Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Genome Biol ; 23(1): 127, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35672799

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is an autoimmune condition of the central nervous system with a well-characterized genetic background. Prior analyses of MS genetics have identified broad enrichments across peripheral immune cells, yet the driver immune subsets are unclear. RESULTS: We utilize chromatin accessibility data across hematopoietic cells to identify cell type-specific enrichments of MS genetic signals. We find that CD4 T and B cells are independently enriched for MS genetics and further refine the driver subsets to Th17 and memory B cells, respectively. We replicate our findings in data from untreated and treated MS patients and find that immunomodulatory treatments suppress chromatin accessibility at driver cell types. Integration of statistical fine-mapping and chromatin interactions nominate numerous putative causal genes, illustrating complex interplay between shared and cell-specific genes. CONCLUSIONS: Overall, our study finds that open chromatin regions in CD4 T cells and B cells independently drive MS genetic signals. Our study highlights how careful integration of genetics and epigenetics can provide fine-scale insights into causal cell types and nominate new genes and pathways for disease.


Subject(s)
Multiple Sclerosis , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes , Chromatin , Humans , Immunity , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism
2.
Nat Commun ; 10(1): 4408, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31562318

ABSTRACT

Intestinal epithelial cells (IEC) have important functions in nutrient absorption, barrier integrity, regeneration, pathogen-sensing, and mucus secretion. Goblet cells are a specialized cell type of IEC that secrete Trefoil factor 3 (TFF3) to regulate mucus viscosity and wound healing, but whether TFF3-responsiveness requires a receptor is unclear. Here, we show that leucine rich repeat receptor and nogo-interacting protein 2 (LINGO2) is essential for TFF3-mediated functions. LINGO2 immunoprecipitates with TFF3, co-localizes with TFF3 on the cell membrane of IEC, and allows TFF3 to block apoptosis. We further show that TFF3-LINGO2 interactions disrupt EGFR-LINGO2 complexes resulting in enhanced EGFR signaling. Excessive basal EGFR activation in Lingo2 deficient mice increases disease severity during colitis and augments immunity against helminth infection. Conversely, TFF3 deficiency reduces helminth immunity. Thus, TFF3-LINGO2 interactions de-repress inhibitory LINGO2-EGFR complexes, allowing TFF3 to drive wound healing and immunity.


Subject(s)
Colitis/immunology , ErbB Receptors/immunology , Helminthiasis/immunology , Intestinal Mucosa/immunology , Membrane Proteins/metabolism , Nerve Tissue Proteins/immunology , Trefoil Factor-3/immunology , Animals , Cell Line, Tumor , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate , ErbB Receptors/genetics , ErbB Receptors/metabolism , Goblet Cells/immunology , Goblet Cells/metabolism , Goblet Cells/parasitology , HEK293 Cells , Helminthiasis/metabolism , Helminthiasis/parasitology , Helminths/immunology , Helminths/physiology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Membrane Proteins/genetics , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organophosphonates , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism , U937 Cells
3.
Mucosal Immunol ; 12(1): 64-76, 2019 01.
Article in English | MEDLINE | ID: mdl-30337651

ABSTRACT

Coordinated efforts between macrophages and epithelia are considered essential for wound healing, but the macrophage-derived molecules responsible for repair are poorly defined. This work demonstrates that lung macrophages rely upon Trefoil factor 2 to promote epithelial proliferation following damage caused by sterile wounding, Nippostrongylus brasiliensis or Bleomycin sulfate. Unexpectedly, the presence of T, B, or ILC populations was not essential for macrophage-driven repair. Instead, conditional deletion of TFF2 in myeloid-restricted CD11cCre TFF2 flox mice exacerbated lung pathology and reduced the proliferative expansion of CD45- EpCAM+ pro-SPC+ alveolar type 2 cells. TFF2 deficient macrophages had reduced expression of the Wnt genes Wnt4 and Wnt16 and reconstitution of hookworm-infected CD11cCre TFF2flox mice with rWnt4 and rWnt16 restored the proliferative defect in lung epithelia post-injury. These data reveal a previously unrecognized mechanism wherein lung myeloid phagocytes utilize a TFF2/Wnt axis as a mechanism that drives epithelial proliferation following lung injury.


Subject(s)
Lung Injury/immunology , Lung/immunology , Macrophages/physiology , Nippostrongylus/immunology , Respiratory Mucosa/physiology , Strongylida Infections/immunology , Trefoil Factor-2/metabolism , Animals , Bleomycin , CD11c Antigen/metabolism , Cell Communication , Cell Proliferation , Cells, Cultured , Humans , Lung/pathology , Lung Injury/chemically induced , Lung Injury/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Trefoil Factor-2/genetics , Wound Healing
4.
Am J Pathol ; 188(5): 1161-1170, 2018 05.
Article in English | MEDLINE | ID: mdl-29458008

ABSTRACT

Trefoil factors (TFFs) are small secreted proteins that regulate tissue integrity and repair at mucosal surfaces, particularly in the gastrointestinal tract. However, their relative contribution(s) to controlling baseline lung function or the extent of infection-induced lung injury are unknown issues. With the use of irradiation bone marrow chimeras, we found that TFF2 produced from both hematopoietic- and nonhematopoietic-derived cells is essential for host protection, proliferation of alveolar type 2 cells, and restoration of pulmonary gas exchange after infection with the hookworm parasite Nippostrongylus brasiliensis. In the absence of TFF2, lung epithelia were unable to proliferate and expressed reduced lung mRNA transcript levels for type 2 response-inducing IL-25 and IL-33 after infectious injury. Strikingly, even in the absence of infection or irradiation, TFF2 deficiency compromised lung structure and function, as characterized by distended alveoli and reduced blood oxygen levels relative to wild-type control mice. Taken together, we show a previously unappreciated role for TFF2, produced by either hematopoietic or nonhematopoietic sources, as a pro-proliferative factor for lung epithelial cells under steady-state and infectious injury conditions.


Subject(s)
Epithelial Cells/metabolism , Lung/metabolism , Pulmonary Alveoli/metabolism , Strongylida Infections/metabolism , Trefoil Factor-2/metabolism , Animals , Cell Proliferation , Epithelial Cells/parasitology , Epithelial Cells/pathology , Lung/parasitology , Lung/pathology , Mice , Mice, Transgenic , Nippostrongylus , Pulmonary Alveoli/parasitology , Pulmonary Alveoli/pathology , Strongylida Infections/immunology , Strongylida Infections/pathology
5.
J Immunol ; 196(11): 4632-40, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183598

ABSTRACT

How the metabolic demand of parasitism affects immune-mediated resistance is poorly understood. Immunity against parasitic helminths requires M2 cells and IL-13, secreted by CD4(+) Th2 and group 2 innate lymphoid cells (ILC2), but whether certain metabolic enzymes control disease outcome has not been addressed. This study demonstrates that AMP-activated protein kinase (AMPK), a key driver of cellular energy, regulates type 2 immunity and restricts lung injury following hookworm infection. Mice with a selective deficiency in the AMPK catalytic α1 subunit in alveolar macrophages and conventional dendritic cells produced less IL-13 and CCL17 and had impaired expansion of ILC2 in damaged lung tissue compared with wild-type controls. Defective type 2 responses were marked by increased intestinal worm burdens, exacerbated lung injury, and increased production of IL-12/23p40, which, when neutralized, restored IL-13 production and improved lung recovery. Taken together, these data indicate that defective AMPK activity in myeloid cells negatively impacts type 2 responses through increased IL-12/23p40 production. These data support an emerging concept that myeloid cells and ILC2 can coordinately regulate tissue damage at mucosal sites through mechanisms dependent on metabolic enzyme function.


Subject(s)
AMP-Activated Protein Kinases/immunology , Hookworm Infections/immunology , Immunity, Innate/immunology , Interleukin-12/immunology , Interleukin-23/immunology , Lung Injury/immunology , Myeloid Cells/immunology , AMP-Activated Protein Kinases/metabolism , Animals , Hookworm Infections/metabolism , Lung Injury/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...