Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Cell Mol Gastroenterol Hepatol ; 11(2): 573-595, 2021.
Article in English | MEDLINE | ID: mdl-32961356

ABSTRACT

BACKGROUND AND AIMS: As the incidence of nonalcoholic steatohepatitis (NASH) continues to rise, understanding how normal liver functions are affected during disease is required before developing novel therapeutics which could reduce morbidity and mortality. However, very little is understood about how the transport of proteins and cells from the liver by the lymphatic vasculature is affected by inflammatory mediators or during disease. METHODS: To answer these questions, we utilized a well-validated mouse model of NASH and exposure to highly oxidized low density lipoprotein (oxLDL). In addition to single cell sequencing, multiplexed immunofluorescence and metabolomic analysis of liver lymphatic endothelial cells (LEC)s we evaluated lymphatic permeability and transport both in vitro and in vivo. RESULTS: Confirming similarities between human and mouse liver lymphatic vasculature in NASH, we found that the lymphatic vasculature expands as disease progresses and results in the downregulation of genes important to lymphatic identity and function. We also demonstrate, in mice with NASH, that fluorescein isothiocyanate (FITC) dextran does not accumulate in the liver draining lymph node upon intrahepatic injection, a defect that was rescued with therapeutic administration of the lymphatic growth factor, recombinant vascular endothelial growth factor C (rVEGFC). Similarly, exposure to oxLDL reduced the amount of FITC-dextran in the portal draining lymph node and through an LEC monolayer. We provide evidence that the mechanism by which oxLDL impacts lymphatic permeability is via a reduction in Prox1 expression which decreases lymphatic specific gene expression, impedes LEC metabolism and reorganizes the highly permeable lymphatic cell-cell junctions which are a defining feature of lymphatic capillaries. CONCLUSIONS: We identify oxLDL as a major contributor to decreased lymphatic permeability in the liver, a change which is consistent with decreased protein homeostasis and increased inflammation during chronic liver disease.


Subject(s)
Lipoproteins, LDL/metabolism , Liver/pathology , Lymphatic Vessels/pathology , Non-alcoholic Fatty Liver Disease/immunology , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Homeodomain Proteins/metabolism , Humans , Intercellular Junctions/pathology , Liver/immunology , Lymphatic Vessels/cytology , Lymphatic Vessels/immunology , Male , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Permeability , Proteostasis/genetics , Proteostasis/immunology , RNA-Seq , Single-Cell Analysis , Tumor Suppressor Proteins/metabolism
3.
Front Immunol ; 10: 1036, 2019.
Article in English | MEDLINE | ID: mdl-31156626

ABSTRACT

Liver lymphatic vessels support liver function by draining interstitial fluid, cholesterol, fat, and immune cells for surveillance in the liver draining lymph node. Chronic liver disease is associated with increased inflammation and immune cell infiltrate. However, it is currently unknown if or how lymphatic vessels respond to increased inflammation and immune cell infiltrate in the liver during chronic disease. Here we demonstrate that lymphatic vessel abundance increases in patients with chronic liver disease and is associated with areas of fibrosis and immune cell infiltration. Using single-cell mRNA sequencing and multi-spectral immunofluorescence analysis we identified liver lymphatic endothelial cells and found that chronic liver disease results in lymphatic endothelial cells (LECs) that are in active cell cycle with increased expression of CCL21. Additionally, we found that LECs from patients with NASH adopt a transcriptional program associated with increased IL13 signaling. Moreover, we found that oxidized low density lipoprotein, associated with NASH pathogenesis, induced the transcription and protein production of IL13 in LECs both in vitro and in a mouse model. Finally, we show that oxidized low density lipoprotein reduced the transcription of PROX1 and decreased lymphatic stability. Together these data indicate that LECs are active participants in the liver, expanding in an attempt to maintain tissue homeostasis. However, when inflammatory signals, such as oxidized low density lipoprotein are increased, as in NASH, lymphatic function declines and liver homeostasis is impeded.


Subject(s)
Cell Differentiation , Endothelial Cells/metabolism , Hepatitis C, Chronic/metabolism , Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Adult , Aged , Aged, 80 and over , Animals , Chronic Disease , Female , Hepatitis C, Chronic/pathology , Humans , Lipoproteins, LDL/pharmacology , Lymphatic Vessels/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/pathology , Sequence Analysis, RNA , Single-Cell Analysis
4.
Front Immunol ; 10: 1313, 2019.
Article in English | MEDLINE | ID: mdl-31244852

ABSTRACT

Post-partum breast cancer patients, or breast cancer patients diagnosed within 10 years of last childbirth, are ~3-5 times more likely to develop metastasis in comparison to non-post-partum, or nulliparous, patients. Additionally, post-partum patients have increased tumor-associated lymphatic vessels and LN involvement, including when controlled for size of the primary tumor. In pre-clinical, immune-competent, mouse mammary tumor models of post-partum breast cancer (PPBC), tumor growth and lymphogenous tumor cell spread occur more rapidly in post-partum hosts. Here we report on PD-L1 expression by lymphatic endothelial cells and CD11b+ cells in the microenvironment of post-partum tumors, which is accompanied by an increase in PD-1 expression by T cells. Additionally, we observed increases in PD-L1 and PD-1 in whole mammary tissues during post-partum mammary gland involution; a known driver of post-partum tumor growth, invasion, and metastasis in pre-clinical models. Importantly, implantation of murine mammary tumor cells during post-partum mammary gland involution elicits a CD8+ T cell population that expresses both the co-inhibitory receptors PD-1 and Lag-3. However, upon anti-PD-1 treatment, during post-partum mammary gland involution, the involution-initiated promotional effects on tumor growth are reversed and the PD-1, Lag-3 double positive population disappears. Consequently, we observed an expansion of poly-functional CD8+ T cells that produced both IFNγ and TNFα. Finally, lymphatic vessel frequency decreased significantly following anti-PD-1 suggesting that anti-PD-1/PD-L1 targeted therapies may have efficacy in reducing tumor growth and dissemination in post-partum breast cancer patients.


Subject(s)
Breast Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , B7-H1 Antigen/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Female , Humans , Lymphatic Vessels/immunology , Lymphatic Vessels/pathology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Postpartum Period/immunology , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment/immunology
5.
J Vis Exp ; (143)2019 01 07.
Article in English | MEDLINE | ID: mdl-30663671

ABSTRACT

Within the liver, lymphatic vessels are found within the portal triad, and their described function is to remove interstitial fluid from the liver to the lymph nodes where cellular debris and antigens can be surveyed. We are very interested in understanding how the lymphatic vasculature might be involved in inflammation and immune cell function within the liver. However, very little has been published establishing digestion protocols for the isolation of lymphatic endothelial cells (LECs) from the liver or specific markers that can be used to evaluate liver LECs on a per cell basis. Therefore, we optimized a method for the digestion and staining of the liver in order to evaluate the LEC population in the liver. We are confident that the method outlined here will be useful for the identification and isolation of LECs from the liver and will strengthen our understanding of how LECs respond to the liver microenvironment.


Subject(s)
Biomarkers/metabolism , Endothelial Cells/pathology , Flow Cytometry/methods , Liver/pathology , Lymphatic Vessels/physiopathology , Animals , Humans , Mice
6.
J Immunol ; 201(6): 1735-1747, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30045970

ABSTRACT

Lymph node (LN) expansion during an immune response is a complex process that involves the relaxation of the fibroblastic network, germinal center formation, and lymphatic vessel growth. These processes require the stromal cell network of the LN to act deliberately to accommodate the influx of immune cells to the LN. The molecular drivers of these processes are not well understood. Therefore, we asked whether the immediate cytokines type 1 IFN produced during viral infection influence the lymphatic network of the LN in mice. We found that following an IFN-inducing stimulus such as viral infection or polyI:C, programmed cell death ligand 1 (PD-L1) expression is dynamically upregulated on lymphatic endothelial cells (LECs). We found that reception of type 1 IFN by LECs is important for the upregulation of PD-L1 of mouse and human LECs and the inhibition of LEC expansion in the LN. Expression of PD-L1 by LECs is also important for the regulation of LN expansion and contraction after an IFN-inducing stimulus. We demonstrate a direct role for both type 1 IFN and PD-L1 in inhibiting LEC division and in promoting LEC survival. Together, these data reveal a novel mechanism for the coordination of type 1 IFN and PD-L1 in manipulating LEC expansion and survival during an inflammatory immune response.


Subject(s)
B7-H1 Antigen/immunology , Cell Proliferation , Endothelial Cells/immunology , Endothelium, Lymphatic/immunology , Interferon Type I/immunology , Animals , B7-H1 Antigen/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Endothelial Cells/pathology , Endothelium, Lymphatic/pathology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interferon Type I/genetics , Mice , Mice, Knockout , Poly I-C/pharmacology
7.
Nat Commun ; 8(1): 2034, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29229919

ABSTRACT

Antigens derived from viral infection or vaccination can persist within a host for many weeks after resolution of the infection or vaccine responses. We previously identified lymphatic endothelial cells (LEC) as the repository for this antigen archival, yet LECs are unable to present their archived antigens to CD8+ T cells, and instead transfer their antigens to CD11c+ antigen-presenting cells (APC). Here we show that the exchange of archived antigens between LECs and APCs is mediated by migratory dendritic cells (DC). After vaccination, both migratory basic leucine zipper ATF-like transcription factor 3 (BatF3)-dependent and BatF3-independent DCs are responsible for antigen exchange and cross-presentation. However, exchange of archived viral antigens is mediated only by BatF3-dependent migratory DCs potentially acquiring apoptotic LECs. In conclusion, LEC-archived antigens are exchanged with migratory DCs, both directly and through LEC apoptosis, to cross-present archived antigens to circulating T cells.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Endothelial Cells/immunology , Lymph Nodes/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/immunology , Female , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics , Repressor Proteins/immunology , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL