two-tailed unpaired Student’s t-test), ** p < 0.01 (one-way ANOVA).
Cells 11 01970 g003
Figure 4. A model of a stepwise contribution of LTα and TNF in Peyer’s patch development. Stromal cells produce ARTN which binds to RET receptor on a CD11c+ hematopoietic progenitor cell. RET activation may lead to the induction of LTα1β2 on CD11c+ cells. CD11c+ cells then interact with LTβR+ stromal cells which produce IL-7 for the survival of LTi cells. LTi cells start to express LTα1β2, which interacts with LTβR on stromal cells leading to the activation of the non-canonical NF-κB pathway. LTi cells may also express tmTNF, sTNF, and LTα3 that bind to TNFR1 on a yet unknown cell type, presumably on stromal cells, leading to the activation of canonical NF-κB pathway. Both non-canonical and canonical NF-κB pathways synergize to produce chemokines and adhesion molecules that attract and retain lymphocytes in PP. Created with BioRender.com.
Figure 4. A model of a stepwise contribution of LTα and TNF in Peyer’s patch development. Stromal cells produce ARTN which binds to RET receptor on a CD11c+ hematopoietic progenitor cell. RET activation may lead to the induction of LTα1β2 on CD11c+ cells. CD11c+ cells then interact with LTβR+ stromal cells which produce IL-7 for the survival of LTi cells. LTi cells start to express LTα1β2, which interacts with LTβR on stromal cells leading to the activation of the non-canonical NF-κB pathway. LTi cells may also express tmTNF, sTNF, and LTα3 that bind to TNFR1 on a yet unknown cell type, presumably on stromal cells, leading to the activation of canonical NF-κB pathway. Both non-canonical and canonical NF-κB pathways synergize to produce chemokines and adhesion molecules that attract and retain lymphocytes in PP. Created with BioRender.com.
Cells 11 01970 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gogoleva, V.S.; Kuprash, D.V.; Grivennikov, S.I.; Tumanov, A.V.; Kruglov, A.A.; Nedospasov, S.A. LTα, TNF, and ILC3 in Peyer’s Patch Organogenesis. Cells 2022, 11, 1970. https://doi.org/10.3390/cells11121970

AMA Style

Gogoleva VS, Kuprash DV, Grivennikov SI, Tumanov AV, Kruglov AA, Nedospasov SA. LTα, TNF, and ILC3 in Peyer’s Patch Organogenesis. Cells. 2022; 11(12):1970. https://doi.org/10.3390/cells11121970

Chicago/Turabian Style

Gogoleva, Violetta S., Dmitry V. Kuprash, Sergei I. Grivennikov, Alexei V. Tumanov, Andrey A. Kruglov, and Sergei A. Nedospasov. 2022. "LTα, TNF, and ILC3 in Peyer’s Patch Organogenesis" Cells 11, no. 12: 1970. https://doi.org/10.3390/cells11121970

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop