Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Ther Oncolytics ; 24: 299-318, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35118189

ABSTRACT

This study determined the influence of intravenous (i.v.) oncolytic vaccinia virus mpJX-594 (mpJX) on antitumor activity of anti-programmed death receptor-1 antibody (aPD1) in functional and metastatic pancreatic neuroendocrine tumors (PanNETs). One i.v. dose of mpJX, engineered for mice with the same plasmid design as clinical virus Pexa-Vec, was administered alone or with repeated dosing of aPD1 (mpJX+aPD1) to two contrasting genetic models of PanNET: one developing benign insulin-secreting tumors (RIP1-Tag2;C57BL/6J mice) and the other developing liver metastases (RIP1-Tag2;AB6F1 mice). Experiments revealed that aPD1 had synergistic actions with mpJX on CD8+ T cell and natural killer (NK) cell influx, apoptosis, and suppression of proliferation in PanNETs. After mpJX+aPD1, the 53-fold increase in apoptosis (5 days) and 85% reduction in proliferation (20 days) exceeded the sum of mpJX and aPD1 given separately. mpJX+aPD1 also stabilized blood insulin and glucose in mice with functional PanNETs, regressed liver metastases in mice with aggressive PanNETs, and prolonged survival of both. The findings revealed that mpJX+aPD1 converted "cold" PanNETs into immunogenic tumors with widespread cytotoxic T cell influx, tumor cell killing, and suppression of proliferation. Reduction of tumor insulin secretion from functional PanNETs prolonged survival, and anti-metastatic actions on aggressive PanNETs reduced the metastatic burden to less than before treatment. The findings support the efficacy of the vaccinia virus with aPD1 for functional and metastatic PanNETs.

2.
Nat Immunol ; 22(5): 571-585, 2021 05.
Article in English | MEDLINE | ID: mdl-33903764

ABSTRACT

Fibroblastic reticular cells (FRCs) are specialized stromal cells that define tissue architecture and regulate lymphocyte compartmentalization, homeostasis, and innate and adaptive immunity in secondary lymphoid organs (SLOs). In the present study, we used single-cell RNA sequencing (scRNA-seq) of human and mouse lymph nodes (LNs) to identify a subset of T cell-zone FRCs defined by the expression of Gremlin1 (Grem1) in both species. Grem1-CreERT2 knock-in mice enabled localization, multi-omics characterization and genetic depletion of Grem1+ FRCs. Grem1+ FRCs primarily localize at T-B cell junctions of SLOs, neighboring pre-dendritic cells and conventional dendritic cells (cDCs). As such, their depletion resulted in preferential loss and decreased homeostatic proliferation and survival of resident cDCs and compromised T cell immunity. Trajectory analysis of human LN scRNA-seq data revealed expression similarities to murine FRCs, with GREM1+ cells marking the endpoint of both trajectories. These findings illuminate a new Grem1+ fibroblastic niche in LNs that functions to maintain the homeostasis of lymphoid tissue-resident cDCs.


Subject(s)
Dendritic Cells, Follicular/immunology , Fibroblasts/immunology , Lymph Nodes/immunology , Stromal Cells/immunology , Aged , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Proliferation/genetics , Cell Survival/genetics , Cell Survival/immunology , Dendritic Cells, Follicular/metabolism , Female , Fibroblasts/metabolism , Gene Expression Regulation/immunology , Gene Knock-In Techniques , Humans , Immunity, Cellular/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lymph Nodes/cytology , Male , Mice , Mice, Transgenic , RNA-Seq , Single-Cell Analysis , Stromal Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
Nat Immunol ; 20(9): 1174-1185, 2019 09.
Article in English | MEDLINE | ID: mdl-31406377

ABSTRACT

Classical type 1 dendritic cells (cDC1s) are required for antiviral and antitumor immunity, which necessitates an understanding of their development. Development of the cDC1 progenitor requires an E-protein-dependent enhancer located 41 kilobases downstream of the transcription start site of the transcription factor Irf8 (+41-kb Irf8 enhancer), but its maturation instead requires the Batf3-dependent +32-kb Irf8 enhancer. To understand this switch, we performed single-cell RNA sequencing of the common dendritic cell progenitor (CDP) and identified a cluster of cells that expressed transcription factors that influence cDC1 development, such as Nfil3, Id2 and Zeb2. Genetic epistasis among these factors revealed that Nfil3 expression is required for the transition from Zeb2hi and Id2lo CDPs to Zeb2lo and Id2hi CDPs, which represent the earliest committed cDC1 progenitors. This genetic circuit blocks E-protein activity to exclude plasmacytoid dendritic cell potential and explains the switch in Irf8 enhancer usage during cDC1 development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Dendritic Cells/cytology , Enhancer Elements, Genetic/genetics , Inhibitor of Differentiation Protein 2/metabolism , Interferon Regulatory Factors/metabolism , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/metabolism , Stem Cells/cytology
4.
Cancer Res ; 78(4): 922-937, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29259007

ABSTRACT

Oncolytic viruses pose many questions in their use in cancer therapy. In this study, we assessed the potential of mpJX-594 (mouse-prototype JX-594), a replication-competent vaccinia virus administered by intravenous injection, to target the tumor vasculature, produce immune activation and tumor cell killing more widespread than the infection, and suppress invasion and metastasis. These actions were examined in RIP-Tag2 transgenic mice with pancreatic neuroendocrine tumors that developed spontaneously and progressed as in humans. mpJX-594 initially infected tumor vascular endothelial cells, leading to vascular pruning and prolonged leakage in tumors but not in normal organs; parallel effects were observed in U87 gliomas. Viral infection spread to tumor cells, where tumor cell killing was much more widespread than the infection. Widespread tumor cell killing at 5 days was prevented by depletion of CD8+ T lymphocytes and did not require GM-CSF, as mpJX-594 variants that expressed human, mouse, or no GM-CSF produced equivalent amounts of killing. The antivascular, antitumor, and antimetastatic effects of mpJX-594 were amplified by concurrent or sequential administration of sunitinib, a multitargeted receptor tyrosine kinase inhibitor. These effects were not mimicked by selective inhibition of VEGFR2 despite equivalent vascular pruning, but were accompanied by suppression of regulatory T cells and greater influx of activated CD8+ T cells. Together, our results showed that mpJX-594 targets tumor blood vessels, spreads secondarily to tumor cells, and produces widespread CD8+ T-cell-dependent tumor cell killing in primary tumors and metastases, and that these effects can be amplified by coadministration of sunitinib.Significance: These findings reveal multiple unrecognized features of the antitumor properties of oncolytic vaccinia viruses, all of which can be amplified by the multitargeted kinase inhibitor sunitinib. Cancer Res; 78(4); 922-37. ©2017 AACR.


Subject(s)
Antineoplastic Agents/therapeutic use , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Sunitinib/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Humans , Mice , Mice, Transgenic , Sunitinib/pharmacology , Vaccinia virus/immunology
5.
J Am Soc Nephrol ; 29(1): 138-154, 2018 01.
Article in English | MEDLINE | ID: mdl-29217759

ABSTRACT

Dendritic cells (DCs) are thought to form a dendritic network across barrier surfaces and throughout organs, including the kidney, to perform an important sentinel function. However, previous studies of DC function used markers, such as CD11c or CX3CR1, that are not unique to DCs. Here, we evaluated the role of DCs in renal inflammation using a CD11c reporter mouse line and two mouse lines with DC-specific reporters, Zbtb46-GFP and Snx22-GFP. Multiphoton microscopy of kidney sections confirmed that most of the dendritically shaped CD11c+ cells forming a network throughout the renal interstitium expressed macrophage-specific markers. In contrast, DCs marked by Zbtb46-GFP or Snx22-GFP were less abundant, concentrated around blood vessels, and round in shape. We confirmed this pattern of localization using imaging mass cytometry. Motility measurements showed that resident macrophages were sessile, whereas DCs were motile before and after inflammation. Although uninflamed glomeruli rarely contained DCs, injury with nephrotoxic antibodies resulted in accumulation of ZBTB46 + cells in the periglomerular region. ZBTB46 identifies all classic DCs, which can be categorized into two functional subsets that express either CD103 or CD11b. Depletion of ZBTB46 + cells attenuated the antibody-induced kidney injury, whereas deficiency of the CD103+ subset accelerated injury through a mechanism that involved increased neutrophil infiltration. RNA sequencing 7 days after nephrotoxic antibody injection showed that CD11b+ DCs expressed the neutrophil-attracting cytokine CXCL2, whereas CD103+ DCs expressed high levels of several anti-inflammatory genes. These results provide new insights into the distinct functions of the two major DC subsets in glomerular inflammation.


Subject(s)
Dendritic Cells/physiology , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Animals , Antigens, CD/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , CD11 Antigens/genetics , CD11b Antigen/genetics , Cell Movement , Chemokine CXCL2/genetics , Dendritic Cells/metabolism , Dendritic Cells/pathology , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/metabolism , Integrin alpha Chains/metabolism , Macrophages , Male , Mice , Mice, Knockout , Neutrophils/pathology , Neutrophils/physiology , Repressor Proteins/genetics , Sequence Analysis, RNA , Sorting Nexins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
6.
Am J Pathol ; 187(9): 1984-1997, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28683257

ABSTRACT

Chylous pleural effusion (chylothorax) frequently accompanies lymphatic vessel malformations and other conditions with lymphatic defects. Although retrograde flow of chyle from the thoracic duct is considered a potential mechanism underlying chylothorax in patients and mouse models, the path chyle takes to reach the thoracic cavity is unclear. Herein, we use a novel transgenic mouse model, where doxycycline-induced overexpression of vascular endothelial growth factor (VEGF)-C was driven by the adipocyte-specific promoter adiponectin (ADN), to determine how chylothorax forms. Surprisingly, 100% of adult ADN-VEGF-C mice developed chylothorax within 7 days. Rapid, consistent appearance of chylothorax enabled us to examine the step-by-step development in otherwise normal adult mice. Dynamic imaging with a fluorescent tracer revealed that lymph in the thoracic duct of these mice could enter the thoracic cavity by retrograde flow into enlarged paravertebral lymphatics and subpleural lymphatic plexuses that had incompetent lymphatic valves. Pleural mesothelium overlying the lymphatic plexuses underwent exfoliation that increased during doxycycline exposure. Together, the findings indicate that chylothorax in ADN-VEGF-C mice results from retrograde flow of chyle from the thoracic duct into lymphatic tributaries with defective valves. Chyle extravasates from these plexuses and enters the thoracic cavity through exfoliated regions of the pleural mesothelium.


Subject(s)
Chylothorax/genetics , Lymphatic System/abnormalities , Vascular Endothelial Growth Factor C/genetics , Animals , Chylothorax/pathology , Lymphatic Vessels/abnormalities , Mice , Mice, Transgenic
7.
J Clin Invest ; 126(9): 3511-25, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27548529

ABSTRACT

Angiopoietin-2 (ANG2) regulates blood vessel remodeling in many pathological conditions through differential effects on Tie2 signaling. While ANG2 competes with ANG1 to inhibit Tie2, it can paradoxically also promote Tie2 phosphorylation (p-Tie2). A related paradox is that both inactivation and overactivation of Tie2 can result in vascular remodeling. Here, we reconciled these opposing actions of ANG2 by manipulating conditions that govern its actions in the vasculature. ANG2 drove vascular remodeling during Mycoplasma pulmonis infection by acting as a Tie2 antagonist, which led to p-Tie2 suppression, forkhead box O1 (FOXO1) activation, increased ANG2 expression, and vessel leakiness. These changes were exaggerated by anti-Tie2 antibody, inhibition of PI3K signaling, or ANG2 overexpression and were reduced by anti-ANG2 antibody or exogenous ANG1. In contrast, under pathogen-free conditions, ANG2 drove vascular remodeling by acting as an agonist, promoting high p-Tie2, low FOXO1 activation, and no leakage. Tie1 activation was strong under pathogen-free conditions, but infection or TNF-α led to Tie1 inactivation by ectodomain cleavage and promoted the Tie2 antagonist action of ANG2. Together, these data indicate that ANG2 activation of Tie2 supports stable enlargement of normal nonleaky vessels, but reduction of Tie1 in inflammation leads to ANG2 antagonism of Tie2 and initiates a positive feedback loop wherein FOXO1-driven ANG2 expression promotes vascular remodeling and leakage.


Subject(s)
Angiopoietin-2/metabolism , Forkhead Box Protein O1/antagonists & inhibitors , Receptor, TIE-2/metabolism , Animals , Antibodies, Monoclonal/chemistry , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Female , Humans , Inflammation , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Mycoplasma pulmonis , Phosphatidylinositol 3-Kinases/metabolism , Protein Domains , Vascular Endothelial Growth Factor A/metabolism , Vascular Remodeling
8.
Cell Rep ; 14(7): 1723-1734, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26876174

ABSTRACT

To induce adaptive immunity, dendritic cells (DCs) migrate through afferent lymphatic vessels (LVs) to draining lymph nodes (dLNs). This process occurs in several consecutive steps. Upon entry into lymphatic capillaries, DCs first actively crawl into downstream collecting vessels. From there, they are next passively and rapidly transported to the dLN by lymph flow. Here, we describe a role for the chemokine CCL21 in intralymphatic DC crawling. Performing time-lapse imaging in murine skin, we found that blockade of CCL21-but not the absence of lymph flow-completely abolished DC migration from capillaries toward collecting vessels and reduced the ability of intralymphatic DCs to emigrate from skin. Moreover, we found that in vitro low laminar flow established a CCL21 gradient along lymphatic endothelial monolayers, thereby inducing downstream-directed DC migration. These findings reveal a role for intralymphatic CCL21 in promoting DC trafficking to dLNs, through the formation of a flow-induced gradient.


Subject(s)
Bone Marrow Cells/cytology , Chemokine CCL21/immunology , Dendritic Cells/cytology , Endothelium, Lymphatic/immunology , Lymph Nodes/immunology , Lymphatic Vessels/immunology , Animals , Bone Marrow Cells/immunology , Cell Movement , Chemokine CCL21/genetics , Dendritic Cells/immunology , Ear , Endothelium, Lymphatic/ultrastructure , Gene Expression , Lymph Nodes/ultrastructure , Lymphatic Vessels/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rheology , Skin/cytology , Skin/immunology , Time-Lapse Imaging
9.
Am J Pathol ; 184(6): 1877-89, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24726646

ABSTRACT

Vascular remodeling is a feature of sustained inflammation in which capillaries enlarge and acquire the phenotype of venules specialized for plasma leakage and leukocyte recruitment. We sought to determine whether neutrophils are required for vascular remodeling in the respiratory tract by using Mycoplasma pulmonis infection as a model of sustained inflammation in mice. The time course of vascular remodeling coincided with the influx of neutrophils during the first few days after infection and peaked at day 5. Depletion of neutrophils with antibody RB6-8C5 or 1A8 reduced neutrophil influx and vascular remodeling after infection by about 90%. Similarly, vascular remodeling after infection was suppressed in Cxcr2(-/-) mice, in which neutrophils adhered to the endothelium of venules but did not extravasate into the tissue. Expression of the venular adhesion molecule P-selectin increased in endothelial cells from day 1 to day 3 after infection, as did expression of the Cxcr2-receptor ligands Cxcl1 and Cxcl2. Tumor necrosis factor α (TNFα) expression increased more than sixfold in the trachea of wild-type and Cxcr2(-/-) mice, but intratracheal administration of TNFα did not induce vascular remodeling similar to that seen in infection. We conclude that neutrophil influx is required for remodeling of capillaries into venules in the airways of mice with Mycoplasma infection and that TNFα signaling is necessary but not sufficient for vascular remodeling.


Subject(s)
Endothelium, Vascular/metabolism , Mycoplasma Infections/metabolism , Mycoplasma pulmonis , Neutrophils/metabolism , Respiratory System/metabolism , Vascular Remodeling , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Endothelium, Vascular/pathology , Female , Mice , Mice, Knockout , Mycoplasma Infections/genetics , Mycoplasma Infections/pathology , Neutrophils/pathology , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Respiratory System/pathology
10.
Lymphat Res Biol ; 11(3): 172-82, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24044757

ABSTRACT

Afferent lymphatic vessels fulfill essential immune functions by transporting leukocytes and lymph-borne antigen to draining lymph nodes (dLNs). An important cell type migrating through lymphatic vessels are dendritic cells (DCs). DCs reside in peripheral tissues like the skin, where they take up antigen and transport it via the lymphatic vascular network to dLNs for subsequent presentation to T cells. As such, DCs play a key role in the induction of adaptive immune responses during infection and vaccination, but also for the maintenance of tolerance. Although the migratory pattern of DCs has been known for long time, interactions between DCs and lymphatic vessels are only now starting to be unraveled at the cellular level. In particular, new tools for visualizing lymphatic vessels in combination with time-lapse microscopy have recently generated valuable insights into the process of DC migration to dLNs. In this review we summarize and discuss current approaches for visualizing DCs and lymphatic vessels in tissues for imaging applications. Furthermore, we review the current state of knowledge about DC migration towards, into and within lymphatic vessels, particularly focusing on the cellular interactions that take place between DCs and the lymphatic endothelium.


Subject(s)
Dendritic Cells/metabolism , Endothelium, Lymphatic/metabolism , Lymph Nodes/metabolism , Lymphatic Vessels/metabolism , Animals , Cell Communication , Cell Movement , Dendritic Cells/cytology , Endothelium, Lymphatic/cytology , Humans , Microscopy, Confocal , Time-Lapse Imaging
11.
Blood ; 120(11): 2249-58, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22855606

ABSTRACT

Dendritic cell (DC) migration via lymphatic vessels to draining lymph nodes (dLNs) is crucial for the initiation of adaptive immunity. We imaged this process by intravital microscopy (IVM) in the ear skin of transgenic mice bearing red-fluorescent vasculature and yellow-fluorescent DCs. DCs within lymphatic capillaries were rarely transported by flow, but actively migrated within lymphatics and were significantly faster than in the interstitium. Pharmacologic blockade of the Rho-associated protein kinase (ROCK), which mediates nuclear contraction and de-adhesion from integrin ligands, significantly reduced DC migration from skin to dLNs in steady-state. IVM revealed that ROCK blockade strongly reduced the velocity of interstitial DC migration, but only marginally affected intralymphatic DC migration. By contrast, during tissue inflammation, ROCK blockade profoundly decreased both interstitial and intralymphatic DC migration. Inhibition of intralymphatic migration was paralleled by a strong up-regulation of ICAM-1 in lymphatic endothelium, suggesting that during inflammation ROCK mediates de-adhesion of DC-expressed integrins from lymphatic-expressed ICAM-1. Flow chamber assays confirmed an involvement of lymphatic-expressed ICAM-1 and DC-expressed ROCK in DC crawling on lymphatic endothelium. Overall, our findings further define the role of ROCK in DC migration to dLNs and reveal a differential requirement for ROCK in intralymphatic DC crawling during steady-state and inflammation.


Subject(s)
Cell Movement , Dendritic Cells/metabolism , Dermatitis, Contact/metabolism , Endothelium, Lymphatic/immunology , rho-Associated Kinases/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Crosses, Genetic , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Dermatitis, Contact/drug therapy , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Endothelium, Lymphatic/drug effects , Endothelium, Lymphatic/metabolism , Endothelium, Lymphatic/pathology , Intercellular Adhesion Molecule-1/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Microscopy, Video , Protein Kinase Inhibitors/pharmacology , Radiation Chimera , Recombinant Fusion Proteins/metabolism , Skin/drug effects , Skin/immunology , Skin/metabolism , Skin/pathology , Up-Regulation/drug effects , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
12.
Blood ; 118(1): 205-15, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-21596851

ABSTRACT

Chemokines and adhesion molecules up-regulated in lymphatic endothelial cells (LECs) during tissue inflammation are thought to enhance dendritic cell (DC) migration to draining lymph nodes, but the in vivo control of this process is not well understood. We performed a transcriptional profiling analysis of LECs isolated from murine skin and found that inflammation induced by a contact hypersensitivity (CHS) response up-regulated the adhesion molecules ICAM-1 and VCAM-1 and inflammatory chemokines. Importantly, the lymphatic markers Prox-1, VEGFR3, and LYVE-1 were significantly down-regulated during CHS. By contrast, skin inflammation induced by complete Freund adjuvant induced a different pattern of chemokine and lymphatic marker gene expression and almost no ICAM-1 up-regulation in LECs. Fluorescein isothiocyanate painting experiments revealed that DC migration to draining lymph nodes was more strongly increased in complete Freund adjuvant-induced than in CHS-induced inflammation. Surprisingly, DC migration did not correlate with the induction of CCL21 and ICAM-1 protein in LECs. Although the requirement for CCR7 signaling became further pronounced during inflammation, CCR7-independent signals had an additional, albeit moderate, impact on enhancing DC migration. Collectively, these findings indicate that DC migration in response to inflammation is stimulus-specific, mainly CCR7-dependent, and overall only moderately enhanced by LEC-induced genes other than CCL21.


Subject(s)
Cell Movement/immunology , Dendritic Cells/immunology , Dermatitis, Contact/immunology , Endothelial Cells/immunology , Lymph Nodes/immunology , Animals , Chemokine CCL21/genetics , Chemokine CCL21/immunology , Chemokine CCL21/metabolism , Dendritic Cells/cytology , Ear, External/immunology , Female , Gene Expression/immunology , Gene Expression Profiling , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Receptors, CCR7/metabolism , Up-Regulation/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
13.
Blood ; 115(23): 4725-33, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20185585

ABSTRACT

Adaptive immune responses are characterized by substantial restructuring of secondary lymphoid organs. The molecular and cellular factors responsible for virus-induced lymphoid remodeling are not well known to date. Here we applied optical projection tomography, a mesoscopic imaging technique, for a global analysis of the entire 3-dimensional structure of mouse peripheral lymph nodes (PLNs), focusing on B-cell areas and high endothelial venule (HEV) networks. Structural homeostasis of PLNs was characterized by a strict correlation between total PLN volume, B-cell volume, B-cell follicle number, and HEV length. After infection with lymphocytic choriomeningitis virus, we observed a substantial, lymphotoxin (LT) beta-receptor-dependent reorganization of the PLN microarchitecture, in which an initial B-cell influx was followed by 3-fold increases in PLN volume and HEV network length on day 8 after infection. Adoptive transfer experiments revealed that virus-induced PLN and HEV network remodeling required LTalpha(1)beta(2)-expressing B cells, whereas the inhibition of vascular endothelial growth factor-A signaling pathways had no significant effect on PLN expansion. In summary, lymphocytic choriomeningitis virus-induced PLN growth depends on a vascular endothelial growth factor-A-independent, LT- and B cell-dependent morphogenic pathway, as revealed by an in-depth mesoscopic analysis of the global PLN structure.


Subject(s)
Adaptive Immunity , B-Lymphocytes/immunology , Lymph Nodes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphotoxin alpha1, beta2 Heterotrimer/immunology , Animals , B-Lymphocytes/pathology , B-Lymphocytes/virology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Homeostasis/genetics , Homeostasis/immunology , Lymph Nodes/pathology , Lymph Nodes/virology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/metabolism , Lymphotoxin alpha1, beta2 Heterotrimer/biosynthesis , Lymphotoxin alpha1, beta2 Heterotrimer/genetics , Lymphotoxin beta Receptor/biosynthesis , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Mice , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
14.
Hybridoma (Larchmt) ; 28(4): 281-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19663701

ABSTRACT

The LST1 gene is located in the human MHC class III region and encodes transmembrane and soluble isoforms that have been suggested to play a role in the regulation of the immune response and are associated with inflammatory diseases such as rheumatoid arthritis. Here we describe the generation and characterization of the first monoclonal antibodies against LST1. Two hybridoma lines secreting monoclonal antibodies designated 7E2 and 8D12 were established. The 7E2 antibody detects recombinant and endogenous LST1 by Western blot analysis while 8D12 reacts with recombinant and endogenous LST1 in immunoprecipitation and flow cytometry procedures. The newly established antibodies were used to survey LST1 protein expression in human cell lines, which was found to be tightly regulated, allowing the expression of transmembrane isoforms but suppressing soluble isoforms.


Subject(s)
Antibodies, Monoclonal/chemistry , Membrane Proteins/chemistry , Membrane Proteins/immunology , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Flow Cytometry/methods , Humans , Hybridomas/immunology , Inflammation , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Protein Isoforms , Rats , Sequence Homology, Amino Acid , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...