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.
Cardiovasc Res ; 119(2): 506-519, 2023 03 31.
Article in English | MEDLINE | ID: mdl-35815623

ABSTRACT

AIMS: Growing evidence correlate the accrual of the sphingolipid ceramide in plasma and cardiac tissue with heart failure (HF). Regulation of sphingolipid metabolism in the heart and the pathological impact of its derangement remain poorly understood. Recently, we discovered that Nogo-B, a membrane protein of endoplasmic reticulum, abundant in the vascular wall, down-regulates the sphingolipid de novo biosynthesis via serine palmitoyltransferase (SPT), first and rate liming enzyme, to impact vascular functions and blood pressure. Nogo-A, a splice isoform of Nogo, is transiently expressed in cardiomyocyte (CM) following pressure overload. Cardiac Nogo is up-regulated in dilated and ischaemic cardiomyopathies in animals and humans. However, its biological function in the heart remains unknown. METHODS AND RESULTS: We discovered that Nogo-A is a negative regulator of SPT activity and refrains ceramide de novo biosynthesis in CM exposed to haemodynamic stress, hence limiting ceramide accrual. At 7 days following transverse aortic constriction (TAC), SPT activity was significantly up-regulated in CM lacking Nogo-A and correlated with ceramide accrual, particularly very long-chain ceramides, which are the most abundant in CM, resulting in the suppression of 'beneficial' autophagy. At 3 months post-TAC, mice lacking Nogo-A in CM showed worse pathological cardiac hypertrophy and dysfunction, with ca. 50% mortality rate. CONCLUSION: Mechanistically, Nogo-A refrains ceramides from accrual, therefore preserves the 'beneficial' autophagy, mitochondrial function, and metabolic gene expression, limiting the progression to HF under sustained stress.


Subject(s)
Heart Failure , Sphingolipids , Humans , Mice , Animals , Nogo Proteins/genetics , Nogo Proteins/metabolism , Sphingolipids/metabolism , Ceramides/metabolism , Heart Failure/genetics , Myocytes, Cardiac/metabolism
2.
Sci Immunol ; 5(45)2020 03 20.
Article in English | MEDLINE | ID: mdl-32198221

ABSTRACT

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


Subject(s)
Antibodies/immunology , Chemokine CCL2/immunology , Stromal Cells/immunology , Animals , Antibody Formation/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Sci Transl Med ; 10(454)2018 08 15.
Article in English | MEDLINE | ID: mdl-30111646

ABSTRACT

Photosensitivity, or skin sensitivity to ultraviolet radiation (UVR), is a feature of lupus erythematosus and other autoimmune and dermatologic conditions, but the mechanistic underpinnings are poorly understood. We identify a Langerhans cell (LC)-keratinocyte axis that limits UVR-induced keratinocyte apoptosis and skin injury via keratinocyte epidermal growth factor receptor (EGFR) stimulation. We show that the absence of LCs in Langerin-diphtheria toxin subunit A (DTA) mice leads to photosensitivity and that, in vitro, mouse and human LCs can directly protect keratinocytes from UVR-induced apoptosis. LCs express EGFR ligands and a disintegrin and metalloprotease 17 (ADAM17), the metalloprotease that activates EGFR ligands. Deletion of ADAM17 from LCs leads to photosensitivity, and UVR induces LC ADAM17 activation and generation of soluble active EGFR ligands, suggesting that LCs protect by providing activated EGFR ligands to keratinocytes. Photosensitive systemic lupus erythematosus (SLE) models and human SLE skin show reduced epidermal EGFR phosphorylation and LC defects, and a topical EGFR ligand reduces photosensitivity. Together, our data establish a direct tissue-protective function for LCs, reveal a mechanistic basis for photosensitivity, and suggest EGFR stimulation as a treatment for photosensitivity in lupus erythematosus and potentially other autoimmune and dermatologic conditions.


Subject(s)
Cytoprotection/radiation effects , Keratinocytes/cytology , Keratinocytes/radiation effects , Langerhans Cells/cytology , Langerhans Cells/radiation effects , Ultraviolet Rays , ADAM17 Protein/metabolism , Animals , Apoptosis/radiation effects , Disease Models, Animal , Epidermis/metabolism , Epidermis/radiation effects , ErbB Receptors/metabolism , Humans , Ligands , Lupus Erythematosus, Systemic/pathology , Mice, Inbred C57BL , Phosphorylation/radiation effects
4.
F1000Res ; 6: 196, 2017.
Article in English | MEDLINE | ID: mdl-28344775

ABSTRACT

Tertiary lymphoid organs are found at sites of chronic inflammation in autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis. These organized accumulations of T and B cells resemble secondary lymphoid organs and generate autoreactive effector cells. However, whether they contribute to disease pathogenesis or have protective functions is unclear. Here, we discuss how tertiary lymphoid organs can generate potentially pathogenic cells but may also limit the extent of the response and damage in autoimmune disease.

5.
J Clin Invest ; 126(11): 4331-4345, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27721238

ABSTRACT

Scleroderma is a group of skin-fibrosing diseases for which there are no effective treatments. A feature of the skin fibrosis typical of scleroderma is atrophy of the dermal white adipose tissue (DWAT). Adipose tissue contains adipose-derived mesenchymal stromal cells (ADSCs) that have regenerative and reparative functions; however, whether DWAT atrophy in fibrosis is accompanied by ADSC loss is poorly understood, as are the mechanisms that might maintain ADSC survival in fibrotic skin. Here, we have shown that DWAT ADSC numbers were reduced, likely because of cell death, in 2 murine models of scleroderma skin fibrosis. The remaining ADSCs showed a partial dependence on dendritic cells (DCs) for survival. Lymphotoxin ß (LTß) expression in DCs maintained ADSC survival in fibrotic skin by activating an LTß receptor/ß1 integrin (LTßR/ß1 integrin) pathway on ADSCs. Stimulation of LTßR augmented the engraftment of therapeutically injected ADSCs, which was associated with reductions in skin fibrosis and improved skin function. These findings provide insight into the effects of skin fibrosis on DWAT ADSCs, identify a DC-ADSC survival axis in fibrotic skin, and suggest an approach for improving mesenchymal stromal cell therapy in scleroderma and other diseases.


Subject(s)
Dendritic Cells/metabolism , Dermis/metabolism , Scleroderma, Diffuse/metabolism , Subcutaneous Fat/metabolism , Animals , Cell Survival/genetics , Dendritic Cells/pathology , Dermis/pathology , Disease Models, Animal , Female , Fibrosis , Integrin beta1/genetics , Integrin beta1/metabolism , Lymphotoxin-beta/genetics , Lymphotoxin-beta/metabolism , Mice , Mice, Knockout , Scleroderma, Diffuse/genetics , Scleroderma, Diffuse/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Subcutaneous Fat/pathology
6.
Trends Immunol ; 37(11): 764-777, 2016 11.
Article in English | MEDLINE | ID: mdl-27638128

ABSTRACT

During normal and pathologic immune responses, lymph nodes can swell considerably. The lymph node vascular-stromal compartment supports and regulates the developing immune responses and undergoes dynamic expansion and remodeling. Recent studies have shown that dendritic cells (DCs), best known for their antigen presentation roles, can directly regulate the vascular-stromal compartment, pointing to a new perspective on DCs as facilitators of lymphoid tissue function. Here, we review the phases of lymph node vascular-stromal growth and remodeling during immune responses, discuss the roles of DCs, and discuss how this understanding can potentially be used for developing novel therapeutic approaches.


Subject(s)
Antigen Presentation , Dendritic Cells/immunology , Immunity , Immunotherapy/methods , Lymph Nodes/immunology , Stromal Cells/physiology , Vascular Remodeling/immunology , Animals , Antigens/immunology , Antigens/metabolism , Cell Communication/immunology , Humans , Immunotherapy/trends , Peptides/immunology , Peptides/metabolism
7.
Immunity ; 42(4): 719-30, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25902483

ABSTRACT

Within secondary lymphoid tissues, stromal reticular cells support lymphocyte function, and targeting reticular cells is a potential strategy for controlling pathogenic lymphocytes in disease. However, the mechanisms that regulate reticular cell function are not well understood. Here we found that during an immune response in lymph nodes, dendritic cells (DCs) maintain reticular cell survival in multiple compartments. DC-derived lymphotoxin beta receptor (LTßR) ligands were critical mediators, and LTßR signaling on reticular cells mediated cell survival by modulating podoplanin (PDPN). PDPN modulated integrin-mediated cell adhesion, which maintained cell survival. This DC-stromal axis maintained lymphocyte survival and the ongoing immune response. Our findings provide insight into the functions of DCs, LTßR, and PDPN and delineate a DC-stromal axis that can potentially be targeted in autoimmune or lymphoproliferative diseases.


Subject(s)
Dendritic Cells/cytology , Lymph Nodes/cytology , Lymphotoxin beta Receptor/immunology , Membrane Glycoproteins/immunology , Stromal Cells/cytology , Animals , Cell Adhesion , Cell Survival/immunology , Dendritic Cells/immunology , Gene Expression Regulation , Immunophenotyping , Lymph Nodes/immunology , Lymphocyte Depletion , Lymphotoxin beta Receptor/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Stromal Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...