scholarly journals Lymph node stromal CCL2 limits antibody responses

2020 ◽  
Vol 5 (45) ◽  
pp. eaaw0693 ◽  
Author(s):  
Dragos C. Dasoveanu ◽  
Hyeung Ju Park ◽  
Catherine L. Ly ◽  
William D. Shipman ◽  
Susan Chyou ◽  
...  

Nonhematopoietic stromal cells in lymph nodes such as fibroblastic reticular cells (FRCs) can support the survival of plasmablasts and plasma cells [together, antibody-forming cells (AFCs)]. However, a regulatory function for the stromal compartment in AFC accumulation has not been appreciated. Here, we show that chemokine ligand 2 (CCL2)–expressing stromal cells limit AFC survival. FRCs express high levels of CCL2 in vessel-rich areas of the T cell zone and the medulla, where AFCs are located. FRC CCL2 is up-regulated during AFC accumulation, and we use lymph node transplantation to show that CCL2 deficiency in BP3+ FRCs and lymphatic endothelial cells increases AFC survival without affecting B or germinal center cell numbers. Monocytes are key expressers of the CCL2 receptor CCR2, as monocyte depletion and transfer late in AFC responses increases and decreases AFC accumulation, respectively. Monocytes express reactive oxygen species (ROS) in an NADPH oxidase 2 (NOX2)–dependent manner, and NOX2-deficient monocytes fail to reduce AFC numbers. Stromal CCL2 modulates both monocyte accumulation and ROS production, and is regulated, in part, by manipulations that modulate vascular permeability. Together, our results reveal that the lymph node stromal compartment, by influencing monocyte accumulation and functional phenotype, has a regulatory role in AFC survival. Our results further suggest a role for inflammation-induced vascular activity in tuning the lymph node microenvironment. The understanding of stromal-mediated AFC regulation in vessel-rich environments could potentially be harnessed to control antibody-mediated autoimmunity.

2022 ◽  
Vol 12 ◽  
Author(s):  
Liwei Jiang ◽  
Mine Yilmaz ◽  
Mayuko Uehara ◽  
Cecilia B. Cavazzoni ◽  
Vivek Kasinath ◽  
...  

Lymph node (LN)-resident stromal cells play an essential role in the proper functioning of LNs. The stromal compartment of the LN undergoes significant compensatory changes to produce a milieu amenable for regulation of the immune response. We have identified a distinct population of leptin receptor-expressing (LepR+) stromal cells, located in the vicinity of the high endothelial venules (HEVs) and lymphatics. These LepR+ stromal cells expressed markers for fibroblastic reticular cells (FRCs), but they lacked markers for follicular dendritic cells (FDCs) and marginal reticular cells (MRCs). Leptin signaling deficiency led to heightened inflammatory responses within the LNs of db/db mice, leakiness of HEVs, and lymphatic fragmentation. Leptin signaling through the JAK/STAT pathway supported LN stromal cell survival and promoted the anti-inflammatory properties of these cells. Conditional knockout of the LepR+ stromal cells in LNs resulted in HEV and extracellular matrix (ECM) abnormalities. Treatment of ob/ob mice with an agonist leptin fusion protein restored the microarchitecture of LNs, reduced intra-LN inflammatory responses, and corrected metabolic abnormalities. Future studies are needed to study the importance of LN stomal cell dysfunction to the pathogenesis of inflammatory responses in type 2 diabetes (T2D) in humans.


Blood ◽  
1963 ◽  
Vol 22 (6) ◽  
pp. 674-689 ◽  
Author(s):  
WILLIAM O. RIEKE ◽  
RUTH W. CAFFREY ◽  
N. B. EVERETT

Abstract Single and multiple injections of tritiated thymidine were combined with radioautography to study the rates of proliferation and interrelationships of the various cell lines in the mesenteric lymph node of the rat. The appearance and labeling patterns of the different cells are described from studies of both smears and tissue sections. Reticular cells exhibit wide variations in labeling intensity, phagocytize labeled lymphocytes, and become labeled in high percentages only when TTH is administered over a period of many days. Other slowly proliferating cell types include small lymphocytes, fat cells, endothelial cells and mast cells. Rapidly proliferating cell lines include plasmablasts, hemohistioblasts, proplasmacytes and large lymphocytes. The generation time of plasmablasts and hemohistioblasts was determined to be approximately 9 and 12 hours respectively. Mature plasma cells constitute a non-dividing population which is renewed in lymph node in not more than 5 days. Evidence is presented that the most primitive cells in the lymphocyte and plasma cell lines are the hemohistioblasts and plasmablasts respectively. Reticular cells most probably are not stem cells. No evidence is found to support previous reports that plasma cells derive from lymphocytes.


2004 ◽  
Vol 200 (6) ◽  
pp. 783-795 ◽  
Author(s):  
Tomoya Katakai ◽  
Takahiro Hara ◽  
Manabu Sugai ◽  
Hiroyuki Gonda ◽  
Akira Shimizu

The sophisticated microarchitecture of the lymph node, which is largely supported by a reticular network of fibroblastic reticular cells (FRCs) and extracellular matrix, is essential for immune function. How FRCs form the elaborate network and remodel it in response to lymphocyte activation is not understood. In this work, we established ERTR7+gp38+VCAM-1+ FRC lines and examined the production of the ER-TR7 antigen. Multiple chemokines produced by FRCs induced T cell and dendritic cell chemotaxis and adhesion to the FRC surface. FRCs can secrete the ER-TR7 antigen as an extracellular matrix component to make a reticular meshwork in response to contact with lymphocytes. The formation of the meshwork is induced by stimulation with tumor necrosis factor-α or lymphotoxin-α in combination with agonistic antibody to lymphotoxin-β receptor in a nuclear factor-κB (RelA)–dependent manner. These findings suggest that signals from lymphocytes induce FRCs to form the network that supports the movement and interactions of immune effectors within the lymph node.


Blood ◽  
2012 ◽  
Vol 120 (24) ◽  
pp. 4675-4683 ◽  
Author(s):  
Lucas Onder ◽  
Priyanka Narang ◽  
Elke Scandella ◽  
Qian Chai ◽  
Maria Iolyeva ◽  
...  

AbstractNonhematopoietic stromal cells of secondary lymphoid organs form important scaffold and fluid transport structures, such as lymph node (LN) trabeculae, lymph vessels, and conduits. Furthermore, through the production of chemokines and cytokines, these cells generate a particular microenvironment that determines lymphocyte positioning and supports lymphocyte homeostasis. IL-7 is an important stromal cell-derived cytokine that has been considered to be derived mainly from T-cell zone fibroblastic reticular cells. We show here that lymphatic endothelial cells (LECs) are a prominent source of IL-7 both in human and murine LNs. Using bacterial artificial chromosome transgenic IL-7–Cre mice, we found that fibroblastic reticular cells and LECs strongly up-regulated IL-7 expression during LN remodeling after viral infection and LN reconstruction after avascular transplantation. Furthermore, IL-7–producing stromal cells contributed to de novo formation of LyveI-positive lymphatic structures connecting reconstructed LNs with the surrounding tissue. Importantly, diphtheria toxin–mediated depletion of IL-7–producing stromal cells completely abolished LN reconstruction. Taken together, this study identifies LN LECs as a major source of IL-7 and shows that IL-7–producing stromal cells are critical for reconstruction and remodeling of the distinct LN microenvironment.


2007 ◽  
Vol 22 (10) ◽  
pp. 2733-2742 ◽  
Author(s):  
Y.-Y. He ◽  
M.-R. Du ◽  
P.-F. Guo ◽  
X.-J. He ◽  
W.-h. Zhou ◽  
...  

2019 ◽  
Vol 97 (Supplement_3) ◽  
pp. 209-210
Author(s):  
Kirill Plemyashov ◽  
Suleyman Suleymanov ◽  
Konstantin Lobodin ◽  
Olga Pavlenko

Abstract In this regard, the cytological picture of the lymphoid tissue in calves’ inguinal lymph node with spontaneous leptospirosis was studied. The study material was taken from the inguinal lymph node of the 11 calves who died of leptospirosis during the enzootic period in Azerbaijan. The study material samples were fixed in 10% neutral formalin solution, followed by pouring in paraffin, coloring azur sections with 2-eosin and counting 13 cell types (lymphoblast, prolymphocyte, lymphocyte, free reticular cell, process sinus reticular cell, endothelium, fibroblast, histiocyte, macrophage, polyblast, plasmablast, protoplasmocyte, plasmacyte) using MOV-15. It was established that the number of lymphoblasts in the inguinal lymph node with subacute leptospirosis decreased 2.2 times, the number of prolymphocytes decreased 1.4 times, the number of lymphocytes decreased 4.4 times. The number of free reticular cells from the cells of the reticuloendothelium decreased 3.7 times. However, the number of grown sinus reticular cells and the endothelium of the sinuses fluctuated within the normal range. The number of fibroblasts increased 1.7 times, histiocytes - 6.6 times, macrophages - 11.8 times, and polyblasts - 11 times (Table 1). At the same time, there was a sharp increase in the number of cells in the plasma row. Of those, the number of plasmablasts increased 8.5 times, protoplasmocytes - 30.4 times, plasma cells - 17 times. Overall, the cytological picture in the inguinal lymph node during spontaneous leptospirosis in calves was characterized by an increase in the number of plasma cells, fibroblasts, histiocytes, macrophages, polyblasts and a decrease in the number of lymphoblasts, prolymphocytes, lymphocytes and free reticular cells.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 447-447
Author(s):  
Yoshiaki Abe ◽  
Mamiko Sakata-Yanagimoto ◽  
Manabu Fujisawa ◽  
Hiroaki Miyoshi ◽  
Yasuhito Suehara ◽  
...  

Abstract Background: Activities of nonhematopoietic cells (NHCs) reportedly underlie lymphomagenesis. In follicular lymphoma (FL), mesenchymal stromal cells (SCs) including follicular dendritic cells (FDCs) have been shown to facilitate FL expansion. However, comprehensive understanding of lymphoma NHC activities have been hampered by indefinite NHC heterogeneity even in normal human lymph node (LN). Indeed, human LN blood endothelial cells (BECs) and non-endothelial stromal cells (NESCs) have not been analyzed at single-cell resolution. Here, we aimed to construct a single-cell atlas of NHCs in human LN applicable to lymphoma researches. We also sought to reveal the landscape of stromal remodeling in lymphomas, particularly in FL, to advance understanding of stromal contributions in lymphomagenesis. Methods: We prospectively performed single-cell RNA sequencing of NHCs (>100,000 cells) extracted from 27 human samples including metastasis-free LN (MFLN; n=9), nodal FL (n=10), peripheral T-cell lymphoma (PTCL; n=5), and diffuse large B-cell lymphoma transformed from FL (tDLBCL; n=3). Data from MFLN samples were used for the construction of NHC atlas. Immunofluorescence (IF) staining was performed to investigate the existence and topological localizations of each NHC subcluster in the LN. Using the NHC atlas, we performed comprehensive comparative analysis with FL NHCs by differentially-expressed gene (DEG) and intercellular ligand-receptor analyses. We also investigated the prognostic impact of putative stroma-derived biomarkers using deposited microarray data of FL patients. Finally, we examined the applicability of the atlas to NHCs from other lymphoma subtypes by analyzing PTCL and tDLBCL NHCs. Data analysis was performed through multiple pipelines including Seurat, Monocle3, and CellphoneDB. Results: Graph-based clustering analysis revealed that the transcriptional features of NHC subpopulations in MFLN are detectable in FL NHCs. Unsupervised sub-clustering analysis of BECs, lymphatic endothelial cells (LECs), and NESCs revealed 10, 8, and 12 subclusters, respectively, including some lacking mouse counterpart. IF staining successfully identified each NHC subcluster and its localization in the LN. In FL NHCs, the proportion of arterial BEC subclusters markedly increased relative to MFLN, while the proportion of LECs decreased. In FL NESCs, the proportion of marginal reticular cells (MRCs) as well as FDCs greatly increased. DEG analysis revealed that the greatest changes in gene expression occurs in NESC subclusters, particularly in MRCs, T-zone reticular cells (TRCs), pericytes, and FDCs. Notably, in some NESC subclusters, we observed marked upregulation of genes relevant to solid cancers but previously not described in lymphomas (e.g. POSTN, EGFL6, and FAP). Combined interactome and DEG analysis revealed 60 FL-specific interactions between NHC subclusters and malignant B cells. For example, interactions mediated through stroma-derived CD70 were enhanced at medullary SC subclusters and SCs at LN capsule adventitia. Additionally, the CCR7-CCL19 interaction and interactions via B-cell activating factor (BAFF) were unexpectedly upregulated at non-TRC SC and medullary SC subclusters, respectively. Also, the CXCL13-CXCR5 axis was highly activated in MRCs, collectively indicating that non-FDC SCs vigorously participate in FL cell expansion and/or infiltration into extra-follicular lesions. Some intercellular interactions were functionally validated by in vitro binding assays. Based on this dataset, we identified putative stroma-derived biomarkers linked to unfavorable prognosis in FL patients including TDO2, encoding immune-modulators, and LY6H and LOX, tip cell markers. We finally confirmed that NHC subclusters identified in our atlas were also detectable in NHCs of more aggressive lymphoma subtypes including PTCL and tDLBCL. Notably, we found that extra-follicular SCs had further differentiated into follicular SCs in tDLBCL, likely representing a terminal form of stromal remodeling in FL. Conclusion: We constructed a comprehensive single-cell atlas of NHCs in human LN highly applicable to lymphoma NHC researches and revealed a total of 30 NHC subclusters. Our study largely updates NHC taxonomy in LNs and provides a rich resource and deeper insights into lymphoma biology, a contribution that should advance lymphoma management and therapy. Figure 1 Figure 1. Disclosures Usuki: Otsuka Pharmaceutical Co., Ltd.: Research Funding, Speakers Bureau; Novartis Pharma K.K.: Research Funding, Speakers Bureau; Ono Pharmaceutical Co., Ltd.: Research Funding, Speakers Bureau; Janssen Pharmaceutical K.K.: Research Funding; Celgene K.K.: Research Funding, Speakers Bureau; Takeda Pharmaceutical Co., Ltd.: Research Funding, Speakers Bureau; Nippon-Boehringer-Ingelheim Co., Ltd.: Research Funding; Mundipharma K.K.: Research Funding; Amgen-Astellas Biopharma K.K.: Research Funding; Nippon-Shinyaku Co., Ltd.: Research Funding, Speakers Bureau; Kyowa-Kirin Co., Ltd.: Research Funding, Speakers Bureau; Pfizer Japan Inc.: Research Funding, Speakers Bureau; Alexion Pharmaceuticals, Inc.: Research Funding, Speakers Bureau; Eisai Co., Ltd.: Speakers Bureau; MSD K.K.: Research Funding, Speakers Bureau; PharmaEssentia Japan KK: Research Funding, Speakers Bureau; Yakult Honsha Co., Ltd.: Research Funding, Speakers Bureau; Daiichi Sankyo Co., Ltd.: Research Funding, Speakers Bureau; Sumitomo-Dainippon Pharma Co., Ltd.: Research Funding; SymBio Pharmaceuticals Ltd.: Research Funding, Speakers Bureau; Gilead Sciences, Inc.: Research Funding; Bristol-Myers-Squibb K.K.: Research Funding, Speakers Bureau; Apellis Pharmaceuticals, Inc.: Research Funding; AbbVie GK: Research Funding, Speakers Bureau; Astellas Pharma Inc.: Research Funding, Speakers Bureau; Incyte Biosciences Japan G.K.: Research Funding; Chugai Pharmaceutical Co., Ltd.: Research Funding, Speakers Bureau; Sanofi K.K.: Speakers Bureau; Amgen K.K.: Research Funding.


2011 ◽  
Vol 2 ◽  
Author(s):  
Anne L. Fletcher ◽  
Deepali Malhotra ◽  
Sophie E. Acton ◽  
Veronika Lukacs-Kornek ◽  
Angelique Bellemare-Pelletier ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document