Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
1.
Front Immunol ; 13: 1009306, 2022.
Article in English | MEDLINE | ID: mdl-36189308

ABSTRACT

Tumor-associated tertiary lymphoid structures are ectopic lymphoid aggregates that have considerable morphological, cellular, and molecular similarity to secondary lymphoid organs, particularly lymph nodes. Tumor vessels expressing peripheral node addressin (PNAd) are hallmark features of these structures. Previous work from our laboratory demonstrated that PNAd is displayed on intratumoral vasculature of murine tumors, and its expression is controlled by the engagement of lymphotoxin-α3, secreted by effector CD8 T cells, with tumor necrosis factor receptors (TNFR) on tumor endothelial cells (TEC). The goals of the present work were: 1) to identify differences in expression of genes encoding the scaffolding proteins and glycosyl transferases associated with PNAd biosynthesis in TEC and lymph node blood endothelial cells (LN BEC); and 2) to determine which of these PNAd associated components are regulated by TNFR signaling. We found that the same genes encoding scaffolding proteins and glycosyl transferases were upregulated in PNAd+ LN BEC and PNAd+ TEC relative to their PNAdneg counterparts. The lower level of PNAd expression on TEC vs LN BEC was associated with relatively lower expression of these genes, particularly the carbohydrate sulfotransferase Chst4. Loss of PNAd on TEC in the absence of TNFR signaling was associated with lack of upregulation of these same genes. A small subset of PNAd+ TEC remaining in the absence of TNFR signaling showed normal upregulation of a subset of these genes, but reduced upregulation of genes encoding the scaffolding proteins podocalyxin and nepmucin, and carbohydrate sulfotransferase Chst2. Lastly, we found that checkpoint immunotherapy augmented both the fraction of TEC expressing PNAd and their surface level of this ligand. This work points to strong similarities in the regulation of PNAd expression on TEC by TNFR signaling and on LN BEC by lymphotoxin-ß receptor signaling, and provides a platform for the development of novel strategies that manipulate PNAd expression on tumor vasculature as an element of cancer immunotherapy.


Subject(s)
Endothelial Cells , Neoplasms , Animals , Ligands , Lymphotoxin-alpha/physiology , Lymphotoxin-beta , Mice , Neoplasms/genetics , Receptors, Tumor Necrosis Factor , Sulfotransferases
2.
Cancer Res ; 79(7): 1383-1397, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30683654

ABSTRACT

The metabolic activity of fumarase (FH) participates in gene transcription linking to tumor cell growth. However, whether this effect is implicated in lung cancer remains unclear. Here, we show TGFß induces p38-mediated FH phosphorylation at Thr 90, which leads to a FH/CSL (also known as RBP-Jκ)/p53 complex formation and FH accumulation at p21 promoter under concomitant activation of Notch signaling; in turn, FH inhibits histone H3 Lys 36 demethylation and thereby promotes p21 transcription and cell growth arrest. In addition, FH is massively phosphorylated at the Ser 46 by PAK4 in non-small cell lung cancer (NSCLC) cells, and PAK4-phosphorylated FH binds to 14-3-3, resulting in cytosolic detention of FH and prohibition of FH/CSL/p53 complex formation. Physiologically, FH Ser 46 phosphorylation promotes tumorigenesis through its suppressive effect on FH Thr 90 phosphorylation-mediated cell growth arrest in NSCLC cells and correlates with poor prognosis in patients with lung cancer. Our findings uncover an uncharacterized mechanism underlying the local effect of FH on TGFß-induced gene transcription, on which the inhibitory effect from PAK4 promotes tumorigenesis in lung cancer. SIGNIFICANCE: Fumarase counteracts CSL via its metabolic activity to facilitate TGFß-induced cell growth arrest, an effect largely blocked by PAK4-mediated phosphorylation of fumarase.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/physiology , Fumarate Hydratase/metabolism , Lung Neoplasms/pathology , Lymphotoxin-alpha/physiology , p21-Activated Kinases/metabolism , 14-3-3 Proteins/metabolism , A549 Cells , Animals , Carcinogenesis , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/metabolism , F-Box Proteins/metabolism , Heterografts , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Lymphotoxin-alpha/antagonists & inhibitors , Male , Mice , Mice, Nude , Phosphorylation , Protein Binding
3.
Exp Biol Med (Maywood) ; 244(1): 1-12, 2019 01.
Article in English | MEDLINE | ID: mdl-30661394

ABSTRACT

IMPACT STATEMENT: The mechanism by which natural products such as resveratrol suppresses TNF-ß-promoted tumor cell proliferation, invasion, and colony formation is unknown. In this study, we explored for the first time the effect of resveratrol on the proinflammatory cytokine TNF-ß-, compared to TNF-α-stimulated proliferative and pro-inflammatory signaling in HCT116 cells. Our findings suggest that expression of TNF-ß and TNF-ß-receptor, like TNF-α, can lead to activation of inflammatory transcription factor (NF-κB) and NF-κB-regulated gene biomarkers, which are involved in the promotion of cancer proliferation, invasion, metastasis, and cell survival of tumor. Resveratrol can block TNF-ß/TNF-ß-receptor-induced activation of NF-κB, NF-κB-modulated gene products, and inhibition of caspase-3 cleavage. These results highlight the therapeutic effect of resveratrol-mediated anti-tumor activity by multitargeting cellular signaling pathways.


Subject(s)
Colorectal Neoplasms/pathology , Cytostatic Agents/therapeutic use , Lymphotoxin-alpha/physiology , Resveratrol/therapeutic use , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Down-Regulation , HCT116 Cells , Humans , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , NF-kappa B/metabolism
4.
PLoS One ; 12(11): e0186993, 2017.
Article in English | MEDLINE | ID: mdl-29095837

ABSTRACT

While Lymphotoxin α (TNF-ß), a product of lymphocytes, is known to play a pivotal role in inflammatory joint environment, resveratrol has been shown to possess anti-inflammatory and chondroprotective effects via activation of the histondeacetylase Sirt1. Whether TNF-ß induction of inflammatory pathways in primary human chondrocytes (PCH) can be modulated by resveratrol, was investigated. Monolayer and alginate cultures of PCH were treated with TNF-ß, anti-TNF-ß, nicotinamide (NAM), antisense oligonucleotides against Sirt1 (Sirt1-ASO) and/or resveratrol and co-cultured with T-lymphocytes. We found that resveratrol suppressed, similar to anti-TNF-ß, TNF-ß-induced increased adhesiveness in an inflammatory microenvironment of T-lymphocytes and PCH. In contrast, knockdown of Sirt1 by mRNA abolished the inhibitory effects of resveratrol on the TNF-ß-induced adhesiveness, suggesting the essential role of this enzyme for resveratrol-mediated anti-inflammatory signaling. Similar results were obtained in PCH stimulated with TNF-α. Sirt1-ASO, NAM or TNF-ß, similar to T-lymphocytes induced inflammatory microenvironment by down-regulation of cartilage-specific proteins, Sox9, Ki67 and enhanced NF-κB-regulated gene products involved in inflammatory and degradative processes in cartilage (MMP-9/-13, COX-2, caspase-3), NF-κB activation and its translocation to the nucleus. Moreover, resveratrol reversed the TNF-ß-, NAM-, T-lymphocytes-induced up-regulation of various NF-κB-regulated gene products. Down-regulation of Sirt1 by mRNA interference abrogated the effect of resveratrol on TNF-ß-induced effects. Ultrastructural and cell viability assay investigations revealed that resveratrol revoked TNF-ß-induced dose-dependent degradative/apoptotic morphological changes, cell viability and proliferation in PCH. Taken together, suppression of TNF-ß-induced inflammatory microenvironment in PCH by resveratrol/Sirt1 might be a novel therapeutic approach for targeting inflammation during rheumatoid arthritis.


Subject(s)
Cartilage, Articular/pathology , Chondrocytes/drug effects , Down-Regulation/drug effects , Inflammation/prevention & control , Lymphotoxin-alpha/physiology , Stilbenes/pharmacology , Tumor Necrosis Factor-alpha/physiology , Apoptosis/drug effects , Cells, Cultured , Chondrocytes/pathology , Gene Knockdown Techniques , Humans , Inflammation/physiopathology , NF-kappa B/metabolism , Resveratrol , Signal Transduction/drug effects , Sirtuin 1/genetics , Up-Regulation/drug effects
5.
Angiología ; 67(3): 206-215, mayo-jun. 2015. ilus
Article in Spanish | IBECS | ID: ibc-136721

ABSTRACT

La isquemia de larga duración es un proceso bastante común durante y tras la cirugía vascular, y la correcta recuperación de la función del órgano afectado depende de que se recupere el flujo sanguíneo del órgano afectado. La isquemia desencadena procesos como la inflamación, angiogénesis o vasculogénesis, que permitirán la reperfusión del órgano. Endoglina es un correceptor para los miembros de la superfamilia de citocinas de TGF-β. En nuestro laboratorio llevamos muchos años estudiando la función y los mecanismos de acción de endoglina, así como su implicación en regulación del tono vascular, angiogénesis, inflamación, fibrosis y en enfermedades como la preeclampsia y la hipertensión. Nuestro objetivo en este trabajo es revisar cómo se suceden los eventos que tienen lugar en la revascularización postisquémica, y mostrar las evidencias que colocan a endoglina como una molécula clave en estos eventos y como una posible diana terapéutica


Long-term ischemia is frequent during and after vascular surgery, and the recovery of affected organ function depends on the re-establishment of blood supply. Ischemia triggers processes such as inflammation, angiogenesis, or vasculogenesis that will lead to tissue remodeling and blood supply restoration. All these events are closely related and highly regulated to achieve complete recovery. Endoglin is a co-receptor for TGF-β cytokines superfamily that plays a central role in vascular physiology, angiogenesis, inflammation and post-ischemic blood supply recovery. Our laboratory has been involved for a long time in the study of endoglin function and its mechanisms of action and involvement in the regulation of vascular function, angiogenesis, inflammation, and fibrosis, as well as in diseases such as hypertension and preeclampsia. The aim here is to review how post-ischemic revascularization takes place, and to assess the role of endoglin in these events, and its importance as a possible therapeutic target


Subject(s)
Humans , Reperfusion/methods , Ischemia/surgery , Neovascularization, Physiologic/physiology , Lymphotoxin-alpha/physiology , Biomarkers/analysis , Inflammation/physiopathology , Cytokines/physiology
6.
J Immunol ; 193(3): 1194-203, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24951816

ABSTRACT

Spleen is a tissue with regenerative capacity, which allows autotransplantation of human spleen fragments to counteract the effects of splenectomy. We now reveal in a murine model that transplant of neonatal spleen capsule alone leads to the regeneration of full spleen tissue. This finding indicates that graft-derived spleen stromal cells, but not lymphocytes, are essential components of tissue neogenesis, a finding verified by transplant and regeneration of Rag1KO spleen capsules. We further demonstrate that lymphotoxin and lymphoid tissue inducer cells participate in two key elements of spleen neogenesis, bulk tissue regeneration and white pulp organization, identifying a lymphotoxin-dependent pathway for neonatal spleen regeneration that contrasts with previously defined lymphotoxin-independent embryonic spleen organogenesis.


Subject(s)
Lymphotoxin-alpha/physiology , Regeneration/immunology , Spleen/immunology , Spleen/transplantation , Animals , Animals, Newborn , Humans , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Animal , Signal Transduction/immunology , Spleen/cytology , Stromal Cells/cytology , Stromal Cells/immunology
8.
Nat Immunol ; 13(10): 947-53, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22922363

ABSTRACT

Microbiota are essential for weight gain in mouse models of diet-induced obesity (DIO), but the pathways that cause the microbiota to induce weight gain are unknown. We report that mice deficient in lymphotoxin, a key molecule in gut immunity, were resistant to DIO. Ltbr(-/-) mice had different microbial community composition compared to their heterozygous littermates, including an overgrowth of segmented filamentous bacteria (SFB). Furthermore, cecal transplantation conferred leanness to germ-free recipients. Housing Ltbr(-/-) mice with their obese siblings rescued weight gain in Ltbr(-/-) mice, demonstrating the communicability of the obese phenotype. Ltbr(-/-) mice lacked interleukin 23 (IL-23) and IL-22, which can regulate SFB. Mice deficient in these pathways also resisted DIO, demonstrating that intact mucosal immunity guides diet-induced changes to the microbiota to enable obesity.


Subject(s)
Immunity, Mucosal , Lymphotoxin beta Receptor/physiology , Lymphotoxin-alpha/physiology , Obesity , Animals , Bacteria/growth & development , Bacteria/immunology , Cecum/microbiology , Cecum/transplantation , Diet , Energy Metabolism , Germ-Free Life , Interleukin-23/deficiency , Interleukin-23/physiology , Interleukins/deficiency , Interleukins/physiology , Lymphotoxin beta Receptor/genetics , Lymphotoxin-alpha/deficiency , Lymphotoxin-alpha/genetics , Metagenome , Mice , Mice, Knockout , Obesity/etiology , Obesity/immunology , Obesity/metabolism , Weight Gain/immunology , Interleukin-22
9.
Nat Immunol ; 12(10): 941-8, 2011 Aug 28.
Article in English | MEDLINE | ID: mdl-21874025

ABSTRACT

Colonic patches (CLPs) and isolated lymphoid follicles (ILFs) are two main lymphoid structures in the colon. Lymphoid tissue-inducer cells (LTi cells) are indispensable for the development of ILFs. LTi cells also produce interleukin 17 (IL-17) and IL-22, signature cytokines secreted by IL-17-producing helper T cells. Here we report that IL-22 acted downstream of the lymphotoxin pathway and regulated the organization and maintenance of mature CLPs and ILFs in the colon during infection with Citrobacter rodentium. Lymphotoxin (LTα(1)ß(2)) regulated the production of IL-22 during infection with C. rodentium, but the lymphotoxin-like protein LIGHT did not. IL-22 signaling was sufficient to restore the organization of CLPs and ILFs and host defense against infection with C. rodentium in mice lacking lymphotoxin signals, which suggests that IL-22 connects the lymphotoxin pathway to mucosal epithelial defense mechanisms.


Subject(s)
Citrobacter rodentium , Colon/immunology , Enterobacteriaceae Infections/immunology , Interleukins/physiology , Lymphoid Tissue/physiology , Lymphotoxin-alpha/physiology , Animals , Colon/microbiology , Interleukin-23/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Interleukin-22
10.
Hepatology ; 52(6): 2158-66, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21064033

ABSTRACT

The matrix metalloproteinase (MMP) family of proteins degrades extracellular matrix (ECM) components as well as processes cytokines and growth factors. MMPs are involved in regulating ECM homeostasis in both normal physiology and disease pathophysiology. Here we report the critical roles of mmp23b in normal zebrafish liver development. Mmp23b was initially identified as a gene linked to the genomic locus of an enhancer trap transgenic zebrafish line in which green fluorescent protein (GFP) expression was restricted to the developing liver. Follow-up analysis of mmp23b messenger RNA (mRNA) expression confirmed its liver-specific expression pattern. Morpholino knockdown of mmp23b resulted in defective hepatocyte proliferation, causing a reduction in liver size while maintaining relatively normal pancreas and gut development. Genetically, we showed that mmp23b functions through the tumor necrosis factor (TNF) signaling pathway. Antisense knockdown of tnfa or tnfb in zebrafish caused similar reductions of liver size, whereas overexpression of tnfa or tnfb rescued liver defects in mmp23b morphants but not vice versa. Biochemically, MMP23B, the human ortholog of Mmp23b, directly interacts with TNF and mediates its release from the cell membrane in a cell culture system. Because mmp23b/MMP23B is highly conserved, our findings in zebrafish warrant further investigation of its role in regulating liver development in mammals.


Subject(s)
Hepatocytes/cytology , Liver/growth & development , Matrix Metalloproteinases/physiology , Zebrafish Proteins/physiology , Animals , Cell Proliferation , Gene Knockdown Techniques , Hepatocytes/drug effects , Humans , Liver/drug effects , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/physiology , Matrix Metalloproteinases/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology , Zebrafish
11.
Blood ; 116(12): 2173-82, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20566898

ABSTRACT

Lymphotoxin-α (LTα), lymphotoxin-ß (LTß), and tumor necrosis factor-α (TNFα) are inflammatory mediators that play crucial roles in lymphoid organ development. We demonstrate here that LTα also contributes to the function of lymphatic vessels and to lymphangiogenesis during inflammation. LTα(-/-) mice exhibited reduced lymph flow velocities and increased interstitial fluid pressure. Airways of LTß(-/-) mice infected with Mycoplasma pulmonis had significantly more lymphangiogenesis than wild type (WT) or LTα(-/-) mice, as did the skin draining immunization sites of LTß(-/-) mice. Macrophages, B cells, and T cells, known sources of LT and TNFα, were apparent in the skin surrounding the immunization sites as were LTα, LTß, and TNFα mRNAs. Ectopic expression of LTα led to the development of LYVE-1 and Prox1-positive lymphatic vessels within tertiary lymphoid organs (TLOs). Quantification of pancreatic lymphatic vessel density in RIPLTαLTß(-/-) and WT mice revealed that LTα was sufficient for inducing lymphangiogenesis and that LTß was not required for this process. Kidneys of inducible LTα transgenic mice developed lymphatic vessels before the appearance of obvious TLOs. These data indicate that LTα plays a significant role in lymphatic vessel function and in inflammation-associated lymphangiogenesis.


Subject(s)
Lymphangiogenesis , Lymphotoxin-alpha/physiology , Animals , Immunization , Inflammation , Kidney/immunology , Lymphatic Vessels , Lymphotoxin-alpha/deficiency , Lymphotoxin-alpha/genetics , Lymphotoxin-beta , Mice , Mice, Knockout , Mycoplasma Infections/pathology , Mycoplasma pulmonis , Skin
12.
Ann Intern Med ; 152(3): 159-66, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20124232

ABSTRACT

Abnormal and exaggerated deposition of extracellular matrix is the hallmark of many fibrotic diseases, including systemic sclerosis and pulmonary, liver, and kidney fibrosis. The spectrum of affected organs, the usually progressive nature of the fibrotic process, the large number of affected persons, and the absence of effective treatment pose an enormous challenge when treating fibrotic diseases. Delineation of the central role of transforming growth factor-beta (TGF-beta) and identification of the specific cellular receptors, kinases, and other mediators involved in the fibrotic process have provided a sound basis for development of effective therapies. The inhibition of signaling pathways activated by TGF-beta represents a novel therapeutic approach for the fibrotic disorders. One of these TGF-beta pathways results in the activation of the nonreceptor tyrosine kinase cellular Abelson (c-Abl), and c-Abl inhibitors, including imatinib mesylate, diminishing the fibrogenic effects of TGF-beta. Thus, recently acquired basic knowledge about the pathogenesis of the fibrotic process has enabled the development of novel therapeutic agents capable of modifying the deleterious effects of the fibrotic diseases.


Subject(s)
Fibrosis/drug therapy , Fibrosis/etiology , Fibrosis/pathology , Humans , Lymphotoxin-alpha/physiology
14.
Infect Immun ; 77(9): 3879-85, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19564380

ABSTRACT

To study the specific role of transmembrane tumor necrosis factor (tmTNF) in protective and pathological responses against the gastrointestinal helminth Trichinella spiralis, we compared the immune responses of TNF-alpha/lymphotoxin alpha (LTalpha)(-/-) mice expressing noncleavable transgenic tmTNF to those of TNF-alpha/LTalpha(-/-) and wild-type mice. The susceptibility of TNF-alpha/LTalpha(-/-) mice to T. spiralis infection was associated with impaired induction of a protective Th2 response and the lack of mucosal mastocytosis. Although tmTNF-expressing transgenic (tmTNF-tg) mice also had a reduced Th2 response, the mast cell response was greater than that observed in TNF-alpha/LTalpha(-/-) mice and was sufficient to induce the expulsion of the parasite. T. spiralis infection of tmTNF-tg mice resulted in significant intestinal pathology characterized by villus atrophy and crypt hyperplasia comparable to that induced following the infection of wild-type mice, while pathology in TNF-alpha/LTalpha(-/-) mice was significantly reduced. Our data thus indicate a role for tmTNF in host defense against gastrointestinal helminths and in the accompanying enteropathy. Furthermore, they also demonstrate that TNF-alpha is required for the induction of Th2 immune responses related to infection with gastrointestinal helminth parasites.


Subject(s)
Intestinal Diseases, Parasitic/immunology , Membrane Proteins/physiology , Trichinella spiralis , Trichinellosis/immunology , Tumor Necrosis Factor-alpha/physiology , Animals , Female , Immunoglobulin E/blood , Interleukin-18/biosynthesis , Interleukin-4/biosynthesis , Lymphotoxin-alpha/physiology , Mice , Mice, Inbred C57BL , Th2 Cells/immunology
15.
Stroke ; 40(3): 970-2, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19182073

ABSTRACT

BACKGROUND AND PURPOSE: Several genes involved in the lymphotoxin-alpha cascade (LTA, LGALS2, and PSMA6) have been linked with the risk of myocardial infarction. Here, we present a comprehensive analysis of these genes in patients with ischemic stroke (IS). METHODS: Twenty-three single nucleotide polymorphisms (SNPs) from LTA, LGALS2, and PSMA6 were genotyped in 601 German IS patients and 736 matched controls. SNPs and haplotypes were tested for association with overall IS, large vessel stroke, and cardioembolic stroke. Significant associations were replicated in an independent sample of 843 IS cases and 933 controls from the UK. RESULTS: Only one SNP (rs1048990 in PSMA6) showed association with overall IS, but this was not replicated in the UK sample. Three SNPs showed significant associations with stroke subtypes (P<0.05), but none of these associations could be replicated in the UK population. CONCLUSIONS: Genetic variation in the lymphotoxin-alpha cascade (LTA, LGALS2, and PSMA6) is not a major risk factor for IS.


Subject(s)
Brain Ischemia/epidemiology , Brain Ischemia/genetics , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/physiology , Stroke/epidemiology , Stroke/genetics , Aged , Case-Control Studies , Female , Galectin 2/genetics , Galectin 2/physiology , Gene Frequency , Genetic Variation , Genotype , Germany/epidemiology , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/physiology , Signal Transduction/physiology , Stroke/classification , United Kingdom/epidemiology
16.
Biochem Biophys Res Commun ; 379(2): 374-8, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19103156

ABSTRACT

We recently reported that the single nucleotide polymorphisms of the lymphotoxin-(LT)alpha gene, a member of the tumor necrosis factor (TNF) family, are closely related to acute myocardial infarction; however, the precise mechanism of LTalpha signaling in atherogenesis remains unclear. We investigated the role of LTalpha3, a secreted homotrimer of LTalpha, in monocyte-endothelial cell adhesion using cultured human umbilical vein endothelial cells (HUVEC). We found that LTalpha3 induced cell adhesion molecules and activated NF-kappaB p50 and p65. LTalpha3 also induced phosphorylation of Akt, phosphorylation and degradation of IkappaB, nuclear translocation of p65, and increased adhesion of THP1 monocytes to HUVEC. These effects were mediated by TNF receptor (TNFR) I and attenuated by the phosphatidylinositol triphosphate-kinase (PI3K) inhibitors LY294002 and Wortmannin. Thus, LTalpha3 mediates the monocyte-endothelial interaction via the classical NF-kappaB pathway following TNFR I/PI3K activation, indicating it may play a role in the development of coronary artery disease.


Subject(s)
Cell Communication , Endothelial Cells/physiology , Lymphotoxin-alpha/physiology , Monocytes/physiology , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Androstadienes/pharmacology , Cell Adhesion , Cell Communication/genetics , Cells, Cultured , Chromones/pharmacology , Coronary Disease/genetics , Coronary Disease/metabolism , Humans , Lymphotoxin-alpha/genetics , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Wortmannin
17.
Eur J Immunol ; 39(1): 280-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19089813

ABSTRACT

In this report we describe a transplantation system where embryonic spleens are grafted into adult hosts. This model can be used to analyze the cellular and molecular requirements for the development and organization of splenic microenvironments. Whole embryonic day 15 (ED15) spleens, grafted under the kidney capsule of adult mice, were colonized by host-derived lymphocytes and DC and developed normal splenic architecture. Grafts were also able to form germinal centers in response to T-dependent antigen. Using this system we demonstrated that adult host-derived lymphotoxin (LT) alpha was sufficient for the development of ED15 LT alpha(-/-) grafts. Grafting of ED15 LT alpha(-/-) spleens into RAG(-/-) hosts followed by transfer of LT alpha(-/-) splenocytes revealed no requirement for lymphocyte-derived LT alpha in the induction of CCL21 or the development of T-zone stroma. These data suggest that interactions between adult lymphoid-tissue inducer-like cells and embryonic stromal cells initiated T-zone development. Furthermore, adult lymphoid tissue inducer-like cells were shown to develop from bone marrow-derived progenitors. The model described here demonstrates a method of transferring whole splenic microenvironments and dissecting the stromal and hematopoietic signals involved in spleen development and organization.


Subject(s)
Cell Communication/immunology , Models, Animal , Spleen/embryology , Spleen/immunology , T-Lymphocytes/immunology , Animals , Antigens, Viral, Tumor/immunology , Cell Differentiation/immunology , Chemokine CCL21/metabolism , Kidney/immunology , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Spleen/cytology , Spleen/transplantation
18.
Genes Immun ; 9(7): 613-23, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18650831

ABSTRACT

Cervical cancer has been associated with specific human leukocyte antigen (HLA) haplotypes/alleles and with polymorphisms at the nearby non-HLA loci TNF, LTA, TAP1 and TAP2. Distinguishing effects of individual loci in the major histocompatibility complex (MHC) region are difficult due to the complex linkage disequilibrium (LD) pattern characterized by high LD, punctuated by recombination hot spots. We have evaluated the association of polymorphism at HLA class II DQB1 and the TNF, LTA, TAP1 and TAP2 genes with cervical cancer risk, using 1306 familial cases and 288 controls. DQB1 was strongly associated; alleles *0301, *0402 and (*)0602 increased cancer susceptibility, whereas *0501 and *0603 decreased susceptibility. Among the non-HLA loci, association was only detected for the TAP2 665 polymorphism, and interallelic disequilibrium analysis indicated that this could be due to LD with DQB1. As the TAP2 665 association was seen predominantly in non-carriers of DQB1 susceptibility alleles, we hypothesized that TAP2 665 may have an effect not attributable to LD with DQB1. However, a logistic regression analysis suggested that TAP2 665 was strongly influenced by LD with DQB1. Our results emphasize the importance of large sample sizes and underscore the necessity of examining both HLA and non-HLA loci in the MHC to assign association to the correct locus.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Genetic Predisposition to Disease , HLA-DQ Antigens/genetics , Lymphotoxin-alpha/genetics , Tumor Necrosis Factor-alpha/genetics , Uterine Cervical Neoplasms/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP Binding Cassette Transporter, Subfamily B, Member 3 , ATP-Binding Cassette Transporters/physiology , Adult , Alleles , Case-Control Studies , Female , HLA-DQ Antigens/physiology , HLA-DQ beta-Chains , Humans , Linkage Disequilibrium/genetics , Lymphotoxin-alpha/physiology , Polymorphism, Single Nucleotide , Risk Factors , Tumor Necrosis Factor-alpha/physiology
19.
Biochem Biophys Res Commun ; 370(3): 461-7, 2008 Jun 06.
Article in English | MEDLINE | ID: mdl-18384745

ABSTRACT

The TNF family is critical for development of lymphoid organs and plays significant roles in regulation of innate and adoptive immune responses. Here, we describe a novel N-ethyl-N-nitrosourea (ENU)-induced mutant strain, HLB382, with a point mutation in the Lta gene, which encodes lymphotoxin (LT) alpha, a member of the TNF family. Mutant mice had no lymph nodes and no Peyer's patches. Microscopically, the spleens had disordered follicles and no germinal centers or discernible marginal zones (MZ). While the development of T cells and follicular B cells was normal, the numbers of NK and MZ B cells were significantly reduced. Interestingly, the numbers of peritoneal B1b cells were significantly increased. Genomic DNA sequences revealed a single base pair insertion in the coding region of Lta resulting in a frame shift and a premature stop codon. This new strain provides opportunities for understanding the full range of Lta gene function on a pure C57BL/6 background.


Subject(s)
Lymph Nodes/abnormalities , Lymphotoxin-alpha/genetics , Mice, Inbred C57BL , Organogenesis/genetics , Peyer's Patches/abnormalities , Animals , B-Lymphocytes/immunology , Bone Marrow/immunology , Chromosome Mapping , Dendritic Cells/immunology , Ethylnitrosourea/toxicity , Granulocytes/immunology , Killer Cells, Natural/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphotoxin-alpha/physiology , Mice , Peritoneum/immunology , Peyer's Patches/cytology , Peyer's Patches/immunology , Point Mutation , T-Lymphocytes/immunology
20.
J Immunol ; 180(4): 2284-93, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18250437

ABSTRACT

Germinal center (GC) responses to T-dependent Ags require effective collaboration between Th cells, activated B cells, and follicular dendritic cells within a highly organized microenvironment. Studies using gene-targeted mice have highlighted nonredundant molecules that are key for initiating and maintaining the GC niche, including the molecules of the ICOS, CD40, and lymphotoxin (LT) pathways. Signaling through ICOS has multiple consequences, including cytokine production, expression of CD40L on Th cells, and differentiation into CXCR5(+) follicular Th cells, all of which are important in the GC reaction. We have therefore taken advantage of ICOS(-/-) mice to dissect which downstream elements are required to initiate the formation of GC. In the context of a T-dependent immune response, we found that GC B cells from ICOS(-/-) mice express lower levels of LTalphabeta compared with wild-type GC B cells in vivo, and stimulation of ICOS on T cells induces LTalphabeta on B cells in vitro. Administration of agonistic anti-LTbeta receptor Ab was unable to restore the GC response in ICOS(-/-) mice, suggesting that additional input from another pathway is required for optimal GC generation. In contrast, treatment with agonistic anti-CD40 Ab in vivo recovered GC networks and restored LTalphabeta expression on GC B cells in ICOS(-/-) mice, and this effect was dependent on LTbeta receptor signaling. Collectively, these data demonstrate that ICOS activation is a prerequisite for the up-regulation of LTalphabeta on GC B cells in vivo and provide a model for cooperation between ICOS, CD40, and LT pathways in the context of the GC response.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/physiology , CD40 Antigens/physiology , Germinal Center/immunology , Germinal Center/metabolism , Lymphotoxin beta Receptor/physiology , Signal Transduction/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Chickens , Germinal Center/pathology , Inducible T-Cell Co-Stimulator Protein , Lymphotoxin beta Receptor/deficiency , Lymphotoxin beta Receptor/genetics , Lymphotoxin-alpha/biosynthesis , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitrophenols/administration & dosage , Nitrophenols/immunology , Phenylacetates , gamma-Globulins/administration & dosage , gamma-Globulins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL