Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nat Immunol ; 22(7): 851-864, 2021 07.
Article in English | MEDLINE | ID: mdl-34099918

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are essential to maintain tissue homeostasis. In cancer, ILC2s can harbor both pro-tumorigenic and anti-tumorigenic functions, but we know little about their underlying mechanisms or whether they could be clinically relevant or targeted to improve patient outcomes. Here, we found that high ILC2 infiltration in human melanoma was associated with a good clinical prognosis. ILC2s are critical producers of the cytokine granulocyte-macrophage colony-stimulating factor, which coordinates the recruitment and activation of eosinophils to enhance antitumor responses. Tumor-infiltrating ILC2s expressed programmed cell death protein-1, which limited their intratumoral accumulation, proliferation and antitumor effector functions. This inhibition could be overcome in vivo by combining interleukin-33-driven ILC2 activation with programmed cell death protein-1 blockade to significantly increase antitumor responses. Together, our results identified ILC2s as a critical immune cell type involved in melanoma immunity and revealed a potential synergistic approach to harness ILC2 function for antitumor immunotherapies.


Subject(s)
Antibodies/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Interleukin-33/pharmacology , Lymphocytes/drug effects , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Cell Line, Tumor , Chemotaxis, Leukocyte/drug effects , Cytotoxicity, Immunologic/drug effects , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/metabolism
2.
Immunity ; 48(4): 618-620, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29669243

ABSTRACT

Generation of functionally diverse effector and memory killer T cells is essential for immediate and long-term protective immunity. Herndler-Brandstetter et al. (2018) report that Bach2 promotes functional plasticity of effector T cells and the transition into the long-term memory compartment by regulating the expression of the inhibitory receptor KLRG1.


Subject(s)
Memory , T-Lymphocytes, Cytotoxic
3.
Genes Dev ; 30(1): 78-91, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26728554

ABSTRACT

Unlike clustered HOX genes, the role of nonclustered homeobox gene family members in hematopoiesis and leukemogenesis has not been extensively studied. Here we found that the hematopoietically expressed homeobox gene Hhex is overexpressed in acute myeloid leukemia (AML) and is essential for the initiation and propagation of MLL-ENL-induced AML but dispensable for normal myelopoiesis, indicating a specific requirement for Hhex for leukemic growth. Loss of Hhex leads to expression of the Cdkn2a-encoded tumor suppressors p16(INK4a) and p19(ARF), which are required for growth arrest and myeloid differentiation following Hhex deletion. Mechanistically, we show that Hhex binds to the Cdkn2a locus and directly interacts with the Polycomb-repressive complex 2 (PRC2) to enable H3K27me3-mediated epigenetic repression. Thus, Hhex is a potential therapeutic target that is specifically required for AML stem cells to repress tumor suppressor pathways and enable continued self-renewal.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Epigenesis, Genetic , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/physiopathology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Gene Deletion , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Inbred C57BL , Protein Binding , Transcription Factors/genetics
4.
Chem Soc Rev ; 46(20): 6255-6275, 2017 Oct 16.
Article in English | MEDLINE | ID: mdl-28816316

ABSTRACT

Natural extracellular matrices (ECMs) have been widely used as a support for the adhesion, migration, differentiation, and proliferation of adipose-derived stem cells (ADSCs). However, poor mechanical behavior and unpredictable biodegradation properties of natural ECMs considerably limit their potential for bioapplications and raise the need for different, synthetic scaffolds. Hydrogels are regarded as the most promising alternative materials as a consequence of their excellent swelling properties and their resemblance to soft tissues. A variety of strategies have been applied to create synthetic biomimetic hydrogels, and their biophysical and biochemical properties have been modulated to be suitable for cell differentiation. In this review, we first give an overview of common methods for hydrogel preparation with a focus on those strategies that provide potential advantages for ADSC encapsulation, before summarizing the physical properties of hydrogel scaffolds that can act as biological cues. Finally, the challenges in the preparation and application of hydrogels with ADSCs are explored and the perspectives are proposed for the next generation of scaffolds.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation , Hydrogels/pharmacology , Stem Cells/cytology , Adipose Tissue/drug effects , Cell Differentiation/drug effects , Extracellular Matrix/drug effects , Humans , Hydrogels/chemical synthesis , Hydrogels/chemistry , Hydrogels/metabolism , Stem Cells/drug effects
5.
Mucosal Immunol ; 17(3): 371-386, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38492744

ABSTRACT

Interleukin-(IL) 22 production by intestinal group 3 innate lymphoid cells (ILC3) is critical to maintain gut homeostasis. However, IL-22 needs to be tightly controlled; reduced IL-22 expression is associated with intestinal epithelial barrier defect while its overexpression promotes tumor development. Here, using a single-cell ribonucleic acid sequencing approach, we identified a core set of genes associated with increased IL-22 production by ILC3. Among these genes, programmed cell death 1 (PD-1), extensively studied in the context of cancer and chronic infection, was constitutively expressed on a subset of ILC3. These cells, found in the crypt of the small intestine and colon, displayed superior capacity to produce IL-22. PD-1 expression on ILC3 was dependent on the microbiota and was induced during inflammation in response to IL-23 but, conversely, was reduced in the presence of Notch ligand. PD-1+ ILC3 exhibited distinct metabolic activity with increased glycolytic, lipid, and polyamine synthesis associated with augmented proliferation compared with their PD-1- counterparts. Further, PD-1+ ILC3 showed increased expression of mitochondrial antioxidant proteins which enable the cells to maintain their levels of reactive oxygen species. Loss of PD-1 signaling in ILC3 led to reduced IL-22 production in a cell-intrinsic manner. During inflammation, PD-1 expression was increased on natural cytotoxicity receptor (NCR)- ILC3 while deficiency in PD-1 expression resulted in increased susceptibility to experimental colitis and failure to maintain gut barrier integrity. Collectively, our findings uncover a new function of the PD-1 and highlight the role of PD-1 signaling in the maintenance of gut homeostasis mediated by ILC3 in mice.


Subject(s)
Homeostasis , Immunity, Innate , Interleukin-22 , Interleukins , Lymphocytes , Mice, Knockout , Programmed Cell Death 1 Receptor , Animals , Mice , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Lymphocytes/immunology , Lymphocytes/metabolism , Interleukins/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Signal Transduction , Colitis/immunology , Intestines/immunology , Mice, Inbred C57BL , Humans , Disease Models, Animal
6.
Sci Immunol ; 9(95): eadj2654, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38820141

ABSTRACT

Tissue-resident innate lymphoid cells (ILCs) play a vital role in the frontline defense of various tissues, including the lung. The development of type 2 ILCs (ILC2s) depends on transcription factors such as GATA3, RORα, GFI1, and Bcl11b; however, the factors regulating lung-resident ILC2s remain unclear. Through fate mapping analysis of the paralog transcription factors GFI1 and GFI1B, we show that GFI1 is consistently expressed during the transition from progenitor to mature ILC2s. In contrast, GFI1B expression is limited to specific subsets of bone marrow progenitors and lung-resident ILC progenitors. We found that GFI1B+ lung ILC progenitors represent a multi-lineage subset with tissue-resident characteristics and the potential to form lung-derived ILC subsets and liver-resident ILC1s. Loss of GFI1B in bone marrow progenitors led to the selective loss of lung-resident IL-18R+ ILCs and mature ILC2, subsequently preventing the emergence of effector ILCs that could protect the lung against inflammatory or tumor challenge.


Subject(s)
Immunity, Innate , Lung , Mice, Inbred C57BL , Proto-Oncogene Proteins , Animals , Lung/immunology , Lung/cytology , Mice , Immunity, Innate/immunology , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/metabolism , Lymphoid Progenitor Cells/immunology , Lymphoid Progenitor Cells/cytology , Repressor Proteins/genetics , Repressor Proteins/immunology , Mice, Knockout , Lymphocytes/immunology , Cell Differentiation/immunology , DNA-Binding Proteins , Transcription Factors
7.
Pharmaceutics ; 15(7)2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37514187

ABSTRACT

Reinvigorating the killing function of tumor-infiltrating immune cells through the targeting of regulatory molecules expressed on lymphocytes has markedly improved the prognosis of cancer patients, particularly in melanoma. While initially thought to solely strengthen adaptive T lymphocyte anti-tumor activity, recent investigations suggest that other immune cell subsets, particularly tissue-resident innate lymphoid cells (ILCs), may benefit from immunotherapy treatment. Here, we describe the recent findings showing immune checkpoint expression on tissue-resident and tumor-infiltrating ILCs and how their effector function is modulated by checkpoint blockade-based therapies in cancer. We discuss the therapeutic potential of ILCs beyond the classical PD-1 and CTLA-4 regulatory molecules, exploring other possibilities to manipulate ILC effector function to further impede tumor growth and quench disease progression.

8.
STAR Protoc ; 3(3): 101534, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35830307

ABSTRACT

Innate lymphoid cells (ILCs) and adaptive T cells remain a challenge to study because of a significant overlap in their transcriptomic profiles. Here, we describe the adoptive transfer of ILC progenitors into mice genetically deficient in innate and adaptive immune cells to allow detailed study of the development and function of ILCs and gene regulation in an in vivo setting. For complete details on the use and execution of this protocol, please refer to Jacquelot et al. (2021) and Seillet et al. (2016).


Subject(s)
Immunity, Innate , Lymphocytes , Animals , Bone Marrow , Lymphoid Progenitor Cells , Mice , T-Lymphocytes
9.
ACS Macro Lett ; 10(8): 1015-1022, 2021 08 17.
Article in English | MEDLINE | ID: mdl-35549122

ABSTRACT

Hierarchical self-assembly offers great possibilities to mimic biological systems with finely arranged complex structures. Herein, we demonstrate the preparation and formation mechanism of an unusual giant polymer vesicle with a latticelike membrane (GVLM). This GVLM is formed by fusion-induced particle assembly (FIPA) of small vesicles that are self-assembled from poly(ethylene oxide)-block-poly[(2-(tetrahydrofuranyloxy)ethyl methacrylate)-stat-(6-(3,3-diphenylnaphthopyranyloxy)hexyl methacrylate)] [PEO43-b-P(TMA22-stat-NMA4)]. Flexible TMA units with high chain mobility and relatively rigid NMA units with intrinsic π-π stacking form the hydrophobic block. These units act as "antifusion" and "profusion" components, respectively. The latticelike membrane of the final GVLM consists of hundreds of small polymer vesicles that are interconnected via multiple interactions. Transmission electron microscopy (TEM) and dynamic light scattering (DLS) studies show that the diameter of the GVLMs is 800-1000 nm. Overall, we provide a new insight into the judicious preparation of hierarchical nanostructures via chemical synthesis and FIPA.


Subject(s)
Nanostructures , Polymers , Hydrophobic and Hydrophilic Interactions , Microscopy, Electron, Transmission , Polyethylene Glycols/chemistry , Polymers/chemistry
10.
Cancers (Basel) ; 13(3)2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33535624

ABSTRACT

Chronic inflammation of the gastrointestinal (GI) tract contributes to colorectal cancer (CRC) progression. While the role of adaptive T cells in CRC is now well established, the role of innate immune cells, specifically innate lymphoid cells (ILCs), is not well understood. To define the role of ILCs in CRC we employed complementary heterotopic and chemically-induced CRC mouse models. We discovered that ILCs were abundant in CRC tumours and contributed to anti-tumour immunity. We focused on ILC2 and showed that ILC2-deficient mice developed a higher tumour burden compared with littermate wild-type controls. We generated an ILC2 gene signature and using machine learning models revealed that CRC patients with a high intratumor ILC2 gene signature had a favourable clinical prognosis. Collectively, our results highlight a critical role for ILC2 in CRC, suggesting a potential new avenue to improve clinical outcomes through ILC2-agonist based therapeutic approaches.

11.
Curr Opin Immunol ; 58: 53-59, 2019 06.
Article in English | MEDLINE | ID: mdl-31125785

ABSTRACT

Adaptive and innate immune cells have typically been functionally and temporally segregated even though they share a number of salient features. Over the past decade, significant advances have been made in understanding the composition and diversity of both innate and adaptive cell populations. This has shed light on how cells from two distinct pathways are so highly complementary. Innate lymphoid cells (ILCs) are pivotally positioned in tissues to form a stable population akin to tissue-resident T cells that protects the body. Nevertheless, the pathway by which different lymphocytes enter tissues, terminally differentiate and are replenished to maintain populations remains incompletely understood. Recent evidence challenges our assumptions about the sedentary lifestyles of so called 'tissue-resident cells' and pushes us to consider their roles in orchestrating protection of the immune system beyond the classical models.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Lymphoid Tissue/immunology , T-Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Cell Movement/immunology , Humans , Lymphocytes/cytology , Lymphocytes/immunology , Lymphoid Tissue/cytology , T-Lymphocytes/cytology
12.
Front Immunol ; 10: 3080, 2019.
Article in English | MEDLINE | ID: mdl-32010138

ABSTRACT

The immune system plays a fundamental role at mucosal barriers in maintaining tissue homeostasis. This is particularly true for the gut where cells are flooded with microbial-derived signals and antigens, which constantly challenge the integrity of the intestinal barrier. Multiple immune cell populations equipped with both pro- and anti-inflammatory functions reside in the gut tissue and these cells tightly regulate intestinal health and functions. Dysregulation of this finely tuned system can progressively lead to autoimmune disease and inflammation-driven carcinogenesis. Over the last decade, the contribution of the adaptive immune system in controlling colorectal cancer has been studied in detail, but the role of the innate system, particularly innate lymphoid cells (ILCs), have been largely overlooked. By sensing their microenvironment, ILCs are essential in supporting gut epithelium repair and controling bacterial- and helminth-mediated intestinal infections, highlighting their important role in maintaining tissue integrity. Accumulating evidence also suggests that they may play an important role in carcinogenesis including intestinal cancers. In this review, we will explore the current knowledge about the pro- and anti-tumor functions of ILCs in colorectal cancer.


Subject(s)
Colorectal Neoplasms/immunology , Immunity, Innate , Intestinal Mucosa/immunology , Lymphocytes/immunology , Tumor Microenvironment/immunology , Colorectal Neoplasms/pathology , Cytokines/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Intestinal Mucosa/pathology , Lymphocytes/pathology
13.
J Exp Med ; 216(7): 1682-1699, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31142588

ABSTRACT

Interleukin (IL)-17-producing CD8+ T (Tc17) cells have emerged as key players in host-microbiota interactions, infection, and cancer. The factors that drive their development, in contrast to interferon (IFN)-γ-producing effector CD8+ T cells, are not clear. Here we demonstrate that the transcription factor TCF-1 (Tcf7) regulates CD8+ T cell fate decisions in double-positive (DP) thymocytes through the sequential suppression of MAF and RORγt, in parallel with TCF-1-driven modulation of chromatin state. Ablation of TCF-1 resulted in enhanced Tc17 cell development and exposed a gene set signature to drive tissue repair and lipid metabolism, which was distinct from other CD8+ T cell subsets. IL-17-producing CD8+ T cells isolated from healthy humans were also distinct from CD8+IL-17- T cells and enriched in pathways driven by MAF and RORγt Overall, our study reveals how TCF-1 exerts central control of T cell differentiation in the thymus by normally repressing Tc17 differentiation and promoting an effector fate outcome.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Interleukin-17/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Animals , Chromatin/metabolism , Chromatin Immunoprecipitation Sequencing , Flow Cytometry , Hepatocyte Nuclear Factor 1-alpha/physiology , Humans , Lipid Metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/physiology
14.
Front Immunol ; 8: 1569, 2017.
Article in English | MEDLINE | ID: mdl-29201028

ABSTRACT

Innate lymphoid cells (ILCs) are a key cell type that are enriched at mucosal surfaces and within tissues. Our understanding of these cells is growing rapidly. Paradoxically, these cells play a role in maintaining tissue integrity but they also function as key drivers of allergy and inflammation. We present here the most recent understanding of how genomics has provided significant insight into how ILCs are generated and the enormous heterogeneity present within the canonical subsets. This has allowed the generation of a detailed blueprint for ILCs to become highly sensitive and adaptive sensors of environmental changes and therefore exquisitely equipped to protect immune surfaces.

SELECTION OF CITATIONS
SEARCH DETAIL