Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
1.
J Autoimmun ; 144: 103177, 2024 04.
Article in English | MEDLINE | ID: mdl-38368767

ABSTRACT

Psoriasis (PS) and atopic dermatitis (AD) are common skin inflammatory diseases characterized by hyper-responsive keratinocytes. Although, some cytokines have been suggested to be specific for each disease, other cytokines might be central to both diseases. Here, we show that Tumor necrosis factor superfamily member 14 (TNFSF14), known as LIGHT, is required for experimental PS, similar to its requirement in experimental AD. Mice devoid of LIGHT, or deletion of either of its receptors, lymphotoxin ß receptor (LTßR) and herpesvirus entry mediator (HVEM), in keratinocytes, were protected from developing imiquimod-induced psoriatic features, including epidermal thickening and hyperplasia, and expression of PS-related genes. Correspondingly, in single cell RNA-seq analysis of PS patient biopsies, LTßR transcripts were found strongly expressed with HVEM in keratinocytes, and LIGHT was upregulated in T cells. Similar transcript expression profiles were also seen in AD biopsies, and LTßR deletion in keratinocytes also protected mice from allergen-induced AD features. Moreover, in vitro, LIGHT upregulated a broad spectrum of genes in human keratinocytes that are clinical features of both PS and AD skin lesions. Our data suggest that agents blocking LIGHT activity might be useful for therapeutic intervention in PS as well as in AD.


Subject(s)
Dermatitis, Atopic , Psoriasis , Humans , Mice , Animals , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Dermatitis, Atopic/genetics , Dermatitis, Atopic/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Keratinocytes/metabolism , Cytokines/metabolism , Psoriasis/genetics , Psoriasis/metabolism , Inflammation/metabolism
2.
EMBO Rep ; 24(3): e54228, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36633157

ABSTRACT

Estrogen is a disease-modifying factor in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) via estrogen receptor alpha (ERα). However, the mechanisms by which ERα signaling contributes to changes in disease pathogenesis have not been completely elucidated. Here, we demonstrate that ERα deletion in dendritic cells (DCs) of mice induces severe neurodegeneration in the central nervous system in a mouse EAE model and resistance to interferon beta (IFNß), a first-line MS treatment. Estrogen synthesized by extragonadal sources is crucial for controlling disease phenotypes. Mechanistically, activated ERα directly interacts with TRAF3, a TLR4 downstream signaling molecule, to degrade TRAF3 via ubiquitination, resulting in reduced IRF3 nuclear translocation and transcription of membrane lymphotoxin (mLT) and IFNß components. Diminished ERα signaling in DCs generates neurotoxic effector CD4+ T cells via mLT-lymphotoxin beta receptor (LTßR) signaling. Lymphotoxin beta receptor antagonist abolished EAE disease symptoms in the DC-specific ERα-deficient mice. These findings indicate that estrogen derived from extragonadal sources, such as lymph nodes, controls TRAF3-mediated cytokine production in DCs to modulate the EAE disease phenotype.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Estrogen Receptor alpha , Mice , Animals , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , Estrogens/pharmacology , Phenotype , Dendritic Cells/metabolism , Mice, Inbred C57BL
3.
J Allergy Clin Immunol ; 151(4): 976-990.e5, 2023 04.
Article in English | MEDLINE | ID: mdl-36473503

ABSTRACT

BACKGROUND: Dysregulation of airway smooth muscle cells (ASM) is central to the severity of asthma. Which molecules dominantly control ASM in asthma is unclear. High levels of the cytokine LIGHT (aka TNFSF14) have been linked to asthma severity and lower baseline predicted FEV1 percentage, implying that signals through its receptors might directly control ASM dysfunction. OBJECTIVE: Our study sought to determine whether signaling via lymphotoxin beta receptor (LTßR) or herpesvirus entry mediator from LIGHT dominantly drives ASM hyperreactivity induced by allergen. METHODS: Conditional knockout mice deficient for LTßR or herpesvirus entry mediator in smooth muscle cells were used to determine their role in ASM deregulation and airway hyperresponsiveness (AHR) in vivo. Human ASM were used to study signals induced by LTßR. RESULTS: LTßR was strongly expressed in ASM from normal and asthmatic subjects compared to several other receptors implicated in smooth muscle deregulation. Correspondingly, conditional deletion of LTßR only in smooth muscle cells in smMHCCreLTßRfl/fl mice minimized changes in their numbers and mass as well as AHR induced by house dust mite allergen in a model of severe asthma. Intratracheal LIGHT administration independently induced ASM hypertrophy and AHR in vivo dependent on direct LTßR signals to ASM. LIGHT promoted contractility, hypertrophy, and hyperplasia of human ASM in vitro. Distinguishing LTßR from the receptors for IL-13, TNF, and IL-17, which have also been implicated in smooth muscle dysregulation, LIGHT promoted NF-κB-inducing kinase-dependent noncanonical nuclear factor kappa-light-chain enhancer of activated B cells in ASM in vitro, leading to sustained accumulation of F-actin, phosphorylation of myosin light chain kinase, and contractile activity. CONCLUSIONS: LTßR signals directly and dominantly drive airway smooth muscle hyperresponsiveness relevant for pathogenesis of airway remodeling in severe asthma.


Subject(s)
Asthma , Receptors, Tumor Necrosis Factor, Member 14 , Humans , Mice , Animals , Lymphotoxin beta Receptor/genetics , Asthma/pathology , Muscle, Smooth , Myocytes, Smooth Muscle/pathology , Mice, Knockout , Allergens , Lung/pathology
4.
Clin Investig Arterioscler ; 35(1): 1-11, 2023.
Article in English, Spanish | MEDLINE | ID: mdl-35738949

ABSTRACT

OBJECTIVE: Vascular smooth muscle cells (VSMCs) undergo a phenotypic-switching process during the generation of unstable atheroma plaques. In this investigation, the potential implication of the tumor necrosis factor superfamily (TNFSF) ligands, in the gene expression signature associated with VSMC plasticity was studied. MATERIAL AND METHODS: Human aortic (ha)VSMCs were obtained commercially and treated with the cytokine TNFSF14, also called LIGHT, the lymphotoxin alpha (LTα), the heterotrimer LTα1ß2 or with vehicle for 72h. The effect of the different treatments on gene expression was analyzed by quantitative PCR and included the study of genes associated with myofibroblast-like cell function, osteochondrogenesis, pluripotency, lymphorganogenesis and macrophage-like cell function. RESULTS: HaVSMCs displayed a change in myofibroblast-like cell genes which consisted in reduced COL1A1 and TGFB1 mRNA levels when treated with LTα or LIGHT and with augmented MMP9 expression levels when treated with LTα. LTα and LIGHT treatments also diminished the expression of genes associated with osteochondrogenesis and pluripotency SOX9, CKIT, and KLF4. By contrary, all the above genes were no affected by the treatment with the trimer LTα1ß2. In addition, haVSMC treatment with LTα, LTα1ß2 and LIGHT altered lymphorganogenic cytokine gene expression which consisted of augmented CCL20 and CCL21 mRNA levels by LTα and a reduction in the gene expression of CCL21 and CXCL13 by LIGHT and LTα1ß2 respectively. Neither, LTα or LIGHT or LTα1ß2 treatments affected the expression of macrophage-like cell markers in haVSMC. CONCLUSIONS: Altogether, indicates that the TNFSF ligands through their interconnected network of signaling, are important in the preservation of VSMC identity against the acquisition of a genetic expression signature compatible with functional cellular plasticity.


Subject(s)
Lymphotoxin beta Receptor , Muscle, Smooth, Vascular , Humans , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Cytokines , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
J Reprod Immunol ; 153: 103693, 2022 09.
Article in English | MEDLINE | ID: mdl-35987137

ABSTRACT

Intrauterine adhesion (IUA) is a fibrotic disease, with complex and multifactorial process, causing menstrual disorders, pregnancy loss or infertility. LIGHT (also named TNFSF14), mainly expressed by immune cells, has been reported to be associated with tissue fibrosis. However, the features of immunocyte subsets, the expression and roles of LIGHT and its receptor HVEM (herpes virus entry mediator) and LTßR (lymphotoxin beta receptor) in IUA remain largely unknown. Compared with the control group, we observed increased ratios of CD45+ cells, neutrophils, T cells, macrophages and decreased natural killer cells proportion, and high LIGHT expression on CD4+ T cells and macrophages in IUA endometrium. Further analysis showed there was a positive correlation between upregulated profibrotic factors (e.g., ɑ-smooth muscle actin, transforming growth factor ß1) and HVEM in IUA endometrial tissue. More importantly, recombinant human LIGHT protein directly up-regulated the expression of HVEM, LTßR, profibrotic and proinflammatory factors expression in human endometrial stromal cells. These findings reveal abnormal changes of immune cell subsets proportion and the overexpression of LIGHT-HVEM/LTßR axis in IUA endometrium, should contribute to inflammation and fibrosis formation of IUA.


Subject(s)
Lymphotoxin beta Receptor , Receptors, Tumor Necrosis Factor, Member 14 , Tumor Necrosis Factor Ligand Superfamily Member 14 , Uterine Diseases , Actins , Female , Fibrosis/genetics , Humans , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/physiology , Pregnancy , Receptors, Tumor Necrosis Factor, Member 14/genetics , Signal Transduction , Transforming Growth Factor beta1 , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Uterine Diseases/genetics , Uterine Diseases/pathology
6.
PLoS One ; 17(8): e0270907, 2022.
Article in English | MEDLINE | ID: mdl-35925983

ABSTRACT

Multiple myeloma (MM), a malignancy of plasma cells (PCs), has diverse genetic underpinnings and in rare cases these include amplification of the lymphotoxin b receptor (Ltbr) locus. LTßR has well defined roles in supporting lymphoid tissue development and function through actions in stromal and myeloid cells, but whether it is functional in PCs is unknown. Here we showed that Ltbr mRNA was upregulated in mouse PCs compared to follicular B cells, but deficiency in the receptor did not cause a reduction in PC responses to a T-dependent or T-independent immunogen. However, LTßR overexpression (OE) enhanced PC formation in vitro after LPS or anti-CD40 stimulation. In vivo, LTßR OE led to increased antigen-specific splenic and bone marrow (BM) plasma cells responses. LTßR OE PCs had increased expression of Nfkb2 and of the NF-kB target genes Bcl2 and Mcl1, factors involved in the formation of long-lived BM PCs. Our findings suggest a pathway by which Ltbr gene amplifications may contribute to MM development through increased NF-kB activity and induction of an anti-apoptotic transcriptional program.


Subject(s)
NF-kappa B , Plasma Cells , Animals , B-Lymphocytes/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Plasma Cells/metabolism , Spleen/metabolism
7.
Mucosal Immunol ; 15(2): 327-337, 2022 02.
Article in English | MEDLINE | ID: mdl-34903876

ABSTRACT

Fibroblasts mediate tissue remodeling in eosinophilic esophagitis (EoE), a chronic allergen-driven inflammatory pathology. Diverse fibroblast subtypes with homeostasis-regulating or inflammatory profiles have been recognized in various tissues, but which mediators induce these alternate differentiation states remain largely unknown. We recently identified that TNFSF14/LIGHT promotes an inflammatory esophageal fibroblast in vitro. Herein we used esophageal biopsies and primary fibroblasts to investigate the role of the LIGHT receptors, herpes virus entry mediator (HVEM) and lymphotoxin-beta receptor (LTßR), and their downstream activated pathways, in EoE. In addition to promoting inflammatory gene expression, LIGHT down-regulated homeostatic factors including WNTs, BMPs and type 3 semaphorins. In vivo, WNT2B+ fibroblasts were decreased while ICAM-1+ and IL-34+ fibroblasts were expanded in EoE, suggesting that a LIGHT-driven gene signature was imprinted in EoE versus normal esophageal fibroblasts. HVEM and LTßR overexpression and deficiency experiments demonstrated that HVEM regulates a limited subset of LIGHT targets, whereas LTßR controls all transcriptional effects. Pharmacologic blockade of the non-canonical NIK/p100/p52-mediated NF-κB pathway potently silenced LIGHT's transcriptional effects, with a lesser role found for p65 canonical NF-κB. Collectively, our results show that LIGHT promotes differentiation of esophageal fibroblasts toward an inflammatory phenotype and represses homeostatic gene expression via a LTßR-NIK-p52 NF-κB dominant pathway.


Subject(s)
Esophagus , Inflammation , Transcriptome , Tumor Necrosis Factor Ligand Superfamily Member 14 , Esophagus/metabolism , Fibroblasts/metabolism , Homeostasis , Humans , Inflammation/genetics , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , NF-kappa B/metabolism , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
8.
Int Immunol ; 34(1): 45-52, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34687536

ABSTRACT

Medullary thymic epithelial cells (mTECs) help shape the thymic microenvironment for T-cell development by expressing a variety of peripheral tissue-restricted antigens (TRAs). The self-tolerance of T cells is established by negative selection of autoreactive T cells that bind to TRAs. To increase the diversity of TRAs, a fraction of mTECs terminally differentiates into distinct subsets resembling atypical types of epithelial cells in specific peripheral tissues. As such, thymic tuft cells that express peripheral tuft cell genes have recently emerged. Here, we show that the transcription factor SRY-box transcription factor 4 (Sox4) is highly expressed in mTECs and is essential for the development of thymic tuft cells. Mice lacking Sox4 specifically in TECs had a significantly reduced number of thymic tuft cells with no effect on the differentiation of other mTEC subsets, including autoimmune regulator (Aire)+ and Ccl21a+ mTECs. Furthermore, Sox4 expression was diminished in mice deficient in TEC-specific lymphotoxin ß receptor (LTßR), indicating a role for the LTßR-Sox4 axis in the differentiation of thymic tuft cells. Given that Sox4 promotes differentiation of peripheral tuft cells, our findings suggest that mTECs employ the same transcriptional program as peripheral epithelial cells. This mechanism may explain how mTECs diversify peripheral antigen expression to project an immunological self within the thymic medulla.


Subject(s)
Lymphotoxin beta Receptor/genetics , SOXC Transcription Factors/genetics , Thymus Gland/immunology , Animals , Cell Differentiation/immunology , Lymphotoxin beta Receptor/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , SOXC Transcription Factors/immunology , Signal Transduction/genetics , Thymus Gland/cytology
9.
Front Immunol ; 12: 712632, 2021.
Article in English | MEDLINE | ID: mdl-34335629

ABSTRACT

Lymphotoxin beta receptor (LTßR) is a promising therapeutic target in autoimmune and infectious diseases as well as cancer. Mice with genetic inactivation of LTßR display multiple defects in development and organization of lymphoid organs, mucosal immune responses, IgA production and an autoimmune phenotype. As these defects are imprinted in embryogenesis and neonate stages, the impact of LTßR signaling in adulthood remains unclear. Here, to overcome developmental defects, we generated mice with inducible ubiquitous genetic inactivation of LTßR in adult mice (iLTßRΔ/Δ mice) and redefined the role of LTßR signaling in organization of lymphoid organs, immune response to mucosal bacterial pathogen, IgA production and autoimmunity. In spleen, postnatal LTßR signaling is required for development of B cell follicles, follicular dendritic cells (FDCs), recruitment of neutrophils and maintenance of the marginal zone. Lymph nodes of iLTßRΔ/Δ mice were reduced in size, lacked FDCs, and had disorganized subcapsular sinus macrophages. Peyer`s patches were smaller in size and numbers, and displayed reduced FDCs. The number of isolated lymphoid follicles in small intestine and colon were also reduced. In contrast to LTßR-/- mice, iLTßRΔ/Δ mice displayed normal thymus structure and did not develop signs of systemic inflammation and autoimmunity. Further, our results suggest that LTßR signaling in adulthood is required for homeostasis of neutrophils, NK, and iNKT cells, but is dispensable for the maintenance of polyclonal IgA production. However, iLTßRΔ/Δ mice exhibited an increased sensitivity to C. rodentium infection and failed to develop pathogen-specific IgA responses. Collectively, our study uncovers new insights of LTßR signaling in adulthood for the maintenance of lymphoid organs, neutrophils, NK and iNKT cells, and IgA production in response to mucosal bacterial pathogen.


Subject(s)
Aging/immunology , Lymphoid Tissue/immunology , Lymphotoxin beta Receptor/physiology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Autoimmunity , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Citrobacter rodentium/immunology , Crosses, Genetic , Gene Expression Regulation, Developmental , Homeostasis/immunology , Immunoglobulin A/biosynthesis , Immunoglobulin A/immunology , Inflammation , Killer Cells, Natural/immunology , Lymphoid Tissue/cytology , Lymphotoxin beta Receptor/biosynthesis , Lymphotoxin beta Receptor/deficiency , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred MRL lpr , Mice, Transgenic , Neutrophils/immunology , Sequence Deletion , Specific Pathogen-Free Organisms , Splenomegaly/immunology
10.
Infect Immun ; 89(6)2021 05 17.
Article in English | MEDLINE | ID: mdl-33753412

ABSTRACT

The lymphotoxin ß receptor (LTßR) plays an essential role in the initiation of immune responses to intracellular pathogens. In mice, the LTßR is crucial for surviving acute toxoplasmosis; however, until now, a functional analysis was largely incomplete. Here, we demonstrate that the LTßR is a key regulator required for the intricate balance of adaptive immune responses. Toxoplasma gondii-infected LTßR-deficient (LTßR-/-) mice show globally altered interferon-γ (IFN-γ) regulation, reduced IFN-γ-controlled host effector molecule expression, impaired T cell functionality, and an absent anti-parasite-specific IgG response, resulting in a severe loss of immune control of the parasites. Reconstitution of LTßR-/- mice with toxoplasma immune serum significantly prolongs survival following T. gondii infection. Notably, analysis of RNA-seq data clearly indicates a specific effect of T. gondii infection on the B cell response and isotype switching. This study uncovers the decisive role of the LTßR in cytokine regulation and adaptive immune responses to control T. gondii.


Subject(s)
Adaptive Immunity , Host-Parasite Interactions/immunology , Immunity, Innate , Lymphotoxin beta Receptor/metabolism , Toxoplasma/immunology , Toxoplasmosis/immunology , Toxoplasmosis/metabolism , Animals , Disease Models, Animal , Lymphotoxin beta Receptor/genetics , Mice , Mice, Knockout , Toxoplasmosis/parasitology
11.
J Exp Med ; 218(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33724364

ABSTRACT

The spleen contains a myriad of conventional dendritic cell (cDC) subsets that protect against systemic pathogen dissemination by bridging antigen detection to the induction of adaptive immunity. How cDC subsets differentiate in the splenic environment is poorly understood. Here, we report that LTα1ß2-expressing Rorgt+ ILC3s, together with B cells, control the splenic cDC niche size and the terminal differentiation of Sirpα+CD4+Esam+ cDC2s, independently of the microbiota and of bone marrow pre-cDC output. Whereas the size of the splenic cDC niche depended on lymphotoxin signaling only during a restricted time frame, the homeostasis of Sirpα+CD4+Esam+ cDC2s required continuous lymphotoxin input. This latter property made Sirpα+CD4+Esam+ cDC2s uniquely susceptible to pharmacological interventions with LTßR agonists and antagonists and to ILC reconstitution strategies. Together, our findings demonstrate that LTα1ß2-expressing Rorgt+ ILC3s drive splenic cDC differentiation and highlight the critical role of ILC3s as perpetual regulators of lymphoid tissue homeostasis.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate , Lymphoid Tissue/immunology , Lymphotoxin-alpha/immunology , Signal Transduction/immunology , Spleen/immunology , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Dendritic Cells/metabolism , Female , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction/genetics , Spleen/cytology , Spleen/metabolism
12.
Mucosal Immunol ; 14(3): 679-690, 2021 05.
Article in English | MEDLINE | ID: mdl-33568785

ABSTRACT

Inflammatory bowel disease is characterized by an exacerbated intestinal immune response, but the critical mechanisms regulating immune activation remain incompletely understood. We previously reported that the TNF-superfamily molecule TNFSF14 (LIGHT) is required for preventing severe disease in mouse models of colitis. In addition, deletion of lymphotoxin beta receptor (LTßR), which binds LIGHT, also led to aggravated colitis pathogenesis. Here, we aimed to determine the cell type(s) requiring LTßR and the mechanism critical for exacerbation of colitis. Specific deletion of LTßR in neutrophils (LTßRΔN), but not in several other cell types, was sufficient to induce aggravated colitis and colonic neutrophil accumulation. Mechanistically, RNA-Seq analysis revealed LIGHT-induced suppression of cellular metabolism, and mitochondrial function, that was dependent on LTßR. Functional studies confirmed increased mitochondrial mass and activity, associated with excessive mitochondrial ROS production and elevated glycolysis at steady-state and during colitis. Targeting these metabolic changes rescued exacerbated disease severity. Our results demonstrate that LIGHT signals to LTßR on neutrophils to suppress metabolic activation and thereby prevents exacerbated immune pathogenesis during colitis.


Subject(s)
Colitis/immunology , Inflammatory Bowel Diseases/immunology , Lymphotoxin beta Receptor/metabolism , Mitochondria/metabolism , Neutrophils/metabolism , Activation, Metabolic , Animals , Dextran Sulfate , Disease Models, Animal , Disease Progression , Humans , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
13.
PLoS One ; 16(2): e0247368, 2021.
Article in English | MEDLINE | ID: mdl-33606781

ABSTRACT

Osteoporosis is a progressive systemic skeletal disease associated with decreased bone mineral density and deterioration of bone quality, and it affects millions of people worldwide. Currently, it is treated mainly using antiresorptive and osteoanabolic agents. However, these drugs have severe adverse effects. Cell replacement therapy using mesenchymal stem cells (MSCs) could serve as a treatment strategy for osteoporosis in the future. LIGHT (HVEM-L, TNFSF14, or CD258) is a member of the tumor necrosis factor superfamily. However, the effect of recombinant LIGHT (rhLIGHT) on osteogenesis in human bone marrow-derived MSCs (hBM-MSCs) is unknown. Therefore, we monitored the effects of LIGHT on osteogenesis of hBM-MSCs. Lymphotoxin-ß receptor (LTßR), which is a LIGHT receptor, was constitutively expressed on the surface of hBM-MSCs. After rhLIGHT treatment, calcium and phosphate deposition in hBM-MSCs, stained by Alizarin red and von Kossa, respectively, significantly increased. We performed quantitative real-time polymerase chain reaction to examine the expressions of osteoprogenitor markers (RUNX2/CBFA1 and collagen I alpha 1) and osteoblast markers (alkaline phosphatase, osterix/Sp7, and osteocalcin) and immunoblotting to assess the underlying biological mechanisms following rhLIGHT treatment. We found that rhLIGHT treatment enhanced von Kossa- and Alizarin red-positive hBM-MSCs and induced the expression of diverse differentiation markers of osteogenesis in a dose-dependent manner. WNT/ß-catenin pathway activation strongly mediated rhLIGHT-induced osteogenesis of hBM-MSCs, accelerating the differentiation of hBM-MSCs into osteocytes. In conclusion, the interaction between LIGHT and LTßR enhances osteogenesis of hBM-MSCs. Therefore, LIGHT might play an important role in stem cell therapy.


Subject(s)
Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genetic Markers , Humans , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Recombinant Proteins/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Wnt Signaling Pathway/drug effects
14.
J Cell Physiol ; 236(6): 4725-4737, 2021 06.
Article in English | MEDLINE | ID: mdl-33269476

ABSTRACT

The differentiation of mature medullary thymic epithelial cells (mTECs) is critical for the induction of central immune tolerance. Although the critical effect of mechanistic target of rapamycin complex 1 (mTORC1) in shaping mTEC differentiation has been studied, the regulatory role of mTORC2 in the differentiation and maturation of mTECs is poorly understood. We herein reported that TEC-specific ablation of a rapamycin-insensitive companion of mTOR (RICTOR), a key component of mTORC2, significantly decreased the thymus size and weight, the total cell number of TECs, and the cell number of mTECs with a smaller degree of reduced cortical thymic epithelial cells. Interestingly, RICTOR deficiency significantly accelerated the mTEC maturation process, as indicated by the increased ratios of mature mTECs (MHCIIhi , CD80+ , and Aire+ ) to immature mTECs (MHCIIlo , CD80- , and Aire- ) in Rictor-deficient mice. The RNA-sequencing assays showed that the upregulated nuclear factor-κB (NF-κB) signaling pathway in Rictor-deficient mTECs was one of the obviously altered pathways compared with wild-type mTECs. Our studies further showed that Rictor-deficient mTECs exhibited upregulated expression of receptor activator of NF-κB (RANK) and lymphotoxin ß receptor (LTßR), as well as increased activity of canonical and noncanonical NF-κB signaling pathways as determined by ImageStream and Simple Western. Finally, our results showed that inhibition of NF-κB signaling pathways could partially reverse the accelerated maturation of mTECs in Rictor conditional KO mice. Thus, mTORC2 negatively controls the kinetics of the mTEC maturation process by inhibiting the LTßR/RANK-NF-κB signal axis.


Subject(s)
Cell Differentiation , Epithelial Cells/enzymology , Lymphotoxin beta Receptor/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , NF-kappa B/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Thymus Gland/enzymology , Animals , Epithelial Cells/pathology , Gene Expression Regulation , Kinetics , Lymphotoxin beta Receptor/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Mice, Knockout , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Signal Transduction , Thymocytes/enzymology , Thymocytes/pathology , Thymus Gland/pathology
15.
Int Immunopharmacol ; 89(Pt A): 106999, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33045563

ABSTRACT

Cisplatin is widely used as a chemotherapeutic agent for treating patients with solid tumors. The most common side effect of cisplatin treatment is nephrotoxicity. Recent studies have shown that mitochondrial apoptotic pathways are involved in cisplatin-induced acute kidney injury (Cis-AKI). LIGHT, the 14th member of the tumor necrosis factor superfamily (TNFSF14), was found to induce apoptosis of certain types of tumor cells. So far, a link between LIGHT and Cis-AKI has not been reported. In this study, we observed that expression of LIGHT and its receptors HVEM and LTßR was increased in kidney tissues of mice after cisplatin treatment. LIGHT deficiency aggravated kidney injury, as evidenced by more severe tubular injury; remarkably increased levels of serum creatinine (Scr), blood urea nitrogen (BUN), and both kidney injury molecule-1 (KIM-1) and inflammatory cytokine mRNAs in renal tissues. Moreover, in the renal tissues of LIGHT KO mice, cisplatin-induced mitochondrion injury and the levels of the pro-apoptotic molecules Bax, Cytochrome C (Cyt C), cleaved caspase-3, and cleaved caspase-9 were dramatically increased; in contrast, the expression of anti-apoptotic molecule Bcl-2 was markedly reduced, compared to those in WT mice, suggesting that LIGHT deficiency accelerated cisplatin-induced mitochondrial apoptosis of renal tubular cells in these mice. Accordingly, treatment with recombinant human LIGHT (rLIGHT) was shown to alleviate cisplatin-induced kidney injury in vivo. Similar results were observed after the human renal tubular epithelial cell line HK-2 cells exposure to rLIGHT stimulation, evidenced by the reduction in the mitochondrion dysfunction (as confirmed by the significant reduced oxidative stress and membrane potential changes) and in the percentage of cells apoptosis. While blocking LIGHT with the soluble fusion protein LTßR-Ig or HVEM-Ig accelerated the HK-2 cells apoptosis. In conclusion, LIGHT deficiency aggravates Cis-AKI by promoting mitochondrial apoptosis pathways.


Subject(s)
Acute Kidney Injury/metabolism , Apoptosis , Cisplatin , Kidney Tubules/metabolism , Mitochondria/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line , Disease Models, Animal , Down-Regulation , Humans , Kidney Tubules/pathology , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
16.
Nat Immunol ; 21(10): 1172-1180, 2020 10.
Article in English | MEDLINE | ID: mdl-32839611

ABSTRACT

Fibroblasts are one of the most common but also neglected types of stromal cells, the heterogeneity of which underlies the specific function of tissue microenvironments in development and regeneration. In the thymus, autoreactive T cells are thought to be negatively selected by reference to the self-antigens expressed in medullary epithelial cells, but the contribution of other stromal cells to tolerance induction has been poorly examined. In the present study, we report a PDGFR+ gp38+ DPP4- thymic fibroblast subset that is required for T cell tolerance induction. The deletion of the lymphotoxin ß-receptor in thymic fibroblasts caused an autoimmune phenotype with decreased expression of tissue-restricted and fibroblast-specific antigens, offering insight into the long-sought target of lymphotoxin signaling in the context of the regulation of autoimmunity. Thus, thymic medullary fibroblasts play an essential role in the establishment of central tolerance by producing a diverse array of self-antigens.


Subject(s)
Fibroblasts/immunology , T-Lymphocytes/immunology , Thymus Gland/metabolism , Animals , Autoantigens/immunology , Autoimmunity , Cells, Cultured , Cellular Microenvironment , Clonal Selection, Antigen-Mediated , Dipeptidyl Peptidase 4/metabolism , Immune Tolerance , Lymphotoxin beta Receptor/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Thymus Gland/cytology
17.
Nat Commun ; 11(1): 2198, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366944

ABSTRACT

The thymus supports multiple αß T cell lineages that are functionally distinct, but mechanisms that control this multifaceted development are poorly understood. Here we examine medullary thymic epithelial cell (mTEC) heterogeneity and its influence on CD1d-restricted iNKT cells. We find three distinct mTEClow subsets distinguished by surface, intracellular and secreted molecules, and identify LTßR as a cell-autonomous controller of their development. Importantly, this mTEC heterogeneity enables the thymus to differentially control iNKT sublineages possessing distinct effector properties. mTEC expression of LTßR is essential for the development thymic tuft cells which regulate NKT2 via IL-25, while LTßR controls CD104+CCL21+ mTEClow that are capable of IL-15-transpresentation for regulating NKT1 and NKT17. Finally, mTECs regulate both iNKT-mediated activation of thymic dendritic cells, and iNKT availability in extrathymic sites. In conclusion, mTEC specialization controls intrathymic iNKT cell development and function, and determines iNKT pool size in peripheral tissues.


Subject(s)
Cell Differentiation/immunology , Epithelial Cells/immunology , Natural Killer T-Cells/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/immunology , Cell Proliferation/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation/immunology , Lymphocyte Activation/immunology , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Lymphotoxin beta Receptor/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
18.
J Immunol ; 204(5): 1085-1090, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31969387

ABSTRACT

Lymphotoxin ß receptor (LTßR) signaling is crucial for lymphoid tissue organogenesis and immune homeostasis. To identify novel regulatory mechanisms for signaling, we implemented a two-step screen that uses coexpression analysis of human fibroblasts undergoing LTßR stimulation and affinity-purification mass spectrometry for the LTßR signaling protein TNFR-associated factor 3 (TRAF3). We identify Ewing sarcoma (EWS) protein as a novel LTßR signaling component that associates with TRAF3 but not with TNFR-associated factor 2 (TRAF2). The EWS:TRAF3 complex forms under unligated conditions that are disrupted following activation of the LTßR. We conclude that EWS limits expression of proinflammatory molecules, GM-CSF, and ERK-2, promoting immune homeostasis.


Subject(s)
Lymphotoxin beta Receptor/immunology , MAP Kinase Signaling System/immunology , Multiprotein Complexes/immunology , RNA-Binding Protein EWS/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , HEK293 Cells , Humans , Lymphotoxin beta Receptor/genetics , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Multiprotein Complexes/genetics , RNA-Binding Protein EWS/genetics , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/immunology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology
19.
Eur J Hum Genet ; 28(3): 313-323, 2020 03.
Article in English | MEDLINE | ID: mdl-31591516

ABSTRACT

Celiac disease (CeD) is a common immune-mediated disease of the small intestine that is triggered by exposure to dietary gluten. While the HLA locus plays a major role in disease susceptibility, 39 non-HLA loci were also identified in a study of 24,269 individuals. We now build on this earlier study by adding 4125 additional Caucasian samples including an Argentinian cohort. In doing so, we not only confirm the previous associations, we also identify two novel independent genome-wide significant associations at loci: 12p13.31 and 22q13.1. By applying a genomics approach and differential expression analysis in CeD intestinal biopsies, we prioritize potential causal genes at these novel loci, including LTBR, CYTH4, and RAC2. Nineteen prioritized causal genes are overlapping known drug targets. Pathway enrichment analysis and expression of these genes in CeD biopsies suggest that they have roles in regulating multiple pathways such as the tumor necrosis factor (TNF) mediated signaling pathway and positive regulation of I-κB kinase/NF-κB signaling.


Subject(s)
Celiac Disease/genetics , Genetic Loci , Polymorphism, Single Nucleotide , Argentina , Celiac Disease/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 22/genetics , Europe , Genome-Wide Association Study , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Intestinal Mucosa/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , RAC2 GTP-Binding Protein
20.
Sci Immunol ; 4(42)2019 12 20.
Article in English | MEDLINE | ID: mdl-31862865

ABSTRACT

Redundant mechanisms support immunoglobulin A (IgA) responses to intestinal antigens. These include multiple priming sites [mesenteric lymph nodes (MLNs), Peyer's patches, and isolated lymphoid follicles] and various cytokines that promote class switch to IgA, even in the absence of T cells. Despite these backup mechanisms, vaccination against enteric pathogens such as rotavirus has limited success in some populations. Genetic and environmental signals experienced during early life are known to influence mucosal immunity, yet the mechanisms for how these exposures operate remain unclear. Here, we used rotavirus infection to follow antigen-specific IgA responses through time and in different gut compartments. Using genetic and pharmacological approaches, we tested the role of the lymphotoxin (LT) pathway-known to support IgA responses-at different developmental stages. We found that LT-ß receptor (LTßR) signaling in early life programs intestinal IgA responses in adulthood by affecting antibody class switch recombination to IgA and subsequent generation of IgA antibody-secreting cells within an intact MLN. In addition, early-life LTßR signaling dictates the phenotype and function of MLN stromal cells to support IgA responses in the adult. Collectively, our studies uncover new mechanistic insights into how early-life LTßR signaling affects mucosal immune responses during adulthood.


Subject(s)
Immunoglobulin A/immunology , Lymph Nodes/immunology , Lymphotoxin beta Receptor/immunology , Lymphotoxin-alpha/immunology , Mesentery/immunology , Stromal Cells/immunology , Animals , Feces/microbiology , Female , Immunity, Mucosal , Lymph Nodes/cytology , Lymphotoxin beta Receptor/genetics , Lymphotoxin-alpha/genetics , Male , Mesentery/cytology , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...