Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Cell ; 165(5): 1120-1133, 2016 May 19.
Article in English | MEDLINE | ID: mdl-27156451

ABSTRACT

Innate lymphoid cells (ILCs) play key roles in host defense, barrier integrity, and homeostasis and mirror adaptive CD4(+) T helper (Th) cell subtypes in both usage of effector molecules and transcription factors. To better understand the relationship between ILC subsets and their Th cell counterparts, we measured genome-wide chromatin accessibility. We find that chromatin in proximity to effector genes is selectively accessible in ILCs prior to high-level transcription upon activation. Accessibility of these regions is acquired in a stepwise manner during development and changes little after in vitro or in vivo activation. Conversely, dramatic chromatin remodeling occurs in naive CD4(+) T cells during Th cell differentiation using a type-2-infection model. This alteration results in a substantial convergence of Th2 cells toward ILC2 regulomes. Our data indicate extensive sharing of regulatory circuitry across the innate and adaptive compartments of the immune system, in spite of their divergent developing pathways.


Subject(s)
Gene Regulatory Networks , Lymphocytes/cytology , Lymphocytes/immunology , Animals , Cell Lineage , Female , Gene Expression Regulation , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Transcriptome
2.
Immunity ; 54(3): 514-525.e6, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33657395

ABSTRACT

MicroRNAs are important regulators of immune responses. Here, we show miR-221 and miR-222 modulate the intestinal Th17 cell response. Expression of miR-221 and miR-222 was induced by proinflammatory cytokines and repressed by the cytokine TGF-ß. Molecular targets of miR-221 and miR-222 included Maf and Il23r, and loss of miR-221 and miR-222 expression shifted the transcriptomic spectrum of intestinal Th17 cells to a proinflammatory signature. Although the loss of miR-221 and miR-222 was tolerated for maintaining intestinal Th17 cell homeostasis in healthy mice, Th17 cells lacking miR-221 and miR-222 expanded more efficiently in response to IL-23. Both global and T cell-specific deletion of miR-221 and miR-222 rendered mice prone to mucosal barrier damage. Collectively, these findings demonstrate that miR-221 and miR-222 are an integral part of intestinal Th17 cell response that are induced after IL-23 stimulation to constrain the magnitude of proinflammatory response.


Subject(s)
Inflammation/immunology , Interleukin-23/metabolism , Intestinal Mucosa/immunology , MicroRNAs/genetics , Th17 Cells/immunology , Animals , Feedback, Physiological , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-maf/metabolism , Receptors, Interleukin/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
3.
Immunity ; 53(4): 745-758.e4, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33010223

ABSTRACT

Innate immune responses rely on rapid and precise gene regulation mediated by accessibility of regulatory regions to transcription factors (TFs). In natural killer (NK) cells and other innate lymphoid cells, competent enhancers are primed during lineage acquisition, and formation of de novo enhancers characterizes the acquisition of innate memory in activated NK cells and macrophages. Here, we investigated how primed and de novo enhancers coordinate to facilitate high-magnitude gene induction during acute activation. Epigenomic and transcriptomic analyses of regions near highly induced genes (HIGs) in NK cells both in vitro and in a model of Toxoplasma gondii infection revealed de novo chromatin accessibility and enhancer remodeling controlled by signal-regulated TFs STATs. Acute NK cell activation redeployed the lineage-determining TF T-bet to de novo enhancers, independent of DNA-sequence-specific motif recognition. Thus, acute stimulation reshapes enhancer function through the combinatorial usage and repurposing of both lineage-determining and signal-regulated TFs to ensure an effective response.


Subject(s)
Enhancer Elements, Genetic/genetics , Enhancer Elements, Genetic/immunology , Killer Cells, Natural/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Animals , Chromatin/genetics , Chromatin/immunology , Female , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Toxoplasma/immunology , Toxoplasmosis/genetics , Toxoplasmosis/immunology
4.
Proc Natl Acad Sci U S A ; 120(18): e2216587120, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37098069

ABSTRACT

Innate lymphoid cells (ILCs) play a key role in tissue-mediated immunity and can be controlled by coreceptor signaling. Here, we define a subset of ILCs that are Tbet+NK1.1- and are present within the tumor microenvironment (TME). We show programmed death-1 receptor (PD-1) expression on ILCs within TME is found in Tbet+NK1.1- ILCs. PD-1 significantly controlled the proliferation and function of Tbet+NK1.1- ILCs in multiple murine and human tumors. We found tumor-derived lactate enhanced PD-1 expression on Tbet+NK1.1- ILCs within the TME, which resulted in dampened the mammalian target of rapamycin (mTOR) signaling along with increased fatty acid uptake. In line with these metabolic changes, PD-1-deficient Tbet+NK1.1- ILCs expressed significantly increased IFNγ and granzyme B and K. Furthermore, PD-1-deficient Tbet+NK1.1- ILCs contributed toward diminished tumor growth in an experimental murine model of melanoma. These data demonstrate that PD-1 can regulate antitumor responses of Tbet+NK1.1- ILCs within the TME.


Subject(s)
Lymphocytes , Neoplasms , Mice , Animals , Humans , Immunity, Innate , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Tumor Microenvironment , Neoplasms/metabolism , Apoptosis , Mammals/metabolism
5.
Proc Natl Acad Sci U S A ; 120(40): e2306761120, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37756335

ABSTRACT

Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1) require signal transducer and activator of transcription 4 (STAT4) to elicit rapid effector responses and protect against pathogens. By combining genetic and transcriptomic approaches, we uncovered divergent roles for STAT4 in regulating effector differentiation of these functionally related cell types. Stat4 deletion in Ncr1-expressing cells led to impaired NK cell terminal differentiation as well as to an unexpected increased generation of cytotoxic ILC1 during intestinal inflammation. Mechanistically, Stat4-deficient ILC1 exhibited upregulation of gene modules regulated by STAT5 in vivo and an aberrant effector differentiation upon in vitro stimulation with IL-2, used as a prototypical STAT5 activator. Moreover, STAT4 expression in NCR+ innate lymphocytes restrained gut inflammation in the dextran sulfate sodium-induced colitis model limiting pathogenic production of IL-13 from adaptive CD4+ T cells in the large intestine. Collectively, our data shed light on shared and distinctive mechanisms of STAT4-regulated transcriptional control in NK cells and ILC1 required for intestinal inflammatory responses.


Subject(s)
Antineoplastic Agents , STAT5 Transcription Factor , Humans , Immunity, Innate , Cell Differentiation , Killer Cells, Natural , Inflammation , STAT4 Transcription Factor/genetics
6.
Nat Immunol ; 13(6): 587-95, 2012 Apr 29.
Article in English | MEDLINE | ID: mdl-22544395

ABSTRACT

Distinct CD4(+) T cell subsets are critical for host defense and immunoregulation. Although these subsets can act as terminally differentiated lineages, they have been increasingly noted to demonstrated plasticity. MicroRNAs are factors that control T cell stability and plasticity. Here we report that naturally occurring regulatory T cells (T(reg) cells) had high expression of the microRNA miR-10a and that miR-10a was induced by retinoic acid and transforming growth factor-ß (TGF-ß) in inducible T(reg) cells. By simultaneously targeting the transcriptional repressor Bcl-6 and the corepressor Ncor2, miR-10a attenuated the phenotypic conversion of inducible T(reg) cells into follicular helper T cells. We also found that miR-10a limited differentiation into the T(H)17 subset of helper T cells and therefore represents a factor that can fine-tune the plasticity and fate of helper T cells.


Subject(s)
MicroRNAs/biosynthesis , Proto-Oncogene Proteins c-bcl-6/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology , Animals , Cell Differentiation/immunology , Down-Regulation/immunology , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/immunology , Nuclear Receptor Co-Repressor 2/immunology , Phenotype , Proto-Oncogene Proteins c-bcl-6/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Transcription, Genetic
7.
Immunity ; 42(5): 877-89, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25992861

ABSTRACT

Interleukin-6 (IL-6) and IL-27 signal through a shared receptor subunit and employ the same downstream STAT transcription proteins, but yet are ascribed unique and overlapping functions. To evaluate the specificity and redundancy for these cytokines, we quantified their global transcriptomic changes and determined the relative contributions of STAT1 and STAT3 using genetic models and chromatin immunoprecipitation-sequencing (ChIP-seq) approaches. We found an extensive overlap of the transcriptomes induced by IL-6 and IL-27 and few examples in which the cytokines acted in opposition. Using STAT-deficient cells and T cells from patients with gain-of-function STAT1 mutations, we demonstrated that STAT3 is responsible for the overall transcriptional output driven by both cytokines, whereas STAT1 is the principal driver of specificity. STAT1 cannot compensate in the absence of STAT3 and, in fact, much of STAT1 binding to chromatin is STAT3 dependent. Thus, STAT1 shapes the specific cytokine signature superimposed upon STAT3's action.


Subject(s)
Chromatin/metabolism , Cytokines/metabolism , Gene Expression Regulation/immunology , Models, Immunological , STAT Transcription Factors/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Chromatin/chemistry , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Mutation , STAT1 Transcription Factor/chemistry , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Transcriptome
8.
Eur J Immunol ; 51(11): 2568-2575, 2021 11.
Article in English | MEDLINE | ID: mdl-34347289

ABSTRACT

Type 1 innate lymphoid cells (ILC1) are tissue-resident lymphocytes that provide early protection against bacterial and viral infections. Discrete transcriptional states of ILC1 have been identified in homeostatic and pathological contexts. However, whether these states delineate ILC1 with different functional properties is not completely understood. Here, we show that liver ILC1 are heterogeneous for the expression of distinct effector molecules and surface receptors, including granzyme A (GzmA) and CD160, in mice. ILC1 expressing high levels of GzmA are enriched in the liver of adult mice, and represent the main hepatic ILC1 population at birth. However, the heterogeneity of GzmA and CD160 expression in hepatic ILC1 begins perinatally and increases with age. GzmA+ ILC1 differ from NK cells for the limited homeostatic requirements of JAK/STAT signals and the transcription factor Nfil3. Moreover, by employing Rorc(γt)-fate map (fm) reporter mice, we established that ILC3-ILC1 plasticity contributes to delineate the heterogeneity of liver ILC1, with RORγt-fm+ cells skewed toward a GzmA- CD160+ phenotype. Finally, we showed that ILC1 defined by the expression of GzmA and CD160 are characterized by graded cytotoxic potential and ability to produce IFN-γ. In conclusion, our findings help deconvoluting ILC1 heterogeneity and provide evidence for functional diversification of liver ILC1.


Subject(s)
Liver/cytology , Liver/immunology , Lymphocyte Subsets/cytology , Lymphocytes/cytology , Animals , Antigens, CD/metabolism , GPI-Linked Proteins/metabolism , Granzymes/metabolism , Immunity, Innate/immunology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Receptors, Immunologic/metabolism
9.
Int J Mol Sci ; 23(14)2022 Jul 16.
Article in English | MEDLINE | ID: mdl-35887206

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignancies and leading causes of cancer-related deaths worldwide. Despite its complex pathogenesis and progression, CRC represents a well-fitting example of how the immune contexture can dictate the disease outcome. The presence of cytotoxic lymphocytes, both CD8+ T cells and natural killer (NK) cells, represents a relevant prognostic factor in CRC and is associated with a better overall survival. Together with NK cells, other innate lymphocytes, namely, innate lymphoid cells (ILCs), have been found both in biopsies of CRC patients and in murine models of intestinal cancer, playing both pro- and anti-tumor activities. In particular, several type 1 innate lymphoid cells (ILC1) with cytotoxic functions have been recently described, and evidence in mice shows a role for both NK cells and ILC1 in controlling CRC metastasis. In this review, we provide an overview of the features of NK cells and the expanding spectrum of innate lymphocytes with cytotoxic functions. We also comment on both the described and the potential roles these innate lymphocytes can play during the progression of intestinal cancer leading to metastasis. Finally, we discuss recent advances in the molecular mechanisms underlying the functional regulation of cytotoxic innate lymphocytes in CRC.


Subject(s)
Colorectal Neoplasms , Lymphocytes , Animals , CD8-Positive T-Lymphocytes , Immunity, Innate , Killer Cells, Natural , Mice
10.
Immunol Rev ; 286(1): 148-159, 2018 11.
Article in English | MEDLINE | ID: mdl-30294965

ABSTRACT

Immunity to pathogens is ensured through integration of early responses mediated by innate cells and late effector functions taking place after terminal differentiation of adaptive lymphocytes. In this context, innate lymphoid cells (ILCs) and adaptive T cells represent a clear example of how prototypical effector functions, including polarized expression of cytokines and/or cytotoxic activity, can occur with overlapping modalities but different timing. The ability of ILCs to provide early protection relies on their poised epigenetic state, which determines their propensity to quickly respond to cytokines and to activate specific patterns of signal-dependent transcription factors. Cytokines activating the Janus kinases (JAKs) and members of the signal transducer and activator of transcription (STAT) pathway are key regulators of lymphoid development and sustain the processes underlying T-cell activation and differentiation. The role of the JAK/STAT pathway has been recently extended to several aspects of ILC biology. Here, we discuss how JAK/STAT signals affect ILC development and effector functions in the context of immune responses, highlighting the molecular mechanisms involved in regulation of gene expression as well as the potential of targeting the JAK/STAT pathway in inflammatory pathologies.


Subject(s)
Janus Kinases/metabolism , Lymphocytes/immunology , STAT Transcription Factors/metabolism , Animals , Cytokines/metabolism , Gene Expression Regulation , Humans , Immunity, Innate , Inflammation , Signal Transduction
11.
Immunity ; 36(6): 1017-30, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22726954

ABSTRACT

Interleukin-27 (IL-27) is a key immunosuppressive cytokine that counters T helper 17 (Th17) cell-mediated pathology. To identify mechanisms by which IL-27 might exert its immunosuppressive effect, we analyzed genes in T cells rapidly induced by IL-27. We found that IL-27 priming of naive T cells upregulated expression of programmed death ligand 1 (PD-L1) in a signal transducer and activator of transcription 1 (STAT1)-dependent manner. When cocultured with naive CD4(+) T cells, IL-27-primed T cells inhibited the differentiation of Th17 cells in trans through a PD-1-PD-L1 interaction. In vivo, coadministration of naive TCR transgenic T cells (2D2 T cells) with IL-27-primed T cells expressing PD-L1 inhibited the development of Th17 cells and protected from severe autoimmune encephalomyelitis. Thus, these data identify a suppressive activity of IL-27, by which CD4(+) T cells can restrict differentiation of Th17 cells in trans.


Subject(s)
B7-H1 Antigen/physiology , Interleukin-17/biosynthesis , Interleukins/pharmacology , Th17 Cells/drug effects , Animals , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/deficiency , B7-H1 Antigen/genetics , Bystander Effect , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Forkhead Transcription Factors/deficiency , Gene Expression Regulation/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-17/genetics , Interleukin-17/physiology , Interleukin-1beta/pharmacology , Interleukin-23/pharmacology , Interleukin-6/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Minor Histocompatibility Antigens , Myelin Proteins/immunology , Myelin Proteins/toxicity , Myelin-Oligodendrocyte Glycoprotein , Receptors, Cytokine/deficiency , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Th17 Cells/cytology , Th17 Cells/metabolism , Th17 Cells/transplantation , Transforming Growth Factor beta/pharmacology
12.
Eur J Immunol ; 48(7): 1174-1180, 2018 07.
Article in English | MEDLINE | ID: mdl-29524223

ABSTRACT

Innate lymphoid cells (ILCs) producing IL-22 and/or IL-17, designated as ILC3, comprise a heterogeneous subset of cells involved in regulation of gut barrier homeostasis and inflammation. Exogenous environmental cues in conjunction with regulated expression of endogenous factors are key determinants of plasticity of ILC3 toward the type 1 fate. Herein, by using mouse models and transcriptomic approaches, we defined at the molecular level, initial events driving ILC3 expressing natural cytotoxicity receptors (NCR+ ILC3) to acquire type 1 features. We observed that NCR+ ILC3 exhibited high basal expression of the signal-dependent transcription factor STAT4 due to T-BET, leading to predisposed potential for the type 1 response. We found that the prototypical inducer of type 3 response, IL-23, played a predominant role over IL-12 by accessing STAT4 and preferentially inducing its phosphorylation in ILC3 expressing T-BET. The early effector program driven by IL-23 was characterized by the expression of IL-22, followed by a production of IFN-γ, which relies on STAT4, T-BET and required chromatin remodeling of the Ifng locus. Altogether, our findings shed light on a feed-forward mechanism involving STAT4 and T-BET that modulates the outcome of IL-23 signaling in ILC3.


Subject(s)
Interleukin-23/metabolism , Intestines/immunology , Lymphocytes/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Ly/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Homeostasis , Humans , Immunity, Mucosal , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Intestines/pathology , Mice , Natural Cytotoxicity Triggering Receptor 1/metabolism , Protein Binding , Signal Transduction , T-Box Domain Proteins/genetics , Th1 Cells/immunology , Interleukin-22
13.
Biomed Microdevices ; 21(2): 33, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30906958

ABSTRACT

We couple a tumor growth model embedded in a microenvironment, with a bio distribution model able to simulate a whole organ. The growth model yields the evolution of tumor cell population, of the differential pressure between cell populations, of porosity of ECM, of consumption of nutrients due to tumor growth, of angiogenesis, and related growth factors as function of the locally available nutrient. The bio distribution model on the other hand operates on a frozen geometry but yields a much refined distribution of nutrient and other molecules. The combination of both models will enable simulating the growth of a tumor in a whole organ, including a realistic distribution of therapeutic agents and allow hence to evaluate the efficacy of these agents.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Models, Biological , Cell Proliferation , Extracellular Matrix/metabolism , Melanoma/blood supply , Neovascularization, Pathologic , Nutrients/pharmacokinetics , Tissue Distribution , Tumor Microenvironment
14.
Nature ; 498(7455): 506-10, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23728300

ABSTRACT

Through their functional diversification, distinct lineages of CD4(+) T cells can act to either drive or constrain immune-mediated pathology. Transcription factors are critical in the generation of cellular diversity, and negative regulators antagonistic to alternate fates often act in conjunction with positive regulators to stabilize lineage commitment. Genetic polymorphisms within a single locus encoding the transcription factor BACH2 are associated with numerous autoimmune and allergic diseases including asthma, Crohn's disease, coeliac disease, vitiligo, multiple sclerosis and type 1 diabetes. Although these associations point to a shared mechanism underlying susceptibility to diverse immune-mediated diseases, a function for BACH2 in the maintenance of immune homeostasis has not been established. Here, by studying mice in which the Bach2 gene is disrupted, we define BACH2 as a broad regulator of immune activation that stabilizes immunoregulatory capacity while repressing the differentiation programs of multiple effector lineages in CD4(+) T cells. BACH2 was required for efficient formation of regulatory (Treg) cells and consequently for suppression of lethal inflammation in a manner that was Treg-cell-dependent. Assessment of the genome-wide function of BACH2, however, revealed that it represses genes associated with effector cell differentiation. Consequently, its absence during Treg polarization resulted in inappropriate diversion to effector lineages. In addition, BACH2 constrained full effector differentiation within TH1, TH2 and TH17 cell lineages. These findings identify BACH2 as a key regulator of CD4(+) T-cell differentiation that prevents inflammatory disease by controlling the balance between tolerance and immunity.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Homeostasis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeostasis/genetics , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/mortality , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/pharmacology
15.
Immunol Rev ; 261(1): 23-49, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25123275

ABSTRACT

The discovery of the specification of CD4(+) helper T cells to discrete effector 'lineages' represented a watershed event in conceptualizing mechanisms of host defense and immunoregulation. However, our appreciation for the actual complexity of helper T-cell subsets continues unabated. Just as the Sami language of Scandinavia has 1000 different words for reindeer, immunologists recognize the range of fates available for a CD4(+) T cell is numerous and may be underestimated. Added to the crowded scene for helper T-cell subsets is the continuously growing family of innate lymphoid cells (ILCs), endowed with common effector responses and the previously defined 'master regulators' for CD4(+) helper T-cell subsets are also shared by ILC subsets. Within the context of this extraordinary complexity are concomitant advances in the understanding of transcriptomes and epigenomes. So what do terms like 'lineage commitment' and helper T-cell 'specification' mean in the early 21st century? How do we put all of this together in a coherent conceptual framework? It would be arrogant to assume that we have a sophisticated enough understanding to seriously answer these questions. Instead, we review the current status of the flexibility of helper T-cell responses in relation to their genetic regulatory networks and epigenetic landscapes. Recent data have provided major surprises as to what master regulators can or cannot do, how they interact with other transcription factors and impact global genome-wide changes, and how all these factors come together to influence helper cell function.


Subject(s)
Gene Regulatory Networks , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Differentiation , Cell Lineage , Epigenesis, Genetic , Gene Regulatory Networks/genetics , Gene Regulatory Networks/immunology , Humans , Immunity, Cellular , Immunity, Innate , Immunomodulation , Transcriptome
16.
Blood ; 123(19): 2978-87, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24632714

ABSTRACT

Mutations of STAT3 underlie the autosomal dominant form of hyperimmunoglobulin E syndrome (HIES). STAT3 has critical roles in immune cells and thus, hematopoietic stem cell transplantation (HSCT), might be a reasonable therapeutic strategy in this disease. However, STAT3 also has critical functions in nonhematopoietic cells and dissecting the protean roles of STAT3 is limited by the lethality associated with germline deletion of Stat3. Thus, predicting the efficacy of HSCT for HIES is difficult. To begin to dissect the importance of STAT3 in hematopoietic and nonhematopoietic cells as it relates to HIES, we generated a mouse model of this disease. We found that these transgenic mice recapitulate multiple aspects of HIES, including elevated serum IgE and failure to generate Th17 cells. We found that these mice were susceptible to bacterial infection that was partially corrected by HSCT using wild-type bone marrow, emphasizing the role played by the epithelium in the pathophysiology of HIES.


Subject(s)
Disease Models, Animal , Job Syndrome/immunology , Mutation/immunology , STAT3 Transcription Factor/immunology , Animals , Bone Marrow Transplantation , Cells, Cultured , Citrobacter rodentium/immunology , Citrobacter rodentium/physiology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Flow Cytometry , Host-Pathogen Interactions/immunology , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Job Syndrome/genetics , Job Syndrome/surgery , Lipopolysaccharides , Mice , Mice, Transgenic , Mutation/genetics , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Shock, Septic/chemically induced , Shock, Septic/genetics , Shock, Septic/immunology , Survival Analysis , Transcriptome/genetics , Transcriptome/immunology
17.
J Immunol ; 191(11): 5684-94, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24184559

ABSTRACT

NK cell differentiation mainly occurs in the bone marrow (BM) where a critical role in the regulation of developing lymphocyte distribution is played by members of the chemokine receptor family. In mouse, the chemokine receptor CX3CR1 identifies a late stage of NK cell development characterized by decreased effector functions and expression of the inhibitory receptor KLRG1. The role of CX3CR1 in the regulation of differentiation and positioning of NK cell subsets in the BM is not known. In this study, we found that CX3CR1 deficiency leads to accumulation of KLRG1(+) NK cells in BM during steady-state conditions. The NK cell subset that expresses the receptor in wild-type mice was expanded in several tissues of CX3CR1-deficient mice, and NK cell degranulation in response to sensitive target cell stimulation was enhanced, suggesting a regulatory role of CX3CR1 in NK cell positioning and differentiation in BM. Indeed, the observed NK cell expansion was not due to altered turnover rate, whereas it was associated with preferential accumulation in the BM parenchyma. In addition, a role of CX3CR1 in NK cell trafficking from BM and spleen was evidenced also during inflammation, as CX3CR1-deficient NK cells were more prompt to exit the BM and did not decrease in spleen in response to polyinosinic-polycytidylic acid-promoted hepatitis. Overall, our results evidenced a relevant role of CX3CR1 in the regulation of NK cell subset exit from BM during homeostasis, and suggest that defect in the CX3CR1/CX3CL1 axis alters NK cell trafficking and functional response during inflammatory conditions.


Subject(s)
Hepatitis/immunology , Killer Cells, Natural/immunology , Receptors, Chemokine/metabolism , Animals , Blood Circulation , Bone Marrow Cells/immunology , CX3C Chemokine Receptor 1 , Cell Degranulation/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Cell Proliferation , Cytotoxicity, Immunologic/genetics , Female , Homeostasis/genetics , Lectins, C-Type , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Poly I-C/immunology , Receptors, Chemokine/genetics , Receptors, Immunologic/metabolism
18.
J Clin Invest ; 134(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949019

ABSTRACT

Type 3 innate lymphoid cells (ILC3s) are key regulators of intestinal homeostasis and epithelial barrier integrity. In this issue of the JCI, Cao and colleagues found that a sensor of endoplasmic reticulum (ER) stress, the inositol-requiring kinase 1α/X-box-binding protein 1 (IRE1α/XBP1) pathway, fine-tuned the functions of ILC3s. Activation of IRE1α and XBP1 in ILC3s limited intestinal inflammation in mice and correlated with the efficacy of ustekinumab, an IL-12/IL-23 blocker, in patients with Crohn's disease. These results advance our understanding in the use of ILCs as biomarkers not only to predict disease outcomes but also to indicate the response to biologicals in patients with inflammatory bowel disease.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases , Protein Serine-Threonine Kinases , X-Box Binding Protein 1 , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/immunology , Animals , Endoribonucleases/metabolism , Endoribonucleases/genetics , Endoribonucleases/immunology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Humans , Mice , Endoplasmic Reticulum Stress/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Signal Transduction/immunology , Crohn Disease/immunology , Crohn Disease/pathology , Crohn Disease/metabolism , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology
19.
J Mech Behav Biomed Mater ; 156: 106584, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38810544

ABSTRACT

Biomechanical parameters have the potential to be used as physical markers for prevention and diagnosis. Finite Element Analysis (FEA) is a widely used tool to evaluate these parameters in vivo. However, the development of clinically relevant FEA requires personalisation of the geometry, boundary conditions, and constitutive parameters. This contribution focuses on the characterisation of mechanical properties in vivo which remains a significant challenge for the community. The aim of this retrospective study is to evaluate the sensitivity of the computed elastic parameters (shear modulus of fat and muscle tissues) derived by inverse analysis as a function of the geometrical modelling assumption (homogenised monolayer vs bilayer) and the formulation of the cost function. The methodology presented here proposes to extract the experimental force-displacement response for each tissue layer (muscle and fat) and construct the associated Finite Element Model for each volunteer, based on data previously collected in our group (N = 7 volunteers) as reported in (Fougeron et al., 2020). The sensitivity analysis indicates that the choice of the cost function has minimal impact on the topology of the response surface in the parametric space. Each surface displays a valley of parameters that minimises the cost function. The constitutive properties of the thigh (reported as median ± interquartile range) were determined to be (µ=198±322Pa,α=37) for the monolayer and (µmuscle=1675±1127Pa,αmuscle=22±14,µfat=537±1131Pa,αfat=32±7) for the bilayer. A comparison of the homogenised monolayer and bilayer models showed that adding a layer reduces the error on the local force displacement curves, increasing the accuracy of the local kinematics of soft tissues during indentation. This allows for an increased understanding of load transmission in soft tissue. The comparison of the two models in terms of strains indicates that the modelling choice significantly influences the localization of maximal compressive strains. These results support the idea that the biomechanical community should conduct further work to develop reliable methodologies for estimating in vivo strain in soft tissue.


Subject(s)
Finite Element Analysis , Thigh , Biomechanical Phenomena , Thigh/physiology , Humans , Compressive Strength , Male , Models, Biological , Adult , Stress, Mechanical , Adipose Tissue
20.
Front Immunol ; 15: 1366197, 2024.
Article in English | MEDLINE | ID: mdl-38601156

ABSTRACT

Introduction: Chemotherapy remains the mainstay treatment for triple-negative breast cancer (TNBC) due to the lack of specific targets. Given a modest response of immune checkpoint inhibitors in TNBC patients, improving immunotherapy is an urgent and crucial task in this field. CD73 has emerged as a novel immunotherapeutic target, given its elevated expression on tumor, stromal, and specific immune cells, and its established role in inhibiting anti-cancer immunity. CD73-generated adenosine suppresses immunity by attenuating tumor-infiltrating T- and NK-cell activation, while amplifying regulatory T cell activation. Chemotherapy often leads to increased CD73 expression and activity, further suppressing anti-tumor immunity. While debulking the tumor mass, chemotherapy also enriches heterogenous cancer stem cells (CSC), potentially leading to tumor relapse. Therefore, drugs targeting both CD73, and CSCs hold promise for enhancing chemotherapy efficacy, overcoming treatment resistance, and improving clinical outcomes. However, safe and effective inhibitors of CD73 have not been developed as of now. Methods: We used in silico docking to screen compounds that may be repurposed for inhibiting CD73. The efficacy of these compounds was investigated through flow cytometry, RT-qPCR, CD73 activity, cell viability, tumorsphere formation, and other in vitro functional assays. For assessment of clinical translatability, TNBC patient-derived xenograft organotypic cultures were utilized. We also employed the ovalbumin-expressing AT3 TNBC mouse model to evaluate tumor-specific lymphocyte responses. Results: We identified quercetin and luteolin, currently used as over-the-counter supplements, to have high in silico complementarity with CD73. When quercetin and luteolin were combined with the chemotherapeutic paclitaxel in a triple-drug regimen, we found an effective downregulation in paclitaxel-enhanced CD73 and CSC-promoting pathways YAP and Wnt. We found that CD73 expression was required for the maintenance of CD44highCD24low CSCs, and co-targeting CD73, YAP, and Wnt effectively suppressed the growth of human TNBC cell lines and patient-derived xenograft organotypic cultures. Furthermore, triple-drug combination inhibited paclitaxel-enriched CSCs and simultaneously improved lymphocyte infiltration in syngeneic TNBC mouse tumors. Discussion: Conclusively, our findings elucidate the significance of CSCs in impairing anti-tumor immunity. The high efficacy of our triple-drug regimen in clinically relevant platforms not only underscores the importance for further mechanistic investigations but also paves the way for potential development of new, safe, and cost-effective therapeutic strategies for TNBC.


Subject(s)
CD47 Antigen , Triple Negative Breast Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Flavonoids/pharmacology , Luteolin/metabolism , Neoplastic Stem Cells/metabolism , Paclitaxel/therapeutic use , Quercetin/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , CD47 Antigen/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL