Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Exp Med ; 221(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38411617

ABSTRACT

In vivo T cell screens are a powerful tool for elucidating complex mechanisms of immunity, yet there is a lack of consensus on the screen design parameters required for robust in vivo screens: gene library size, cell transfer quantity, and number of mice. Here, we describe the Framework for In vivo T cell Screens (FITS) to provide experimental and analytical guidelines to determine optimal parameters for diverse in vivo contexts. As a proof-of-concept, we used FITS to optimize the parameters for a CD8+ T cell screen in the B16-OVA tumor model. We also included unique molecular identifiers (UMIs) in our screens to (1) improve statistical power and (2) track T cell clonal dynamics for distinct gene knockouts (KOs) across multiple tissues. These findings provide an experimental and analytical framework for performing in vivo screens in immune cells and illustrate a case study for in vivo T cell screens with UMIs.


Subject(s)
CD8-Positive T-Lymphocytes , Animals , Mice , Gene Knockout Techniques
2.
Nat Biotechnol ; 42(4): 582-586, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37291427

ABSTRACT

Full-length RNA-sequencing methods using long-read technologies can capture complete transcript isoforms, but their throughput is limited. We introduce multiplexed arrays isoform sequencing (MAS-ISO-seq), a technique for programmably concatenating complementary DNAs (cDNAs) into molecules optimal for long-read sequencing, increasing the throughput >15-fold to nearly 40 million cDNA reads per run on the Sequel IIe sequencer. When applied to single-cell RNA sequencing of tumor-infiltrating T cells, MAS-ISO-seq demonstrated a 12- to 32-fold increase in the discovery of differentially spliced genes.


Subject(s)
High-Throughput Nucleotide Sequencing , RNA Isoforms , DNA, Complementary/genetics , RNA Isoforms/genetics , High-Throughput Nucleotide Sequencing/methods , Protein Isoforms/genetics , Sequence Analysis, RNA/methods , Transcriptome , Gene Expression Profiling/methods , RNA/genetics
3.
Science ; 381(6657): 508-514, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37535724

ABSTRACT

Proton leakage from organelles is a common signal for noncanonical light chain 3B (LC3B) lipidation and inflammasome activation, processes induced upon stimulator of interferon genes (STING) activation. On the basis of structural analysis, we hypothesized that human STING is a proton channel. Indeed, we found that STING activation induced a pH increase in the Golgi and that STING reconstituted in liposomes enabled transmembrane proton transport. Compound 53 (C53), a STING agonist that binds the putative channel interface, blocked STING-induced proton flux in the Golgi and in liposomes. STING-induced LC3B lipidation and inflammasome activation were also inhibited by C53, suggesting that STING's channel activity is critical for these two processes. Thus, STING's interferon-induction function can be decoupled from its roles in LC3B lipidation and inflammasome activation.


Subject(s)
Ion Channels , Membrane Proteins , Protons , Humans , Golgi Apparatus/metabolism , Hydrogen-Ion Concentration , Inflammasomes/metabolism , Ion Channels/agonists , Ion Channels/chemistry , Ion Channels/metabolism , Liposomes , Membrane Proteins/agonists , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Protein Domains , HEK293 Cells
4.
Nat Commun ; 14(1): 611, 2023 02 04.
Article in English | MEDLINE | ID: mdl-36739287

ABSTRACT

Stimulator of interferon genes (STING) is an intracellular sensor of cyclic di-nucleotides involved in the innate immune response against pathogen- or self-derived DNA. STING trafficking is tightly linked to its function, and its dysregulation can lead to disease. Here, we systematically characterize genes regulating STING trafficking and examine their impact on STING-mediated responses. Using proximity-ligation proteomics and genetic screens, we demonstrate that an endosomal sorting complex required for transport (ESCRT) complex containing HGS, VPS37A and UBAP1 promotes STING degradation, thereby terminating STING-mediated signaling. Mechanistically, STING oligomerization increases its ubiquitination by UBE2N, forming a platform for ESCRT recruitment at the endosome that terminates STING signaling via sorting in the lysosome. Finally, we show that expression of a UBAP1 mutant identified in patients with hereditary spastic paraplegia and associated with disrupted ESCRT function, increases steady-state STING-dependent type I IFN responses in healthy primary monocyte-derived dendritic cells and fibroblasts. Based on these findings, we propose that STING is subject to a tonic degradative flux and that the ESCRT complex acts as a homeostatic regulator of STING signaling.


Subject(s)
Membrane Proteins , Nucleotides, Cyclic , Humans , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Immunity, Innate , Membrane Proteins/metabolism , Nucleotides, Cyclic/pharmacology
5.
Am J Hum Genet ; 109(4): 601-617, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35395208

ABSTRACT

Neurodevelopmental disorders are highly heterogenous conditions resulting from abnormalities of brain architecture and/or function. FBXW7 (F-box and WD-repeat-domain-containing 7), a recognized developmental regulator and tumor suppressor, has been shown to regulate cell-cycle progression and cell growth and survival by targeting substrates including CYCLIN E1/2 and NOTCH for degradation via the ubiquitin proteasome system. We used a genotype-first approach and global data-sharing platforms to identify 35 individuals harboring de novo and inherited FBXW7 germline monoallelic chromosomal deletions and nonsense, frameshift, splice-site, and missense variants associated with a neurodevelopmental syndrome. The FBXW7 neurodevelopmental syndrome is distinguished by global developmental delay, borderline to severe intellectual disability, hypotonia, and gastrointestinal issues. Brain imaging detailed variable underlying structural abnormalities affecting the cerebellum, corpus collosum, and white matter. A crystal-structure model of FBXW7 predicted that missense variants were clustered at the substrate-binding surface of the WD40 domain and that these might reduce FBXW7 substrate binding affinity. Expression of recombinant FBXW7 missense variants in cultured cells demonstrated impaired CYCLIN E1 and CYCLIN E2 turnover. Pan-neuronal knockdown of the Drosophila ortholog, archipelago, impaired learning and neuronal function. Collectively, the data presented herein provide compelling evidence of an F-Box protein-related, phenotypically variable neurodevelopmental disorder associated with monoallelic variants in FBXW7.


Subject(s)
F-Box-WD Repeat-Containing Protein 7 , Neurodevelopmental Disorders , Ubiquitination , F-Box-WD Repeat-Containing Protein 7/chemistry , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Germ Cells , Germ-Line Mutation , Humans , Neurodevelopmental Disorders/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
6.
Ann Clin Transl Neurol ; 8(3): 716-722, 2021 03.
Article in English | MEDLINE | ID: mdl-33497533

ABSTRACT

We report two siblings with intractable epilepsy, developmental regression, and progressive cerebellar atrophy due to biallelic variants in the gene CAD. For the affected girl, uridine started at age 5 resulted in dramatic improvements in seizure control and development, cessation of cerebellar atrophy, and resolution of hematological abnormalities. Her older brother had a more severe course and only modest response to uridine started at 14 years old. Treatment of this progressive condition via uridine supplementation provides an example of precision diagnosis and treatment using clear outcome measures and biomarkers to monitor efficacy.


Subject(s)
Aspartate Carbamoyltransferase/genetics , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/genetics , Dihydroorotase/genetics , Drug Resistant Epilepsy/drug therapy , Drug Resistant Epilepsy/genetics , Uridine/pharmacology , Atrophy/pathology , Cerebellar Diseases/drug therapy , Cerebellar Diseases/genetics , Cerebellar Diseases/pathology , Child , Child, Preschool , Developmental Disabilities/drug therapy , Developmental Disabilities/genetics , Disease Progression , Female , Humans , Male , Pedigree , Siblings , Uridine/administration & dosage
7.
Pediatr Rheumatol Online J ; 17(1): 7, 2019 Feb 14.
Article in English | MEDLINE | ID: mdl-30764840

ABSTRACT

BACKGROUND: Hemophagocytic lymphohistiocytosis (HLH) and macrophage activation syndrome (MAS) were historically thought to be distinct entities, often managed in isolation. In fact, these conditions are closely related. A collaborative approach, which incorporates expertise from subspecialties that previously treated HLH/MAS independently, is needed. We leveraged quality improvement (QI) techniques in the form of an Evidence-Based Guideline (EBG) to build consensus across disciplines on the diagnosis and treatment of HLH/MAS. METHODS: A multidisciplinary work group was convened that met monthly to develop the HLH/MAS EBG. Literature review and expert opinion were used to develop a management strategy for HLH/MAS. The EBG was implemented, and quality metrics were selected to monitor outcomes. RESULTS: An HLH/MAS clinical team was formed with representatives from subspecialties involved in the care of patients with HLH/MAS. Broad entry criteria for the HLH/MAS EBG were established and included fever and ferritin ≥500 ng/mL. The rheumatology team was identified as the "gate-keeper," charged with overseeing the diagnostic evaluation recommended in the EBG. First-line medications were recommended based on the acuity of illness and risk of concurrent infection. Quality metrics to be tracked prospectively based on time to initiation of treatment and clinical response were selected. CONCLUSION: HLH/MAS are increasingly considered to be a spectrum of related conditions, and joint management across subspecialties could improve patient outcomes. Our experience in creating a multidisciplinary approach to HLH/MAS management can serve as a model for care at other institutions.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/diagnosis , Macrophage Activation Syndrome/diagnosis , Algorithms , Consensus , Cytokines/blood , Diagnosis, Differential , Evidence-Based Medicine/methods , Humans , Lymphohistiocytosis, Hemophagocytic/therapy , Macrophage Activation Syndrome/therapy , Practice Guidelines as Topic , Quality Improvement
8.
J Exp Med ; 212(10): 1663-77, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26371186

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is an X-linked immunodeficiency disorder frequently associated with systemic autoimmunity, including autoantibody-mediated cytopenias. WAS protein (WASp)-deficient B cells have increased B cell receptor (BCR) and Toll-like receptor (TLR) signaling, suggesting that these pathways might impact establishment of the mature, naive BCR repertoire. To directly investigate this possibility, we evaluated naive B cell specificity and composition in WASp-deficient mice and WAS subjects (n = 12). High-throughput sequencing and single-cell cloning analysis of the BCR repertoire revealed altered heavy chain usage and enrichment for low-affinity self-reactive specificities in murine marginal zone and human naive B cells. Although negative selection mechanisms including deletion, anergy, and receptor editing were relatively unperturbed, WASp-deficient transitional B cells showed enhanced proliferation in vivo mediated by antigen- and Myd88-dependent signals. Finally, using both BCR sequencing and cell surface analysis with a monoclonal antibody recognizing an intrinsically autoreactive heavy chain, we show enrichment in self-reactive cells specifically at the transitional to naive mature B cell stage in WAS subjects. Our combined data support a model wherein modest alterations in B cell-intrinsic, BCR, and TLR signals in WAS, and likely other autoimmune disorders, are sufficient to alter B cell tolerance via positive selection of self-reactive transitional B cells.


Subject(s)
B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Receptors, Antigen, B-Cell/metabolism , Toll-Like Receptors/metabolism , Wiskott-Aldrich Syndrome/metabolism , Adolescent , Adult , Animals , B-Cell Activating Factor/blood , B-Lymphocytes/immunology , Case-Control Studies , Child , Child, Preschool , Cytoprotection , High-Throughput Nucleotide Sequencing , Humans , Infant , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/metabolism , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism , Young Adult
9.
Eur J Immunol ; 45(10): 2773-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26256668

ABSTRACT

Patients with Wiskott-Aldrich syndrome (WAS) exhibit prominent defects in splenic marginal zone (MZ), resulting in abnormal T-cell-independent antibody responses and increased bacterial infections. B-cell-intrinsic deletion of the affected gene WAS protein (WASp) markedly reduces splenic MZ B cells, without impacting the rate of MZ B-cell development, suggesting that abnormal B-cell retention within the MZ accounts for MZ defects in WAS. Since WASp regulates integrin-dependent actin cytoskeletal rearrangement, we previously hypothesized that defective B-cell integrin function promotes MZ depletion. In contrast, we now report that B-cell-intrinsic deletion of the TLR signaling adaptor MyD88 is sufficient to restore the MZ in WAS. We further identify TLR7, an endosomal single-stranded RNA (ssRNA) receptor, as the MyD88-dependent receptor responsible for WAS MZ depletion. These findings implicate spontaneous activation of MZ B cells by ssRNA-containing self-ligands (likely derived from circulating apoptotic material) as the mechanism underlying MZ depletion in WAS. Together, these data suggest a previously unappreciated role for B-cell intrinsic TLR signals in MZ homeostasis, of relevance to both pathogen responses and to the development of systemic autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Membrane Glycoproteins/immunology , Signal Transduction/immunology , Spleen/immunology , Toll-Like Receptor 7/immunology , Wiskott-Aldrich Syndrome/immunology , Animals , B-Lymphocytes/pathology , Cytoskeleton/genetics , Cytoskeleton/immunology , Disease Models, Animal , Integrins/genetics , Integrins/immunology , Lymphocyte Depletion , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Signal Transduction/genetics , Spleen/pathology , Toll-Like Receptor 7/genetics , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/pathology
10.
J Immunol ; 193(7): 3492-502, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25172502

ABSTRACT

Control of peripheral B cell development and homeostasis depends critically on coordinate signals received through the BAFFRs and BCRs. The extent to which other signals contribute to this process, however, remains undefined. We present data indicating that CD4(+) T cells directly influence naive B cell development via CD40 signaling. Loss of CD4(+) T cells or CD40-CD40L interaction leads to reduced B cell homeostatic proliferation and hindered B cell reconstitution posttransplantation. Furthermore, we demonstrate that in the absence of CD40 signals, these events are modulated by BCR self-reactivity. Strikingly, murine models lacking CD40 reveal a broadly altered BCR specificity and limited diversity by both single-cell cloning and high-throughput sequencing techniques. Collectively, our results imply that any setting of T cell lymphopenia or reduced CD40 function, including B cell recovery following transplantation, will impact the naive B cell repertoire.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , Homeostasis/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Animals , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/pathology , CD40 Antigens/genetics , Homeostasis/genetics , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Knockout , Receptors, Antigen, B-Cell/genetics , Signal Transduction/genetics
11.
J Immunol ; 192(10): 4525-32, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24711620

ABSTRACT

Systemic lupus erythematosus is a multisystem autoimmune disease characterized by autoantibodies targeting nucleic acid-associated Ags. The endosomal TLRs TLR7 and TLR9 are critical for generation of Abs targeting RNA- or DNA-associated Ags, respectively. In murine lupus models, deletion of TLR7 limits autoimmune inflammation, whereas deletion of TLR9 exacerbates disease. Whether B cell or myeloid TLR7/TLR9 signaling is responsible for these effects has not been fully addressed. In this study, we use a chimeric strategy to evaluate the effect of B cell-intrinsic deletion of TLR7 versus TLR9 in parallel lupus models. We demonstrate that B cell-intrinsic TLR7 deletion prevents RNA-associated Ab formation, decreases production of class-switched Abs targeting nonnuclear Ags, and limits systemic autoimmunity. In contrast, B cell-intrinsic TLR9 deletion results in decreased DNA-reactive Ab, but increased Abs targeting a broad range of systemic autoantigens. Further, we demonstrate that B cell-intrinsic TLR9 deletion results in increased systemic inflammation and immune complex glomerulonephritis, despite intact TLR signaling within the myeloid compartment. These data stress the critical importance of dysregulated B cell-intrinsic TLR signaling in the pathogenesis of systemic lupus erythematosus.


Subject(s)
Antibodies, Antinuclear/immunology , B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/immunology , Signal Transduction/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/immunology , Animals , Antibodies, Antinuclear/genetics , B-Lymphocytes/pathology , Disease Models, Animal , Inflammation/genetics , Inflammation/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , RNA/genetics , RNA/immunology , Signal Transduction/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 9/genetics
12.
Nat Rev Immunol ; 12(4): 282-94, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22421786

ABSTRACT

Unlike other immune cells, B cells express both an antigen-specific B cell receptor (BCR) and Toll-like receptors (TLRs). Dual BCR and TLR engagement can fine-tune functional B cell responses, directly linking cell-intrinsic innate and adaptive immune programmes. Although most data regarding B cell-specific functions of the TLR signalling pathway have been obtained in mice, the discovery of patients with a deficiency in this pathway has recently provided an insight into human B cell responses. Here, we highlight the importance of the integration of signalling pathways downstream of BCRs and TLRs in modulating B cell function, focusing when possible on B cell-intrinsic roles.


Subject(s)
B-Lymphocytes/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Animals , Humans
13.
J Exp Med ; 208(10): 2033-42, 2011 Sep 26.
Article in English | MEDLINE | ID: mdl-21875954

ABSTRACT

Patients with the immunodeficiency Wiskott-Aldrich syndrome (WAS) frequently develop systemic autoimmunity. Here, we demonstrate that mutation of the WAS gene results in B cells that are hyperresponsive to B cell receptor and Toll-like receptor (TLR) signals in vitro, thereby promoting a B cell-intrinsic break in tolerance. Whereas this defect leads to autoantibody production in WAS protein-deficient (WASp(-/-)) mice without overt disease, chimeric mice in which only the B cell lineage lacks WASp exhibit severe autoimmunity characterized by spontaneous germinal center formation, class-switched autoantibodies, renal histopathology, and early mortality. Both T cell help and B cell-intrinsic TLR engagement play important roles in promoting disease in this model, as depletion with anti-CD4 antibodies or generation of chimeric mice with B cells deficient in both WASp and MyD88 prevented development of autoimmune disease. These data highlight the potentially harmful role for cell-intrinsic loss of B cell tolerance in the setting of normal T cell function, and may explain why WAS patients with mixed chimerism after stem cell transplantation often develop severe humoral autoimmunity.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , Wiskott-Aldrich Syndrome Protein/deficiency , Animals , Antibodies, Antinuclear/immunology , Autoantibodies/immunology , B-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/immunology , Chimera , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Receptors, Antigen, B-Cell/immunology , Toll-Like Receptors/immunology , Wiskott-Aldrich Syndrome Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...