Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 64
1.
Elife ; 122024 Apr 03.
Article En | MEDLINE | ID: mdl-38567749

Vitamin D possesses immunomodulatory functions and vitamin D deficiency has been associated with the rise in chronic inflammatory diseases, including asthma (Litonjua and Weiss, 2007). Vitamin D supplementation studies do not provide insight into the molecular genetic mechanisms of vitamin D-mediated immunoregulation. Here, we provide evidence for vitamin D regulation of two human chromosomal loci, Chr17q12-21.1 and Chr17q21.2, reliably associated with autoimmune and chronic inflammatory diseases. We demonstrate increased vitamin D receptor (Vdr) expression in mouse lung CD4+ Th2 cells, differential expression of Chr17q12-21.1 and Chr17q21.2 genes in Th2 cells based on vitamin D status and identify the IL-2/Stat5 pathway as a target of vitamin D signaling. Vitamin D deficiency caused severe lung inflammation after allergen challenge in mice that was prevented by long-term prenatal vitamin D supplementation. Mechanistically, vitamin D induced the expression of the Ikzf3-encoded protein Aiolos to suppress IL-2 signaling and ameliorate cytokine production in Th2 cells. These translational findings demonstrate mechanisms for the immune protective effect of vitamin D in allergic lung inflammation with a strong molecular genetic link to the regulation of both Chr17q12-21.1 and Chr17q21.2 genes and suggest further functional studies and interventional strategies for long-term prevention of asthma and other autoimmune disorders.


Asthma , Pneumonia , Vitamin D Deficiency , Mice , Animals , Humans , Vitamin D/pharmacology , Interleukin-2 , Inflammation , Th2 Cells , Vitamin D Deficiency/metabolism , Vitamins
2.
Nat Immunol ; 25(1): 129-141, 2024 Jan.
Article En | MEDLINE | ID: mdl-37985858

Lymphocyte development consists of sequential and mutually exclusive cell states of proliferative selection and antigen receptor gene recombination. Transitions between each state require large, coordinated changes in epigenetic landscapes and transcriptional programs. How this occurs remains unclear. Here we demonstrate that in small pre-B cells, the lineage and stage-specific epigenetic reader bromodomain and WD repeat-containing protein 1 (BRWD1) reorders three-dimensional chromatin topology to affect the transition between proliferative and gene recombination molecular programs. BRWD1 regulated the switch between poised and active enhancers interacting with promoters, and coordinated this switch with Igk locus contraction. BRWD1 did so by converting chromatin-bound static to dynamic cohesin competent to mediate long-range looping. ATP-depletion revealed cohesin conversion to be the main energetic mechanism dictating dynamic chromatin looping. Our findings provide a new mechanism of cohesin regulation and reveal how cohesin function can be dictated by lineage contextual mechanisms to facilitate specific cell fate transitions.


Chromatin , Cohesins , Chromatin/genetics , Precursor Cells, B-Lymphoid , Gene Expression Regulation , Cell Differentiation , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
3.
Cell ; 186(24): 5269-5289.e22, 2023 11 22.
Article En | MEDLINE | ID: mdl-37995656

A generic level of chromatin organization generated by the interplay between cohesin and CTCF suffices to limit promiscuous interactions between regulatory elements, but a lineage-specific chromatin assembly that supersedes these constraints is required to configure the genome to guide gene expression changes that drive faithful lineage progression. Loss-of-function approaches in B cell precursors show that IKAROS assembles interactions across megabase distances in preparation for lymphoid development. Interactions emanating from IKAROS-bound enhancers override CTCF-imposed boundaries to assemble lineage-specific regulatory units built on a backbone of smaller invariant topological domains. Gain of function in epithelial cells confirms IKAROS' ability to reconfigure chromatin architecture at multiple scales. Although the compaction of the Igκ locus required for genome editing represents a function of IKAROS unique to lymphocytes, the more general function to preconfigure the genome to support lineage-specific gene expression and suppress activation of extra-lineage genes provides a paradigm for lineage restriction.


Chromatin , Genome , B-Lymphocytes/metabolism , CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly , Humans , Animals , Mice
4.
Proc Natl Acad Sci U S A ; 120(35): e2220853120, 2023 08 29.
Article En | MEDLINE | ID: mdl-37607223

Ly6Clo monocytes are a myeloid subset that specializes in the surveillance of vascular endothelium. Ly6Clo monocytes have been shown to derive from Ly6Chi monocytes. NOTCH2 signaling has been implicated as a trigger for Ly6Clo monocyte development, but the basis for this effect is unclear. Here, we examined the impact of NOTCH2 signaling of myeloid progenitors on the development of Ly6Clo monocytes in vitro. NOTCH2 signaling induced by delta-like ligand 1 (DLL1) efficiently induced the transition of Ly6Chi TREML4- monocytes into Ly6Clo TREML4+ monocytes. We further identified two additional transcriptional requirements for development of Ly6Clo monocytes. Deletion of BCL6 from myeloid progenitors abrogated development of Ly6Clo monocytes. IRF2 was also required for Ly6Clo monocyte development in a cell-intrinsic manner. DLL1-induced in vitro transition into Ly6Clo TREML4+ monocytes required IRF2 but unexpectedly could occur in the absence of NUR77 or BCL6. These results imply a transcriptional hierarchy for these factors in controlling Ly6Clo monocyte development.


Endothelium, Vascular , Monocytes , Hematopoiesis , Signal Transduction
5.
Nat Immunol ; 23(11): 1628-1643, 2022 11.
Article En | MEDLINE | ID: mdl-36316479

T cell differentiation requires Notch1 signaling. In the present study, we show that an enhancer upstream of Notch1 active in double-negative (DN) mouse thymocytes is responsible for raising Notch1 signaling intrathymically. This enhancer is required to expand multipotent progenitors intrathymically while delaying early differentiation until lineage restrictions have been established. Early thymic progenitors lacking the enhancer show accelerated differentiation through the DN stages and increased frequency of B, innate lymphoid (IL) and natural killer (NK) cell differentiation. Transcription regulators for T cell lineage restriction and commitment are expressed normally, but IL and NK cell gene expression persists after T cell lineage commitment and T cell receptor ß VDJ recombination, Cd3 expression and ß-selection have been impaired. This Notch1 enhancer is inactive in double-positive (DP) thymocytes. Its aberrant reactivation at this stage in Ikaros mutants is required for leukemogenesis. Thus, the DN-specific Notch1 enhancer harnesses the regulatory architecture of DN and DP thymocytes to achieve carefully orchestrated changes in Notch1 signaling required for early lineage restrictions and normal T cell differentiation.


Immunity, Innate , Thymocytes , Mice , Animals , Thymocytes/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Lymphocytes/metabolism , Thymus Gland , Cell Differentiation/genetics , Cell Lineage/genetics
6.
J Immunol ; 208(12): 2613-2621, 2022 06 15.
Article En | MEDLINE | ID: mdl-35623662

Keratinocytes, the epithelial cells of the skin, reprogram their gene expression and produce immune effector molecules when exposed to environmental and endogenous triggers of inflammation. It remains unclear how keratinocytes process physiological signals generated during skin irritation and switch from a homeostatic to an inflammatory state. In this article, we show that the stress-activated protein kinase p38α is crucial for keratinocytes to prompt changes in their transcriptome upon cytokine stimulation and drive inflammation in allergen-exposed skin. p38α serves this function by phosphorylating p63, a transcription factor essential for the lineage identity and stemness of the skin epithelium. Phosphorylation by p38α alters the activity of p63 and redeploys this developmental transcription factor to a gene expression program linked to inflammation. Genetic ablation and pharmacological inhibition of p38α or the p38α-p63 target gene product MMP13 attenuate atopic dermatitis-like disease in mice. Our study reveals an epithelial molecular pathway promoting skin inflammation and actionable through treatment with topical small-molecule therapeutics.


Dermatitis, Atopic , Mitogen-Activated Protein Kinase 14/metabolism , Transcription Factors , Animals , Dermatitis, Atopic/metabolism , Inflammation/metabolism , Keratinocytes/metabolism , Mice , Phosphorylation , Transcription Factors/metabolism
7.
Nat Commun ; 13(1): 1859, 2022 04 06.
Article En | MEDLINE | ID: mdl-35388001

The cohesin complex participates in the organization of 3D genome through generating and maintaining DNA loops. Stromal antigen 2 (STAG2), a core subunit of the cohesin complex, is frequently mutated in various cancers. However, the impact of STAG2 inactivation on 3D genome organization, especially the long-range enhancer-promoter contacts and subsequent gene expression control in cancer, remains poorly understood. Here we show that depletion of STAG2 in melanoma cells leads to expansion of topologically associating domains (TADs) and enhances the formation of acetylated histone H3 lysine 27 (H3K27ac)-associated DNA loops at sites where binding of STAG2 is switched to its paralog STAG1. We further identify Interferon Regulatory Factor 9 (IRF9) as a major direct target of STAG2 in melanoma cells via integrated RNA-seq, STAG2 ChIP-seq and H3K27ac HiChIP analyses. We demonstrate that loss of STAG2 activates IRF9 through modulating the 3D genome organization, which in turn enhances type I interferon signaling and increases the expression of PD-L1. Our findings not only establish a previously unknown role of the STAG2 to STAG1 switch in 3D genome organization, but also reveal a functional link between STAG2 and interferon signaling in cancer cells, which may enhance the immune evasion potential in STAG2-mutant cancer.


Chromosomal Proteins, Non-Histone , Melanoma , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Genome , Humans , Interferons/genetics , Melanoma/genetics
8.
Leukemia ; 36(1): 42-57, 2022 01.
Article En | MEDLINE | ID: mdl-34193976

Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) is a high-risk subtype of B-ALL often associated with genetic variants that alter cytokine receptor signaling, including mutations in the interleukin-7 receptor (IL7R). To investigate whether IL7R variants are leukemia-initiating, we built mouse models expressing activated Il7r (aIL7R). B-cell intrinsic aIL7R mice developed spontaneous B-ALL, demonstrating sufficiency of Il7r activating mutations in leukemogenesis. Concomitant introduction of a knock-out allele in the associated adapter protein Lnk (encoded by Sh2b3) or a dominant-negative variant of the transcription factor Ikaros (Ikzf1) increased disease penetrance. The resulting murine leukemias displayed monoclonality and recurrent somatic Kras mutations and efficiently engrafted into immunocompetent mice. Phosphoproteomic analyses of aIL7R leukemic cells revealed constitutive Stat5 signaling and B cell receptor (BCR)-like signaling despite the absence of surface pre-BCR. Finally, in vitro treatment of aIL7R leukemic B-cells with Jak, mTOR, or Syk inhibitors blocked growth, confirming that each pathway is active in this mouse model of IL7R-driven B-ALL.


Gene Expression Regulation, Leukemic , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Interleukin-7/metabolism , Animals , Apoptosis , Cell Proliferation , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Interleukin-7/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cancer Cell ; 39(3): 380-393.e8, 2021 03 08.
Article En | MEDLINE | ID: mdl-33689703

Hotspot mutation of IKZF3 (IKZF3-L162R) has been identified as a putative driver of chronic lymphocytic leukemia (CLL), but its function remains unknown. Here, we demonstrate its driving role in CLL through a B cell-restricted conditional knockin mouse model. Mutant Ikzf3 alters DNA binding specificity and target selection, leading to hyperactivation of B cell receptor (BCR) signaling, overexpression of nuclear factor κB (NF-κB) target genes, and development of CLL-like disease in elderly mice with a penetrance of ~40%. Human CLL carrying either IKZF3 mutation or high IKZF3 expression was associated with overexpression of BCR/NF-κB pathway members and reduced sensitivity to BCR signaling inhibition by ibrutinib. Our results thus highlight IKZF3 oncogenic function in CLL via transcriptional dysregulation and demonstrate that this pro-survival function can be achieved by either somatic mutation or overexpression of this CLL driver. This emphasizes the need for combinatorial approaches to overcome IKZF3-mediated BCR inhibitor resistance.


B-Lymphocytes/pathology , Ikaros Transcription Factor/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation/genetics , Transcription, Genetic/genetics , Animals , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , NF-kappa B/genetics , Receptors, Antigen, B-Cell/genetics , Signal Transduction/genetics
10.
Mol Cancer Res ; 19(3): 507-515, 2021 03.
Article En | MEDLINE | ID: mdl-33234577

Hematopoietic-derived cells are integral components of the tumor microenvironment and serve as critical mediators of tumor-host interactions. Host cells derived from myeloid and lymphoid lineages perform well-established functions linked to cancer development, progression, and response to therapy. It is unclear whether host erythroid cells also contribute to shaping the path that cancer can take, but emerging evidence points to this possibility. Here, we show that tumor-promoting environmental stress and tumor-induced hemodynamic changes trigger renal erythropoietin production and erythropoietin-dependent expansion of splenic erythroid cell populations in mice. These erythroid cells display molecular features indicative of an immature erythroid phenotype, such as the expression of both CD71 and TER119 and the retention of intact nuclei, and express genes encoding immune checkpoint molecules. Nucleated erythroid cells with similar properties are present in mouse and human tumor tissues. Antibody-mediated erythropoietin blockade reduces tumor-responsive erythroid cell induction and tumor growth. These findings reveal the potential of tumor-induced erythropoietin and erythroid cells as targets for cancer treatment. IMPLICATIONS: : Our study identifies erythropoietin and erythroid cells as novel players in tumor-host interactions and highlights the involvement of multiorgan signaling events in their induction in response to environmental stress and tumor growth.


Erythroid Cells/metabolism , Immune Checkpoint Proteins/metabolism , Animals , Cell Differentiation , Humans , Mice , Signal Transduction
11.
J Exp Med ; 217(3)2020 03 02.
Article En | MEDLINE | ID: mdl-31834931

Keratinocytes respond to environmental signals by eliciting induction of genes that preserve skin's integrity. Here we show that the transcriptional response to stress signaling is supported by short-lived epigenetic changes. Comparison of chromatin accessibility and transcriptional changes induced by barrier disruption or by loss of the nucleosome remodeler Mi-2ß identified their striking convergence in mouse and human keratinocytes. Mi-2ß directly repressed genes induced by barrier disruption by restricting AP1-enriched promoter-distal sites, occupied by Mi-2ß and JUNB at steady state and by c-JUN after Mi-2ß depletion or stress signaling. Barrier disruption led to a modest reduction in Mi-2ß expression and a further selective reduction of Mi-2ß localization at stress response genes, possibly through competition with activated c-JUN. Consistent with a repressive role at stress response genes, genetic ablation of Mi-2ß did not prevent reestablishment of barrier integrity but was required for return to homeostasis. Thus, a competition between Mi-2ß-repressive and activating AP1 complexes may permit rapid transcriptional response to and resolution from stress signaling.


Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Skin/metabolism , Transcription Factor AP-1/metabolism , Animals , Cells, Cultured , Homeostasis/physiology , Humans , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/genetics , Signal Transduction/physiology , Transcription, Genetic/genetics
12.
Genes Dev ; 33(13-14): 763-781, 2019 07 01.
Article En | MEDLINE | ID: mdl-31123064

Coordinated induction, but also repression, of genes are key to normal differentiation. Although the role of lineage-specific transcription regulators has been studied extensively, their functional integration with chromatin remodelers, one of the key enzymatic machineries that control chromatin accessibility, remains ill-defined. Here we investigate the role of Mi-2ß, a SNF-2-like nucleosome remodeler and key component of the nucleosome remodeling and histone deacetylase (NuRD) complex in early B cells. Inactivation of Mi-2ß arrested differentiation at the large pre-B-cell stage and caused derepression of cell adhesion and cell migration signaling factors by increasing chromatin access at poised enhancers and chromosome architectural elements. Mi-2ß also supported IL-7R signaling, survival, and proliferation by repressing negative effectors of this pathway. Importantly, overexpression of Bcl2, a mitochondrial prosurvival gene and target of IL-7R signaling, partly rescued the differentiation block caused by Mi-2ß loss. Mi-2ß stably associated with chromatin sites that harbor binding motifs for IKAROS and EBF1 and physically associated with these transcription factors both on and off chromatin. Notably, Mi-2ß shared loss-of-function cellular and molecular phenotypes with IKAROS and EBF1, albeit in a distinct fashion. Thus, the nucleosome remodeler Mi-2ß promotes pre-B-cell differentiation by providing repression capabilities to distinct lineage-specific transcription factor-based regulatory networks.


B-Lymphocytes/cytology , Cell Differentiation/genetics , Chromatin/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Expression Regulation, Developmental , Animals , Cell Lineage , Cell Proliferation/genetics , Cell Survival/genetics , Cells, Cultured , Mice , Transcription Factors
13.
PLoS Genet ; 15(3): e1007765, 2019 03.
Article En | MEDLINE | ID: mdl-30897080

Rare individuals with inactivating mutations in the Huntington's disease gene (HTT) exhibit variable abnormalities that imply essential HTT roles during organ development. Here we report phenotypes produced when increasingly severe hypomorphic mutations in the murine HTT orthologue Htt, (HdhneoQ20, HdhneoQ50, HdhneoQ111), were placed over a null allele (Hdhex4/5). The most severe hypomorphic allele failed to rescue null lethality at gastrulation, while the intermediate, though still severe, alleles yielded recessive perinatal lethality and a variety of fetal abnormalities affecting body size, skin, skeletal and ear formation, and transient defects in hematopoiesis. Comparative molecular analysis of wild-type and Htt-null retinoic acid-differentiated cells revealed gene network dysregulation associated with organ development that nominate polycomb repressive complexes and miRNAs as molecular mediators. Together these findings demonstrate that Htt is required both pre- and post-gastrulation to support normal development.


Huntingtin Protein/genetics , Huntington Disease/genetics , Alleles , Animals , Cell Differentiation/genetics , Disease Models, Animal , Gene Frequency/genetics , Genotype , Huntingtin Protein/physiology , Mice/embryology , Mutation , Nerve Tissue Proteins/genetics , Phenotype
14.
Dev Cell ; 47(6): 693-695, 2018 12 17.
Article En | MEDLINE | ID: mdl-30562513

The transcriptional activators YAP and TAZ integrate mechanical and biochemical signals to support growth and regeneration. A recent study in Nature (Chang et al., 2018) showed that SWI/SNF sequesters YAP from its DNA-binding partner TEAD. Mechanical coupling between cytoskeleton and nucleus reverses this antagonism by nuclear F-actin sequestering of SWI/SNF.


Actins , Transcription Factors , Actin Cytoskeleton , Cell Nucleus , Cytoskeleton
15.
Curr Opin Hematol ; 24(4): 345-352, 2017 Jul.
Article En | MEDLINE | ID: mdl-28463873

PURPOSE OF REVIEW: Loss of IKAROS in committed B cell precursors causes a block in differentiation while at the same time augments aberrant cellular properties, such as bone marrow stromal adhesion, self-renewal and resistance to glucocorticoid-mediated cell death. B cell acute lymphoblastic leukaemias originating from these early stages of B cell differentiation and associated with IKAROS mutations share a high-risk cellular phenotype suggesting that deregulation of IKAROS-based mechanisms cause a highly malignant disease process. RECENT STUDIES: Recent studies show that IKAROS is critical for the activity of super-enhancers at genes required for pre-B cell receptor (BCR) signalling and differentiation, working either downstream of or in parallel with B cell master regulators such as EBF1 and PAX5. IKAROS also directly represses a cryptic regulatory network of transcription factors prevalent in mesenchymal and epithelial precursors that includes YAP1, TEAD1/2, LHX2 and LMO2, and their targets, which are not normally expressed in lymphocytes. IKAROS prevents not only expression of these 'extra-lineage' transcription factors but also their cooperation with endogenous B cell master regulators, such as EBF1 and PAX5, leading to the formation of a de novo for lymphocytes super-enhancer network. IKAROS coordinates with the Polycomb repression complex (PRC2) to provide stable repression of associated genes during B cell development. However, induction of regulatory factors normally repressed by IKAROS starts a feed-forward loop that activates de-novo enhancers and elevates them to super-enhancer status, thereby diminishing PRC2 repression and awakening aberrant epithelial-like cell properties in B cell precursors. SUMMARY: Insight into IKAROS-based transcriptional circuits not only sets new paradigms for cell differentiation but also provides new approaches for classifying and treating high-risk human B-ALL that originates from these early stages of B cell differentiation.


B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation , Gene Regulatory Networks , Transcription, Genetic , Animals , B-Lymphocytes/pathology , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Self Renewal/genetics , Enhancer Elements, Genetic , Humans , Ikaros Transcription Factor/metabolism , Polycomb-Group Proteins/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Protein Binding
16.
Genes Dev ; 31(5): 439-450, 2017 03 01.
Article En | MEDLINE | ID: mdl-28385788

Lymphocyte differentiation is set to produce myriad immune effector cells with the ability to respond to multitudinous foreign substances. The uniqueness of this developmental system lies in not only the great diversity of cellular functions that it can generate but also the ability of its differentiation intermediates and mature effector cells to expand upon demand, thereby providing lifelong immunity. Surprisingly, the goals of this developmental system are met by a relatively small group of DNA-binding transcription factors that work in concert to control the timing and magnitude of gene expression and fulfill the demands for cellular specialization, expansion, and maintenance. The cellular and molecular mechanisms through which these lineage-promoting transcription factors operate have been a focus of basic research in immunology. The mechanisms of development discerned in this effort are guiding clinical research on disorders with an immune cell base. Here, I focus on IKAROS, one of the earliest regulators of lymphoid lineage identity and a guardian of lymphocyte homeostasis.


Cell Differentiation/genetics , Cell Differentiation/immunology , Ikaros Transcription Factor/genetics , Lymphocytes/cytology , Lymphocytes/immunology , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression Regulation, Developmental , Homeostasis/genetics , Homeostasis/immunology , Humans , Ikaros Transcription Factor/immunology , Transcription Factors/genetics , Transcription Factors/immunology
17.
Genes Dev ; 31(2): 85-87, 2017 01 15.
Article En | MEDLINE | ID: mdl-28202537

The organization of the genome within the confines of the nuclear compartment is considered a key contributor to transcription and DNA replication, repair, and recombination. A typical higher eukaryotic cell has a spherical nucleus that is ∼10 µm in diameter. This is not the case for a neutrophil, a short-lived innate immune cell with an unusual multilobular nuclear structure that may serve purposes outside nuclear functions. In this issue of Genes & Development, Zhu and colleagues (pp. 141-153) investigate the neutrophil's genome organization and the mechanisms that contribute to its unique nuclear shape.


Cell Nucleus , Neutrophils/cytology , DNA Replication , Genome , Humans
18.
Nat Immunol ; 18(3): 334-343, 2017 03.
Article En | MEDLINE | ID: mdl-28092372

Environmental challenges to epithelial cells trigger gene expression changes that elicit context-appropriate immune responses. We found that the chromatin remodeler Mi-2ß controls epidermal homeostasis by regulating the genes involved in keratinocyte and immune-cell activation to maintain an inactive state. Mi-2ß depletion resulted in rapid deployment of both a pro-inflammatory and an immunosuppressive response in the skin. A key target of Mi-2ß in keratinocytes is the pro-inflammatory cytokine thymic stromal lymphopoietin (TSLP). Loss of TSLP receptor (TSLPR) signaling specifically in regulatory T (Treg) cells prevented their activation and permitted rapid progression from a skin pro-inflammatory response to a lethal systemic condition. Thus, in addition to their well-characterized role in pro-inflammatory responses, keratinocytes also directly support immune-suppressive responses that are critical for re-establishing organismal homeostasis.


Cytokines/metabolism , DNA Helicases/metabolism , Immunoglobulins/metabolism , Keratinocytes/physiology , Receptors, Cytokine/metabolism , T-Lymphocytes, Regulatory/physiology , Animals , Cell Communication , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , DNA Helicases/genetics , Immunoglobulins/genetics , Inflammation/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytokine/genetics , Signal Transduction/genetics , Thymic Stromal Lymphopoietin
19.
J Biol Chem ; 292(5): 1762-1772, 2017 02 03.
Article En | MEDLINE | ID: mdl-28011639

The evolutionarily conserved protein kinase p38 mediates innate resistance to environmental stress and microbial infection. Four p38 isoforms exist in mammals and may have been co-opted for new roles in adaptive immunity. Murine T cells deficient in p38α, the ubiquitously expressed p38 isoform, showed no readily apparent cell-autonomous defects while expressing elevated amounts of another isoform, p38ß. Mice with T cells simultaneously lacking p38α and p38ß displayed lymphoid atrophy and elevated Foxp3+ regulatory T cell frequencies. Double deficiency of p38α and p38ß in naïve CD4+ T cells resulted in an attenuation of MAPK-activated protein kinase (MK)-dependent mTOR signaling after T cell receptor engagement, and enhanced their differentiation into regulatory T cells under appropriate inducing conditions. Pharmacological inhibition of the p38-MK-mTOR signaling module produced similar effects, revealing potential for therapeutic applications.


MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinase 11/immunology , Mitogen-Activated Protein Kinase 14/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 14/genetics , Receptors, Antigen, T-Cell/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology
20.
Genes Dev ; 30(17): 1971-90, 2016 09 01.
Article En | MEDLINE | ID: mdl-27664237

IKAROS is required for the differentiation of highly proliferative pre-B-cell precursors, and loss of IKAROS function indicates poor prognosis in precursor B-cell acute lymphoblastic leukemia (B-ALL). Here we show that IKAROS regulates this developmental stage by positive and negative regulation of superenhancers with distinct lineage affiliations. IKAROS defines superenhancers at pre-B-cell differentiation genes together with B-cell master regulators such as PAX5, EBF1, and IRF4 but is required for a highly permissive chromatin environment, a function that cannot be compensated for by the other transcription factors. IKAROS is also highly enriched at inactive enhancers of genes normally expressed in stem-epithelial cells. Upon IKAROS loss, expression of pre-B-cell differentiation genes is attenuated, while a group of extralineage transcription factors that are directly repressed by IKAROS and depend on EBF1 relocalization at their enhancers for expression is induced. LHX2, LMO2, and TEAD-YAP1, normally kept separate from native B-cell transcription regulators by IKAROS, now cooperate directly with them in a de novo superenhancer network with its own feed-forward transcriptional reinforcement. Induction of de novo superenhancers antagonizes Polycomb repression and superimposes aberrant stem-epithelial cell properties in a B-cell precursor. This dual mechanism of IKAROS regulation promotes differentiation while safeguarding against a hybrid stem-epithelial-B-cell phenotype that underlies high-risk B-ALL.


Cell Differentiation/genetics , Enhancer Elements, Genetic/physiology , Epithelial Cells/cytology , Gene Expression Regulation, Leukemic , Ikaros Transcription Factor/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Precursor Cells, B-Lymphoid/cytology , Animals , Epigenesis, Genetic , Epithelial Cells/pathology , Ikaros Transcription Factor/genetics , Mice , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cells, B-Lymphoid/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
...