Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(1): 186-199, 2023 01.
Article in English | MEDLINE | ID: mdl-36536106

ABSTRACT

Most studies of adaptive immunity to SARS-CoV-2 infection focus on peripheral blood, which may not fully reflect immune responses at the site of infection. Using samples from 110 children undergoing tonsillectomy and adenoidectomy during the COVID-19 pandemic, we identified 24 samples with evidence of previous SARS-CoV-2 infection, including neutralizing antibodies in serum and SARS-CoV-2-specific germinal center and memory B cells in the tonsils and adenoids. Single-cell B cell receptor (BCR) sequencing indicated virus-specific BCRs were class-switched and somatically hypermutated, with overlapping clones in the two tissues. Expanded T cell clonotypes were found in tonsils, adenoids and blood post-COVID-19, some with CDR3 sequences identical to previously reported SARS-CoV-2-reactive T cell receptors (TCRs). Pharyngeal tissues from COVID-19-convalescent children showed persistent expansion of germinal center and antiviral lymphocyte populations associated with interferon (IFN)-γ-type responses, particularly in the adenoids, and viral RNA in both tissues. Our results provide evidence for persistent tissue-specific immunity to SARS-CoV-2 in the upper respiratory tract of children after infection.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Child , Pandemics , Adaptive Immunity , Palatine Tonsil , Antibodies, Viral
2.
Nat Immunol ; 20(7): 890-901, 2019 07.
Article in English | MEDLINE | ID: mdl-31209400

ABSTRACT

Progenitor-like CD8+ T cells mediate long-term immunity to chronic infection and cancer and respond potently to immune checkpoint blockade. These cells share transcriptional regulators with memory precursor cells, including T cell-specific transcription factor 1 (TCF1), but it is unclear whether they adopt distinct programs to adapt to the immunosuppressive environment. By comparing the single-cell transcriptomes and epigenetic profiles of CD8+ T cells responding to acute and chronic viral infections, we found that progenitor-like CD8+ T cells became distinct from memory precursor cells before the peak of the T cell response. We discovered a coexpression gene module containing Tox that exhibited higher transcriptional activity associated with more abundant active histone marks in progenitor-like cells than memory precursor cells. Moreover, thymocyte selection-associated high mobility group box protein TOX (TOX) promoted the persistence of antiviral CD8+ T cells and was required for the programming of progenitor-like CD8+ T cells. Thus, long-term CD8+ T cell immunity to chronic viral infection requires unique transcriptional and epigenetic programs associated with the transcription factor TOX.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , Infections/etiology , Single-Cell Analysis , Animals , Biomarkers , Chromatin Immunoprecipitation , Epigenesis, Genetic , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Homeodomain Proteins/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunologic Memory , Infections/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Time Factors , Transcriptome
3.
BMC Bioinformatics ; 24(1): 102, 2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36941541

ABSTRACT

BACKGROUND: Epigenetic modification of chromatin plays a pivotal role in regulating gene expression during cell differentiation. The scale and complexity of epigenetic data pose significant challenges for biologists to identify the regulatory events controlling cell differentiation. RESULTS: To reduce the complexity, we developed a package, called Snapshot, for clustering and visualizing candidate cis-regulatory elements (cCREs) based on their epigenetic signals during cell differentiation. This package first introduces a binarized indexing strategy for clustering the cCREs. It then provides a series of easily interpretable figures for visualizing the signal and epigenetic state patterns of the cCREs clusters during the cell differentiation. It can also use different hierarchies of cell types to highlight the epigenetic history specific to any particular cell lineage. We demonstrate the utility of Snapshot using data from a consortium project for ValIdated Systematic IntegratiON (VISION) of epigenomic data in hematopoiesis. CONCLUSION: The package Snapshot can identify all distinct clusters of genomic locations with unique epigenetic signal patterns during cell differentiation. It outperforms other methods in terms of interpreting and reproducing the identified cCREs clusters. The package of Snapshot is available at GitHub: https://github.com/guanjue/Snapshot .


Subject(s)
Chromatin , Epigenomics , Epigenomics/methods , Cell Differentiation/genetics , Epigenesis, Genetic , Cluster Analysis
4.
Immunity ; 31(4): 587-97, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19818650

ABSTRACT

T helper 17 (Th17) cells play major roles in autoimmunity and bacterial infections, yet how T cell receptor (TCR) signaling affects Th17 cell differentiation is relatively unknown. We demonstrate that CD4(+) T cells lacking Itk, a tyrosine kinase required for full TCR-induced phospholipase C-gamma (PLC-gamma1) activation, exhibit decreased interleukin-17A (IL-17A) expression in vitro and in vivo, despite relatively normal expression of retinoic acid receptor-related orphan receptor-gammaT (ROR-gammaT) and IL-17F. IL-17A expression was rescued by pharmacologically induced Ca(2+) influx or constitutively activated nuclear factor of activated T cells (NFAT). Conversely, decreased TCR stimulation or calcineurin inhibition preferentially reduced IL-17A expression. We further found that the promoter of Il17a but not Il17f has a conserved NFAT binding site that bound NFATc1 in wild-type but not Itk-deficient cells, even though both exhibited open chromatin conformations. Finally, Itk(-/-) mice also showed differential regulation of IL-17A and IL-17F in vivo. Our results suggest that Itk specifically couples TCR signaling to Il17a expression and the differential regulation of Th17 cell cytokines through NFATc1.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Interleukin-17/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Calcium/immunology , Calcium/metabolism , Cytokines/metabolism , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3 , Phospholipase C gamma/immunology , Phospholipase C gamma/metabolism , Promoter Regions, Genetic/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Retinoic Acid/immunology , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/immunology , Receptors, Thyroid Hormone/metabolism , Signal Transduction/immunology
5.
Haematologica ; 102(4): 656-665, 2017 04.
Article in English | MEDLINE | ID: mdl-28011901

ABSTRACT

Gaucher disease, the inherited deficiency of lysosomal glucocerebrosidase, is characterized by the presence of glucosylceramide-laden macrophages resulting from impaired digestion of aged erythrocytes or apoptotic leukocytes. Studies of macrophages from patients with type 1 Gaucher disease with genotypes N370S/N370S, N370S/L444P or N370S/c.84dupG revealed that Gaucher macrophages have impaired efferocytosis resulting from reduced levels of p67phox and Rab7. The decreased Rab7 expression leads to impaired fusion of phagosomes with lysosomes. Moreover, there is defective translocation of p67phox to phagosomes, resulting in reduced intracellular production of reactive oxygen species. These factors contribute to defective deposition and clearance of apoptotic cells in phagolysosomes, which may have an impact on the inflammatory response and contribute to the organomegaly and inflammation seen in patients with Gaucher disease.


Subject(s)
Gaucher Disease/genetics , Gaucher Disease/immunology , Macrophages/immunology , Macrophages/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Biomarkers , Cytophagocytosis/genetics , Cytophagocytosis/immunology , Genotype , Glucosylceramidase/genetics , Humans , Immunohistochemistry , Mutation , Phagosomes/metabolism , Reactive Oxygen Species/metabolism , Respiratory Burst/genetics , Respiratory Burst/immunology
6.
J Med Genet ; 53(5): 318-29, 2016 05.
Article in English | MEDLINE | ID: mdl-27095636

ABSTRACT

BACKGROUND: Laminins are heterotrimeric complexes, consisting of α, ß and γ subunits that form a major component of basement membranes and extracellular matrix. Laminin complexes have different, but often overlapping, distributions and functions. METHODS: Under our clinical protocol, NCT00068224, we have performed extensive clinical and neuropsychiatric phenotyping, neuroimaging and molecular analysis in patients with laminin α1 (LAMA1)-associated lamininopathy. We investigated the consequence of mutations in LAMA1 using patient-derived fibroblasts and neuronal cells derived from neuronal stem cells. RESULTS: In this paper we describe individuals with biallelic mutations in LAMA1, all of whom had the cerebellar dysplasia, myopia and retinal dystrophy, in addition to obsessive compulsive traits, tics and anxiety. Patient-derived fibroblasts have impaired adhesion, reduced migration, abnormal morphology and increased apoptosis due to impaired activation of Cdc42, a member of the Rho family of GTPases that is involved in cytoskeletal dynamics. LAMA1 knockdown in human neuronal cells also showed abnormal morphology and filopodia formation, supporting the importance of LAMA1 in neuronal migration, and marking these cells potentially useful tools for disease modelling and therapeutic target discovery. CONCLUSION: This paper broadens the phenotypes associated with LAMA1 mutations. We demonstrate that LAMA1 deficiency can lead to alteration in cytoskeletal dynamics, which may invariably lead to alteration in dendrite growth and axonal formation. Estimation of disease prevalence based on population studies in LAMA1 reveals a prevalence of 1-20 in 1 000 000. TRIAL REGISTRATION NUMBER: NCT00068224.


Subject(s)
Cerebellar Diseases/metabolism , Laminin/genetics , Mutation , Myopia/metabolism , Obsessive-Compulsive Disorder/metabolism , Adult , Cell Adhesion , Cell Movement , Cerebellar Diseases/genetics , Cerebellar Diseases/physiopathology , Child , Female , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Male , Myopia/genetics , Myopia/physiopathology , Neurons/metabolism , Neurons/physiology , Obsessive-Compulsive Disorder/genetics , Obsessive-Compulsive Disorder/physiopathology , Pedigree , Retinal Dystrophies/genetics , Retinal Dystrophies/metabolism , Retinal Dystrophies/physiopathology , Syndrome , Tic Disorders/genetics , Tic Disorders/metabolism , Tic Disorders/physiopathology , Young Adult , cdc42 GTP-Binding Protein
7.
Hum Genet ; 134(7): 775-87, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25939664

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is a pediatric myeloproliferative neoplasm that arises from malignant transformation of the stem cell compartment and results in increased production of myeloid cells. Somatic and germline variants in CBL (Casitas B-lineage lymphoma proto-oncogene) have been associated with JMML. We report an incompletely penetrant CBL Y371C mutation discovered by whole-exome sequencing in three individuals with JMML in a large pedigree with 35 years of follow-up. The Y371 residue is highly evolutionarily conserved among CBL orthologs and paralogs. In silico bioinformatics prediction programs suggested that the Y371C mutation is highly deleterious. Protein structural modeling revealed that the Y371C mutation abrogated the ability of the CBL protein to adopt a conformation that is required for ubiquitination. Clinically, the three mutation-positive JMML individuals exhibited variable clinical courses; in two out of three, primary hematologic abnormalities persisted into adulthood with minimal clinical symptoms. The penetrance of the CBL Y371C mutation was 30% for JMML and 40% for all leukemia. Of the 8 mutation carriers in the family with available photographs, only one had significant dysmorphic features; we found no evidence of a clinical phenotype consistent with a "CBL syndrome". Although CBL Y371C has been previously reported in familial JMML, we are the first group to follow a complete pedigree harboring this mutation for an extended period, revealing additional information about this variant's penetrance, function and natural history.


Subject(s)
Germ-Line Mutation , Leukemia, Myelomonocytic, Juvenile/genetics , Mutation, Missense , Pedigree , Proto-Oncogene Proteins c-cbl/genetics , Ubiquitination/genetics , Adolescent , Adult , Child , Child, Preschool , Exome , Female , Follow-Up Studies , Humans , Infant , Male , Models, Molecular , Penetrance , Protein Structure, Tertiary , Proto-Oncogene Mas , Proto-Oncogene Proteins c-cbl/chemistry
8.
Blood ; 119(6): 1511-21, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22160378

ABSTRACT

KIT mutations are the most common secondary mutations in inv(16) acute myeloid leukemia (AML) patients and are associated with poor prognosis. It is therefore important to verify that KIT mutations cooperate with CBFB-MYH11, the fusion gene generated by inv(16), for leukemogenesis. Here, we transduced wild-type and conditional Cbfb-MYH11 knockin (KI) mouse bone marrow (BM) cells with KIT D816V/Y mutations. KIT transduction caused massive BM Lin(-) cell death and fewer colonies in culture that were less severe in the KI cells. D816Y KIT but not wild-type KIT enhanced proliferation in Lin(-) cells and led to more mixed lineage colonies from transduced KI BM cells. Importantly, 60% and 80% of mice transplanted with KI BM cells expressing D816V or D816Y KIT, respectively, died from leukemia within 9 months, whereas no control mice died. Results from limiting dilution transplantations indicate higher frequencies of leukemia-initiating cells in the leukemia expressing mutated KIT. Signaling pathway analysis revealed that p44/42 MAPK and Stat3, but not AKT and Stat5, were strongly phosphorylated in the leukemia cells. Finally, leukemia cells carrying KIT D816 mutations were sensitive to the kinase inhibitor PKC412. Our data provide clear evidence for cooperation between mutated KIT and CBFB-MYH11 during leukemogenesis.


Subject(s)
Leukemia/genetics , Mutation , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-kit/genetics , Animals , Blotting, Western , Bone Marrow Transplantation , Cell Survival/drug effects , Cells, Cultured , Disease Progression , Female , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukemia/metabolism , Leukemia/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
9.
bioRxiv ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38529501

ABSTRACT

Inducible pluripotent stem cells (iPSCs) derived from patient samples have significantly enhanced our ability to model neurological diseases. Comparative studies of dopaminergic (DA) neurons differentiated from iPSCs derived from siblings with Gaucher disease discordant for parkinsonism provides a valuable avenue to explore genetic modifiers contributing to GBA1-associated parkinsonism in disease-relevant cells. However, such studies are often complicated by the inherent heterogeneity in differentiation efficiency among iPSC lines derived from different individuals. To address this technical challenge, we devised a selection strategy to enrich dopaminergic (DA) neurons expressing tyrosine hydroxylase (TH). A neomycin resistance gene (neo) was inserted at the C-terminus of the TH gene following a T2A self-cleavage peptide, placing its expression under the control of the TH promoter. This allows for TH+ DA neuron enrichment through geneticin selection. This method enabled us to generate comparable, high-purity DA neuron cultures from iPSC lines derived from three sisters that we followed for over a decade: one sibling is a healthy individual, and the other two have Gaucher disease (GD) with GBA1 genotype N370S/c.203delC+R257X (p.N409S/c.203delC+p.R296X). Notably, the younger sister with GD later developed Parkinson disease (PD). A comprehensive analysis of these high-purity DA neurons revealed that although GD DA neurons exhibited decreased levels of glucocerebrosidase (GCase), there was no substantial difference in GCase protein levels or lipid substrate accumulation between DA neurons from the GD and GD/PD sisters, suggesting that the PD discordance is related to of other genetic modifiers.

10.
Mol Genet Metab ; 110(1-2): 188-90, 2013.
Article in English | MEDLINE | ID: mdl-23850077

ABSTRACT

Mutations in individuals with the lysosomal storage disorder Niemann-Pick disease, type C1 (NPC1) are heterogeneous, not localized to specific protein domains, and not correlated to time of onset or disease severity. We demonstrate direct correlation of the time of neurological symptom onset with the severity of lysosomal defects in NPC1 patient-derived fibroblasts. This is a novel assay for NPC1 individuals that may be predictive of NPC1 disease progression and broadly applicable to other lysosomal disorders.


Subject(s)
Lysosomal Storage Diseases/genetics , Lysosomes/metabolism , Membrane Glycoproteins/genetics , Niemann-Pick Disease, Type C/genetics , Adolescent , Adult , Biological Transport/genetics , Cells, Cultured , Child , Child, Preschool , Disease Progression , Female , Fibroblasts , Humans , Infant , Infant, Newborn , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Lysosomes/genetics , Lysosomes/pathology , Male , Membrane Glycoproteins/metabolism , Mutation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Protein Structure, Tertiary
11.
Blood ; 117(24): 6608-11, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21515824

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is an inherited immunodeficiency characterized by high incidence of autoantibody-mediated autoimmune complications. Such a feature has been associated with defective suppressor activity of WAS protein-deficient, naturally occurring CD4(+)CD25(+)Foxp3(+) regulatory T cells on responder T cells. However, it remains to be established whether the altered B-cell tolerance reported in WAS patients and Was knockout (WKO) mice is secondary to abnormalities in the direct suppression of B-cell function by nTreg cells or to impaired regulation of T-helper function. Because activated nTreg cells are known to induce granzyme B-mediated B-cell killing, we decided to evaluate the regulatory capabilities of WKO nTregs on B lymphocytes. We found that preactivated WKO nTreg cells failed to effectively suppress B-cell proliferation and that such a defect was associated with reduced killing of B cells and significantly decreased degranulation of granzyme B. Altogether, these results provide additional mechanistic insights into the loss of immune tolerance in WAS.


Subject(s)
B-Lymphocytes/physiology , Cell Proliferation , T-Lymphocytes, Regulatory/physiology , Wiskott-Aldrich Syndrome Protein/genetics , Animals , B-Lymphocytes/metabolism , Cell Death/genetics , Cell Death/immunology , Cell Degranulation/genetics , Cell Degranulation/immunology , Cells, Cultured , Down-Regulation/genetics , Down-Regulation/immunology , Granzymes/metabolism , Immune Tolerance/genetics , Immune Tolerance/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/metabolism , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome Protein/metabolism
12.
J Exp Med ; 203(6): 1551-65, 2006 Jun 12.
Article in English | MEDLINE | ID: mdl-16754717

ABSTRACT

X-linked lymphoproliferative disease is caused by mutations affecting SH2D1A/SAP, an adaptor that recruits Fyn to signal lymphocyte activation molecule (SLAM)-related receptors. After infection, SLAM-associated protein (SAP)-/- mice show increased T cell activation and impaired humoral responses. Although SAP-/- mice can respond to T-independent immunization, we find impaired primary and secondary T-dependent responses, with defective B cell proliferation, germinal center formation, and antibody production. Nonetheless, transfer of wild-type but not SAP-deficient CD4 cells rescued humoral responses in reconstituted recombination activating gene 2-/- and SAP-/- mice. To investigate these T cell defects, we examined CD4 cell function in vitro and in vivo. Although SAP-deficient CD4 cells have impaired T cell receptor-mediated T helper (Th)2 cytokine production in vitro, we demonstrate that the humoral defects can be uncoupled from cytokine expression defects in vivo. Instead, SAP-deficient T cells exhibit decreased and delayed inducible costimulator (ICOS) induction and heightened CD40L expression. Notably, in contrast to Th2 cytokine defects, humoral responses, ICOS expression, and CD40L down-regulation were rescued by retroviral reconstitution with SAP-R78A, a SAP mutant that impairs Fyn binding. We further demonstrate a role for SLAM/SAP signaling in the regulation of early surface CD40L expression. Thus, SAP affects expression of key molecules required for T-B cell collaboration by mechanisms that are distinct from its role in cytokine regulation.


Subject(s)
Antibody Formation , Cytokines/immunology , Glycoproteins/immunology , Immunoglobulins/immunology , Lymphoproliferative Disorders/immunology , T-Lymphocytes/immunology , X Chromosome , Animals , Antigens, CD , Glycoproteins/deficiency , Glycoproteins/genetics , Immunoglobulins/deficiency , Immunoglobulins/genetics , Lymphoproliferative Disorders/genetics , Mice , Mice, Knockout , Mutation , Receptors, Cell Surface , Signaling Lymphocytic Activation Molecule Family Member 1
13.
Blood ; 115(7): 1433-43, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-20007544

ABSTRACT

It is known that CBFB-MYH11, the fusion gene generated by inversion of chromosome 16 in human acute myeloid leukemia, is causative for oncogenic transformation. However, the mechanism by which CBFB-MYH11 initiates leukemogenesis is not clear. Previously published reports showed that CBFB-MYH11 dominantly inhibits RUNX1 and CBFB, and such inhibition has been suggested as the mechanism for leukemogenesis. Here we show that Cbfb-MYH11 caused Cbfb/Runx1 repression-independent defects in both primitive and definitive hematopoiesis. During primitive hematopoiesis, Cbfb-MYH11 delayed differentiation characterized by sustained expression of Gata2, Il1rl1, and Csf2rb, a phenotype not found in Cbfb and Runx1 knockout mice. Expression of Cbfb-MYH11 in the bone marrow induced the accumulation of abnormal progenitor-like cells expressing Csf2rb in preleukemic mice. The expression of all 3 genes was detected in most human and murine CBFB-MYH11(+) leukemia samples. Interestingly, Cbfb-MYH11(+) preleukemic progenitors and leukemia-initiating cells did not express Csf2rb, although the majority of leukemia cells in our Cbfb-MYH11 knockin mice were Csf2rb(+). Therefore Csf2rb can be used as a negative selection marker to enrich preleukemic progenitor cells and leukemia-initiating cells from Cbfb-MYH11 mice. These results suggest that Cbfb/Runx1 repression-independent activities contribute to leukemogenesis by Cbfb-MYH11.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Cytokine Receptor Common beta Subunit/metabolism , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/metabolism , Animals , Apoptosis/physiology , Biomarkers , Cell Differentiation/physiology , Cell Division/physiology , Core Binding Factor Alpha 2 Subunit/genetics , Cytokine Receptor Common beta Subunit/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Leukemic , Hematopoiesis/physiology , Hematopoietic Stem Cells/pathology , Humans , Interleukin-1 Receptor-Like 1 Protein , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins, Fusion/genetics , Phenotype , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Up-Regulation/physiology
14.
Nat Commun ; 13(1): 805, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35145086

ABSTRACT

T follicular helper (Tfh) cells provide signals to initiate and maintain the germinal center (GC) reaction and are crucial for the generation of robust, long-lived antibody responses, but how the GC microenvironment affects Tfh cells is not well understood. Here we develop an in vivo T cell-intrinsic CRISPR-knockout screen to evaluate Tfh and Th1 cells in an acute viral infection model to identify regulators of Tfh cells in their physiological setting. Using a screen of druggable-targets, alongside genetic, transcriptomic and cellular analyses, we identify a function of HIF-1α in suppressing mTORC1-mediated and Myc-related pathways, and provide evidence that VHL-mediated degradation of HIF-1α is required for Tfh development; an expanded in vivo CRISPR screen reveals multiple components of these pathways that regulate Tfh versus Th1 cells, including signaling molecules, cell-cycle regulators, nutrient transporters, metabolic enzymes and autophagy mediators. Collectively, our data serve as a resource for studying Tfh versus Th1 decisions, and implicate the VHL-HIF-1α axis in fine-tuning Tfh generation.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Animals , Antibody Formation , Cell Differentiation/immunology , Gene Expression , Gene Knockout Techniques , Germinal Center/immunology , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunity, Humoral/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Virus Diseases/immunology
15.
Res Sq ; 2022 Mar 23.
Article in English | MEDLINE | ID: mdl-35350206

ABSTRACT

SARS-CoV-2 infection triggers adaptive immune responses from both T and B cells. However, most studies focus on peripheral blood, which may not fully reflect immune responses in lymphoid tissues at the site of infection. To evaluate both local and systemic adaptive immune responses to SARS-CoV-2, we collected peripheral blood, tonsils, and adenoids from 110 children undergoing tonsillectomy/adenoidectomy during the COVID-19 pandemic and found 24 with evidence of prior SARS-CoV-2 infection, including detectable neutralizing antibodies against multiple viral variants. We identified SARS-CoV-2-specific germinal center (GC) and memory B cells; single cell BCR sequencing showed that these virus-specific B cells were class-switched and somatically hypermutated, with overlapping clones in the adenoids and tonsils. Oropharyngeal tissues from COVID-19-convalescent children showed persistent expansion of GC and anti-viral lymphocyte populations associated with an IFN-γ-type response, with particularly prominent changes in the adenoids, as well as evidence of persistent viral RNA in both tonsil and adenoid tissues of many participants. Our results show robust, tissue-specific adaptive immune responses to SARS-CoV-2 in the upper respiratory tract of children weeks to months after acute infection, providing evidence of persistent localized immunity to this respiratory virus.

16.
Blood ; 114(25): 5162-72, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19843882

ABSTRACT

The transcription factor Gata1 is required for the development of erythrocytes and megakaryocytes. Previous studies with a complementation rescue approach showed that the zinc finger domains are required for both primitive and definitive hematopoiesis. Here we report a novel zebrafish gata1 mutant with an N-ethyl-N-nitrosourea-induced point mutation in the C-finger (gata1(T301K)). The Gata1 protein with this mutation bound to its DNA target sequence with reduced affinity and transactivated inefficiently in a reporter assay. gata1(T301K/T301K) fish had a decreased number of erythrocytes during primitive hematopoiesis but normal adult hematopoiesis. We crossed the gata1(T301K/T301K) fish with those carrying the R339X mutation, also known as vlad tepes (vlt), which abolishes DNA binding and transactivation activities. As we reported previously, gata1(vlt/vlt) embryos were "bloodless" and died approximately 11 to 15 days after fertilization. Interestingly, the gata1(T301K/vlt) fish had nearly a complete block of primitive hematopoiesis, but they resumed hematopoiesis between 7 and 14 days after fertilization and grew to phenotypically normal fish with normal adult hematopoiesis. Our findings suggest that the impact of Gata1 on hematopoiesis correlates with its DNA-binding ability and that primitive hematopoiesis is more sensitive to reduction in Gata1 function than definitive hematopoiesis.


Subject(s)
DNA/metabolism , GATA1 Transcription Factor/metabolism , Hematopoiesis , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , DNA/chemistry , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Female , Flow Cytometry , GATA1 Transcription Factor/chemistry , GATA1 Transcription Factor/genetics , Gene Expression Regulation, Developmental , In Situ Hybridization , Male , Models, Molecular , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Transcriptional Activation , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 105(34): 12337-42, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18719100

ABSTRACT

A major goal for developmental biologists is to define the behaviors and molecular contents of differentiating cells. We have devised a strategy for isolating cells from diverse embryonic regions and stages in the zebrafish, using computer-guided laser photoconversion of injected Kaede protein and flow cytometry. This strategy enabled us to perform a genome-wide transcriptome comparison of germ layer precursor cells. Mesendoderm and ectoderm precursors cells isolated by this method differentiated appropriately in transplantation assays. Microarray analysis of these cells reidentified known genes at least as efficiently as previously reported strategies that relied on artificial mesendoderm activation or inhibition. We also identified a large set of uncharacterized mesendoderm-enriched genes as well as ectoderm-enriched genes. Loss-of-function studies revealed that one of these genes, the MAP kinase inhibitor dusp4, is essential for early development. Embryos injected with antisense morpholino oligonucleotides that targeted Dusp4 displayed necrosis of head tissues. Marker analysis during late gastrulation revealed a specific loss of sox17, but not of other endoderm markers, and analysis at later stages revealed a loss of foregut and pancreatic endoderm. This specific loss of sox17 establishes a new class of endoderm specification defect.


Subject(s)
DNA-Binding Proteins/deficiency , Dual-Specificity Phosphatases/genetics , Germ Layers/cytology , High Mobility Group Proteins/deficiency , Mitogen-Activated Protein Kinase Phosphatases/genetics , Transcription Factors/deficiency , Transcription, Genetic , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Animals , Dual-Specificity Phosphatases/physiology , Ectoderm/cytology , Ectoderm/embryology , Embryo, Nonmammalian , Embryonic Development/genetics , Embryonic Induction/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Germ Layers/embryology , Mesoderm/cytology , Mesoderm/embryology , Mitogen-Activated Protein Kinase Phosphatases/physiology , SOXF Transcription Factors , Zebrafish Proteins/physiology
18.
Blood Adv ; 5(23): 4949-4962, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34492681

ABSTRACT

RUNX1 is essential for the generation of hematopoietic stem cells (HSCs). Runx1-null mouse embryos lack definitive hematopoiesis and die in mid-gestation. However, although zebrafish embryos with a runx1 W84X mutation have defects in early definitive hematopoiesis, some runx1W84X/W84X embryos can develop to fertile adults with blood cells of multilineages, raising the possibility that HSCs can emerge without RUNX1. Here, using 3 new zebrafish runx1-/- lines, we uncovered the compensatory mechanism for runx1-independent hematopoiesis. We show that, in the absence of a functional runx1, a cd41-green fluorescent protein (GFP)+ population of hematopoietic precursors still emerge from the hemogenic endothelium and can colonize the hematopoietic tissues of the mutant embryos. Single-cell RNA sequencing of the cd41-GFP+ cells identified a set of runx1-/--specific signature genes during hematopoiesis. Significantly, gata2b, which normally acts upstream of runx1 for the generation of HSCs, was increased in the cd41-GFP+ cells in runx1-/- embryos. Interestingly, genetic inactivation of both gata2b and its paralog gata2a did not affect hematopoiesis. However, knocking out runx1 and any 3 of the 4 alleles of gata2a and gata2b abolished definitive hematopoiesis. Gata2 expression was also upregulated in hematopoietic cells in Runx1-/- mice, suggesting the compensatory mechanism is conserved. Our findings indicate that RUNX1 and GATA2 serve redundant roles for HSC production, acting as each other's safeguard.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , GATA2 Transcription Factor/metabolism , Hemangioblasts , Zebrafish Proteins/metabolism , Animals , Core Binding Factor Alpha 2 Subunit/genetics , GATA2 Transcription Factor/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells , Mice , Zebrafish , Zebrafish Proteins/genetics
19.
J Cell Biol ; 161(1): 169-86, 2003 Apr 14.
Article in English | MEDLINE | ID: mdl-12682089

ABSTRACT

Neurogenesis is known to persist in the adult mammalian central nervous system (CNS). The identity of the cells that generate new neurons in the postnatal CNS has become a crucial but elusive issue. Using a transgenic mouse, we show that NG2 proteoglycan-positive progenitor cells that express the 2',3'-cyclic nucleotide 3'-phosphodiesterase gene display a multipotent phenotype in vitro and generate electrically excitable neurons, as well as astrocytes and oligodendrocytes. The fast kinetics and the high rate of multipotent fate of these NG2+ progenitors in vitro reflect an intrinsic property, rather than reprogramming. We demonstrate in the hippocampus in vivo that a sizeable fraction of postnatal NG2+ progenitor cells are proliferative precursors whose progeny appears to differentiate into GABAergic neurons capable of propagating action potentials and displaying functional synaptic inputs. These data show that at least a subpopulation of postnatal NG2-expressing cells are CNS multipotent precursors that may underlie adult hippocampal neurogenesis.


Subject(s)
Antigens/metabolism , Cell Differentiation/genetics , Hippocampus/growth & development , Hippocampus/metabolism , Multipotent Stem Cells/metabolism , Nerve Tissue Proteins , Neurons/metabolism , Proteoglycans/metabolism , 2',3'-Cyclic-Nucleotide Phosphodiesterases/genetics , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Action Potentials/genetics , Animals , Animals, Newborn , Antigens/genetics , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Hippocampus/cytology , Intermediate Filament Proteins/metabolism , Mice , Mice, Transgenic , Models, Animal , Multipotent Stem Cells/cytology , Nestin , Neural Pathways/cytology , Neural Pathways/growth & development , Neural Pathways/metabolism , Neurons/cytology , Oligodendroglia/cytology , Oligodendroglia/metabolism , Phenotype , Promoter Regions, Genetic/genetics , Proteoglycans/genetics , Recombinant Fusion Proteins , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , gamma-Aminobutyric Acid/metabolism
20.
J Clin Invest ; 111(9): 1389-97, 2003 May.
Article in English | MEDLINE | ID: mdl-12727931

ABSTRACT

Revertant mosaicism due to true back mutations or second-site mutations has been identified in several inherited disorders. The occurrence of revertants is considered rare, and the underlying genetic mechanisms remain mostly unknown. Here we describe somatic mosaicism in two brothers affected with Wiskott-Aldrich syndrome (WAS). The original mutation causing disease in this family is a single base insertion (1305insG) in the WAS protein (WASP) gene, which results in frameshift and abrogates protein expression. Both patients, however, showed expression of WASP in a fraction of their T cells that were demonstrated to carry a second-site mutation causing the deletion of 19 nucleotides from nucleotide 1299 to 1316. This deletion abrogated the effects of the original mutation and restored the WASP reading frame. In vitro expression studies indicated that mutant protein encoded by the second-site mutation was expressed and functional, since it was able to bind to cellular partners and mediate T cell receptor/CD3 downregulation. These observations were consistent with evidence of in vivo selective advantage of WASP-expressing lymphocytes. Molecular analysis revealed that the sequence surrounding the deletion contained two 4-bp direct repeats and that a hairpin structure could be formed by five GC pairs within the deleted fragment. These findings strongly suggest that slipped mispairing was the cause of this second-site mutation and that selective accumulation of WASP-expressing T lymphocytes led to revertant mosaicism in these patients.


Subject(s)
Mosaicism , Mutation , Proteins/genetics , Wiskott-Aldrich Syndrome/genetics , Adolescent , Child , Child, Preschool , Genes, T-Cell Receptor beta , Genotype , Humans , Infant , Male , Pedigree , Protein Binding , Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein
SELECTION OF CITATIONS
SEARCH DETAIL