Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cells ; 12(22)2023 11 20.
Article in English | MEDLINE | ID: mdl-37998397

ABSTRACT

Bardet-Biedl syndrome (BBS) is an archetypal ciliopathy caused by dysfunction of primary cilia. BBS affects multiple tissues, including the kidney, eye and hypothalamic satiety response. Understanding pan-tissue mechanisms of pathogenesis versus those which are tissue-specific, as well as gauging their associated inter-individual variation owing to genetic background and stochastic processes, is of paramount importance in syndromology. The BBSome is a membrane-trafficking and intraflagellar transport (IFT) adaptor protein complex formed by eight BBS proteins, including BBS1, which is the most commonly mutated gene in BBS. To investigate disease pathogenesis, we generated a series of clonal renal collecting duct IMCD3 cell lines carrying defined biallelic nonsense or frameshift mutations in Bbs1, as well as a panel of matching wild-type CRISPR control clones. Using a phenotypic screen and an unbiased multi-omics approach, we note significant clonal variability for all assays, emphasising the importance of analysing panels of genetically defined clones. Our results suggest that BBS1 is required for the suppression of mesenchymal cell identities as the IMCD3 cell passage number increases. This was associated with a failure to express epithelial cell markers and tight junction formation, which was variable amongst clones. Transcriptomic analysis of hypothalamic preparations from BBS mutant mice, as well as BBS patient fibroblasts, suggested that dysregulation of epithelial-to-mesenchymal transition (EMT) genes is a general predisposing feature of BBS across tissues. Collectively, this work suggests that the dynamic stability of the BBSome is essential for the suppression of mesenchymal cell identities as epithelial cells differentiate.


Subject(s)
Bardet-Biedl Syndrome , Humans , Mice , Animals , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/metabolism , Bardet-Biedl Syndrome/pathology , Mice, Knockout , Proteins/metabolism , Cilia/metabolism , Microtubule-Associated Proteins/metabolism
2.
Nat Immunol ; 24(1): 174-185, 2023 01.
Article in English | MEDLINE | ID: mdl-36564464

ABSTRACT

The kinase LCK and CD4/CD8 co-receptors are crucial components of the T cell antigen receptor (TCR) signaling machinery, leading to key T cell fate decisions. Despite decades of research, the roles of CD4-LCK and CD8-LCK interactions in TCR triggering in vivo remain unknown. In this study, we created animal models expressing endogenous levels of modified LCK to resolve whether and how co-receptor-bound LCK drives TCR signaling. We demonstrated that the role of LCK depends on the co-receptor to which it is bound. The CD8-bound LCK is largely dispensable for antiviral and antitumor activity of cytotoxic T cells in mice; however, it facilitates CD8+ T cell responses to suboptimal antigens in a kinase-dependent manner. By contrast, the CD4-bound LCK is required for efficient development and function of helper T cells via a kinase-independent stabilization of surface CD4. Overall, our findings reveal the role of co-receptor-bound LCK in T cell biology, show that CD4- and CD8-bound LCK drive T cell development and effector immune responses using qualitatively different mechanisms and identify the co-receptor-LCK interactions as promising targets for immunomodulation.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , T-Lymphocytes, Cytotoxic , Mice , Animals , T-Lymphocytes, Cytotoxic/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , CD4 Antigens , Signal Transduction , Receptors, Antigen, T-Cell/metabolism , CD8 Antigens/metabolism
3.
EMBO Rep ; 22(2): e50785, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33426789

ABSTRACT

Bardet-Biedl Syndrome (BBS) is a pleiotropic genetic disease caused by the dysfunction of primary cilia. The immune system of patients with ciliopathies has not been investigated. However, there are multiple indications that the impairment of the processes typically associated with cilia may have influence on the hematopoietic compartment and immunity. In this study, we analyze clinical data of BBS patients and corresponding mouse models carrying mutations in Bbs4 or Bbs18. We find that BBS patients have a higher prevalence of certain autoimmune diseases. Both BBS patients and animal models have altered red blood cell and platelet compartments, as well as elevated white blood cell levels. Some of the hematopoietic system alterations are associated with BBS-induced obesity. Moreover, we observe that the development and homeostasis of B cells in mice is regulated by the transport complex BBSome, whose dysfunction is a common cause of BBS. The BBSome limits canonical WNT signaling and increases CXCL12 levels in bone marrow stromal cells. Taken together, our study reveals a connection between a ciliopathy and dysregulated immune and hematopoietic systems.


Subject(s)
Autoimmune Diseases , Bardet-Biedl Syndrome , Hematopoiesis , Animals , Bardet-Biedl Syndrome/complications , Bardet-Biedl Syndrome/genetics , Cilia , Disease Models, Animal , Hematopoiesis/genetics , Humans , Mice , Microtubule-Associated Proteins/genetics , Mutation
4.
J Biol Chem ; 295(42): 14279-14290, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32759308

ABSTRACT

Bardet-Biedl syndrome (BBS) is a pleiotropic ciliopathy caused by dysfunction of primary cilia. More than half of BBS patients carry mutations in one of eight genes encoding for subunits of a protein complex, the BBSome, which mediates trafficking of ciliary cargoes. In this study, we elucidated the mechanisms of the BBSome assembly in living cells and how this process is spatially regulated. We generated a large library of human cell lines deficient in a particular BBSome subunit and expressing another subunit tagged with a fluorescent protein. We analyzed these cell lines utilizing biochemical assays, conventional and expansion microscopy, and quantitative fluorescence microscopy techniques: fluorescence recovery after photobleaching and fluorescence correlation spectroscopy. Our data revealed that the BBSome formation is a sequential process. We show that the pre-BBSome is nucleated by BBS4 and assembled at pericentriolar satellites, followed by the translocation of the BBSome into the ciliary base mediated by BBS1. Our results provide a framework for elucidating how BBS-causative mutations interfere with the biogenesis of the BBSome.


Subject(s)
Microtubule-Associated Proteins/metabolism , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/metabolism , Bardet-Biedl Syndrome/pathology , CRISPR-Cas Systems/genetics , Cell Line , Cilia/metabolism , Cytoplasm/metabolism , Fluorescence Recovery After Photobleaching , Gene Editing , Humans , Microscopy, Fluorescence , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Mutation , Protein Subunits/genetics , Protein Subunits/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...