Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38876796

ABSTRACT

Innate lymphoid cells (ILCs) are critical for intestinal adaptation to microenvironmental challenges, and the gut mucosa is characterized by low oxygen. Adaptation to low oxygen is mediated by hypoxia-inducible transcription factors (HIFs), and the HIF-1α subunit shapes an ILC phenotype upon acute colitis that contributes to intestinal damage. However, the impact of HIF signaling in NKp46+ ILCs in the context of repetitive mucosal damage and chronic inflammation, as it typically occurs during inflammatory bowel disease, is unknown. In chronic colitis, mice lacking the HIF-1α isoform in NKp46+ ILCs show a decrease in NKp46+ ILC1s but a concomitant rise in neutrophils and Ly6Chigh macrophages. Single-nucleus RNA sequencing suggests enhanced interaction of mesenchymal cells with other cell compartments in the colon of HIF-1α KO mice and a loss of mucus-producing enterocytes and intestinal stem cells. This was, furthermore, associated with increased bone morphogenetic pathway-integrin signaling, expansion of fibroblast subsets, and intestinal fibrosis. In summary, this suggests that HIF-1α-mediated ILC1 activation, although detrimental upon acute colitis, protects against excessive inflammation and fibrosis during chronic intestinal damage.


Subject(s)
Colitis , Fibrosis , Hypoxia-Inducible Factor 1, alpha Subunit , Lymphocytes , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1 , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Natural Cytotoxicity Triggering Receptor 1/genetics , Mice , Colitis/metabolism , Colitis/genetics , Lymphocytes/metabolism , Lymphocytes/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Inflammation/metabolism , Mice, Inbred C57BL , Chronic Disease , Immunity, Innate , Signal Transduction , Disease Models, Animal , Male , Intestines/pathology , Antigens, Ly
2.
EMBO Rep ; 24(6): e56156, 2023 06 05.
Article in English | MEDLINE | ID: mdl-36987917

ABSTRACT

Natural killer (NK) cells are forced to cope with different oxygen environments even under resting conditions. The adaptation to low oxygen is regulated by oxygen-sensitive transcription factors, the hypoxia-inducible factors (HIFs). The function of HIFs for NK cell activation and metabolic rewiring remains controversial. Activated NK cells are predominantly glycolytic, but the metabolic programs that ensure the maintenance of resting NK cells are enigmatic. By combining in situ metabolomic and transcriptomic analyses in resting murine NK cells, our study defines HIF-1α as a regulator of tryptophan metabolism and cellular nicotinamide adenine dinucleotide (NAD+ ) levels. The HIF-1α/NAD+ axis prevents ROS production during oxidative phosphorylation (OxPhos) and thereby blocks DNA damage and NK cell apoptosis under steady-state conditions. In contrast, in activated NK cells under hypoxia, HIF-1α is required for glycolysis, and forced HIF-1α expression boosts glycolysis and NK cell performance in vitro and in vivo. Our data highlight two distinct pathways by which HIF-1α interferes with NK cell metabolism. While HIF-1α-driven glycolysis is essential for NK cell activation, resting NK cell homeostasis relies on HIF-1α-dependent tryptophan/NAD+ metabolism.


Subject(s)
NAD , Tryptophan , Mice , Animals , Tryptophan/metabolism , Killer Cells, Natural , Glycolysis/genetics , Hypoxia/metabolism , Cell Hypoxia , Oxygen/metabolism , Homeostasis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
3.
Sci Adv ; 9(2): eadc8825, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36638181

ABSTRACT

Metastatic disease is a major cause of death for patients with melanoma. Melanoma cells can become metastatic not only due to cell-intrinsic plasticity but also due to cancer-induced protumorigenic remodeling of the immune microenvironment. Here, we report that innate immune surveillance by natural killer (NK) cells is bypassed by human melanoma cells expressing the stem cell marker NGFR. Using in vitro and in vivo cytotoxic assays, we show that NGFR protects melanoma cells from NK cell-mediated killing and, furthermore, boosts metastasis formation in a mouse model with adoptively transferred human NK cells. Mechanistically, NGFR leads to down-regulation of NK cell activating ligands and simultaneous up-regulation of the fatty acid stearoyl-coenzyme A desaturase (SCD) in melanoma cells. Notably, pharmacological and small interfering RNA-mediated inhibition of SCD reverted NGFR-induced NK cell evasion in vitro and in vivo. Hence, NGFR orchestrates immune control antagonizing pathways to protect melanoma cells from NK cell clearance, which ultimately favors metastatic disease.


Subject(s)
Antineoplastic Agents , Melanoma , Mice , Animals , Humans , Cell Line, Tumor , Melanoma/pathology , Killer Cells, Natural , Lipids , Tumor Microenvironment , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism
4.
Cell Stem Cell ; 29(10): 1459-1474.e9, 2022 Oct 06.
Article in English | MEDLINE | ID: mdl-36113462

ABSTRACT

Fibrosis is the final path of nearly every form of chronic disease, regardless of the pathogenesis. Upon chronic injury, activated, fibrogenic fibroblasts deposit excess extracellular matrix, and severe tissue fibrosis can occur in virtually any organ. However, antifibrotic therapies that target fibrogenic cells, while sparing homeostatic fibroblasts in healthy tissues, are limited. We tested whether specific immunization against endogenous proteins, strongly expressed in fibrogenic cells but highly restricted in quiescent fibroblasts, can elicit an antigen-specific cytotoxic T cell response to ameliorate organ fibrosis. In silico epitope prediction revealed that activation of the genes Adam12 and Gli1 in profibrotic cells and the resulting "self-peptides" can be exploited for T cell vaccines to ablate fibrogenic cells. We demonstrate the efficacy of a vaccination approach to mount CD8+ T cell responses that reduce fibroblasts and fibrosis in the liver and lungs in mice. These results provide proof of principle for vaccination-based immunotherapies to treat fibrosis.


Subject(s)
Fibroblasts , Lung , Animals , Epitopes/metabolism , Fibroblasts/metabolism , Fibrosis , Immunotherapy , Liver/pathology , Lung/metabolism , Mice , Vaccination , Zinc Finger Protein GLI1/metabolism
5.
J Exp Med ; 219(2)2022 02 07.
Article in English | MEDLINE | ID: mdl-35024767

ABSTRACT

Gut innate lymphoid cells (ILCs) show remarkable phenotypic diversity, yet microenvironmental factors that drive this plasticity are incompletely understood. The balance between NKp46+, IL-22-producing, group 3 ILCs (ILC3s) and interferon (IFN)-γ-producing group 1 ILCs (ILC1s) contributes to gut homeostasis. The gut mucosa is characterized by physiological hypoxia, and adaptation to low oxygen is mediated by hypoxia-inducible transcription factors (HIFs). However, the impact of HIFs on ILC phenotype and gut homeostasis is not well understood. Mice lacking the HIF-1α isoform in NKp46+ ILCs show a decrease in IFN-γ-expressing, T-bet+, NKp46+ ILC1s and a concomitant increase in IL-22-expressing, RORγt+, NKp46+ ILC3s in the gut mucosa. Single-cell RNA sequencing revealed HIF-1α as a driver of ILC phenotypes, where HIF-1α promotes the ILC1 phenotype by direct up-regulation of T-bet. Loss of HIF-1α in NKp46+ cells prevents ILC3-to-ILC1 conversion, increases the expression of IL-22-inducible genes, and confers protection against intestinal damage. Taken together, our results suggest that HIF-1α shapes the ILC phenotype in the gut.


Subject(s)
Antigens, Ly/metabolism , Cell Plasticity/immunology , Gastrointestinal Tract/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunity, Innate , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Animals , Biomarkers , Disease Susceptibility , Gene Expression , Gene Expression Profiling , Homeostasis , Immunity, Mucosal , Immunophenotyping , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lymphocyte Subsets , Mice , Mice, Knockout , Microbiota , Single-Cell Analysis
6.
Nat Commun ; 12(1): 4700, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34349124

ABSTRACT

During skin injury, immune response and repair mechanisms have to be coordinated for rapid skin regeneration and the prevention of microbial infections. Natural Killer (NK) cells infiltrate hypoxic skin lesions and Hypoxia-inducible transcription factors (HIFs) mediate adaptation to low oxygen. We demonstrate that mice lacking the Hypoxia-inducible factor (HIF)-1α isoform in NK cells show impaired release of the cytokines Interferon (IFN)-γ and Granulocyte Macrophage - Colony Stimulating Factor (GM-CSF) as part of a blunted immune response. This accelerates skin angiogenesis and wound healing. Despite rapid wound closure, bactericidal activity and the ability to restrict systemic bacterial infection are impaired. Conversely, forced activation of the HIF pathway supports cytokine release and NK cell-mediated antibacterial defence including direct killing of bacteria by NK cells despite delayed wound closure. Our results identify, HIF-1α in NK cells as a nexus that balances antimicrobial defence versus global repair in the skin.


Subject(s)
Killer Cells, Natural/immunology , Skin/immunology , Skin/microbiology , Wound Healing , Animals , Cell Hypoxia , Cytokines/metabolism , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Mice , Neovascularization, Physiologic , Skin/blood supply , Skin Diseases, Bacterial/prevention & control
7.
Theranostics ; 8(14): 3856-3869, 2018.
Article in English | MEDLINE | ID: mdl-30083264

ABSTRACT

Monoclonal antibodies (mAbs) have significantly improved the treatment of certain cancers. However, in general mAbs alone have limited therapeutic activity. One of their main mechanisms of action is to induce antibody-dependent cell-mediated cytotoxicity (ADCC), which is mediated by natural killer (NK) cells. Unfortunately, most cancer patients have severe immune dysfunctions affecting NK activity. This can be circumvented by the injection of allogeneic, expanded NK cells, which is safe. Nevertheless, despite their strong cytolytic potential against different tumors, clinical results have been poor. Methods: We combined allogeneic NK cells and mAbs to improve cancer treatment. We generated expanded NK cells (e-NK) with strong in vitro and in vivo ADCC responses against different tumors and using different therapeutic mAbs, namely rituximab, obinutuzumab, daratumumab, cetuximab and trastuzumab. Results: Remarkably, e-NK cells can be stored frozen and, after thawing, armed with mAbs. They mediate ADCC through degranulation-dependent and -independent mechanisms. Furthermore, they overcome certain anti-apoptotic mechanisms found in leukemic cells. Conclusion: We have established a new protocol for activation/expansion of NK cells with high ADCC activity. The use of mAbs in combination with e-NK cells could potentially improve cancer treatment.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents, Immunological/administration & dosage , Immunotherapy/methods , Killer Cells, Natural/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Transplantation, Homologous/methods , Animals , Disease Models, Animal , Humans , Mice, SCID , Treatment Outcome
8.
Biomedicines ; 6(2)2018 May 15.
Article in English | MEDLINE | ID: mdl-29762526

ABSTRACT

Hypoxia is a hallmark of inflamed, infected or damaged tissue, and the adaptation to inadequate tissue oxygenation is regulated by hypoxia-inducible factors (HIFs). HIFs are key mediators of the cellular response to hypoxia, but they are also associated with pathological stress such as inflammation, bacteriological infection or cancer. In addition, HIFs are central regulators of many innate and adaptive immunological functions, including migration, antigen presentation, production of cytokines and antimicrobial peptides, phagocytosis as well as cellular metabolic reprogramming. A characteristic feature of immune cells is their ability to infiltrate and operate in tissues with low level of nutrients and oxygen. The objective of this article is to discuss the role of HIFs in the function of innate and adaptive immune cells in hypoxia, with a focus on how hypoxia modulates immunometabolism.

9.
Nat Commun ; 8(1): 1597, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29150606

ABSTRACT

Productive angiogenesis, a prerequisite for tumour growth, depends on the balanced release of angiogenic and angiostatic factors by different cell types within hypoxic tumours. Natural killer (NK) cells kill cancer cells and infiltrate hypoxic tumour areas. Cellular adaptation to low oxygen is mediated by Hypoxia-inducible factors (HIFs). We found that deletion of HIF-1α in NK cells inhibited tumour growth despite impaired tumour cell killing. Tumours developing in these conditions were characterised by a high-density network of immature vessels, severe haemorrhage, increased hypoxia, and facilitated metastasis due to non-productive angiogenesis. Loss of HIF-1α in NK cells increased the bioavailability of the major angiogenic cytokine vascular endothelial growth factor (VEGF) by decreasing the infiltration of NK cells that express angiostatic soluble VEGFR-1. In summary, this identifies the hypoxic response in NK cells as an inhibitor of VEGF-driven angiogenesis, yet, this promotes tumour growth by allowing the formation of functionally improved vessels.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Killer Cells, Natural/metabolism , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor , Cells, Cultured , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics
10.
Oncotarget ; 8(9): 15085-15100, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28118605

ABSTRACT

We have recently shown that targeting Vascular Endothelial Growth Factor (VEGF) specifically in scar-infiltrating myeloid cells prevented remodeling of the sinusoidal vasculature and abrogated the resolution of murine liver fibrosis, thereby unmasking an unanticipated link between angiogenesis and resolution of fibrosis. In a gain of function approach, we wanted to test the impact of VEGF overexpression in myeloid cells on fibrolysis. We observe that genetic inactivation of the von Hippel Lindau protein (VHL), a negative regulator of Hypoxia-inducible factors (HIF) in myeloid cells, leads to increased VEGF expression and most importantly, accelerated matrix degradation and reduced myofibroblast numbers after CCl4 challenge. This is associated with enhanced expression of MMP-2 and -14 as well as lower expression of TIMP-2 in liver endothelial cells. In addition, we report increased expression of MMP-13 in scar-associated macrophages as well as improved liver regeneration upon ablation of VHL in myeloid cells. Finally, therapeutic infusion of macrophages nulli-zygous for VHL or treated with the pharmacologic hydroxylase inhibitor and HIF-inducer Dimethyloxalylglycine (DMOG) accelerates resolution of fibrosis. Hence, boosting the HIF-VEGF signaling axis in macrophages represents a promising therapeutic avenue for the treatment of liver fibrosis.


Subject(s)
Cell Hypoxia/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Cirrhosis/prevention & control , Liver Regeneration/physiology , Myeloid Cells/physiology , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Female , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Macrophages/cytology , Macrophages/metabolism , Male , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
11.
Nat Commun ; 7: 12528, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27538380

ABSTRACT

Chemotherapy remains a mainstay of cancer treatment but its use is often limited by the development of adverse reactions. Severe loss of body weight (cachexia) is a frequent cause of death in cancer patients and is exacerbated by chemotherapy. We show that genetic inactivation of vascular endothelial growth factor (VEGF)-A in myeloid cells prevents chemotherapy-induced cachexia by inhibiting skeletal muscle loss and the lipolysis of white adipose tissue. It also improves clearance of senescent tumour cells by natural killer cells and inhibits tumour regrowth after chemotherapy. The effects depend on the chemoattractant chemerin, which is released by the tumour endothelium in response to chemotherapy. The findings define chemerin as a critical mediator of the immune response, as well as an important inhibitor of cancer cachexia. Targeting myeloid cell-derived VEGF signalling should impede the lipolysis and weight loss that is frequently associated with chemotherapy, thereby substantially improving the therapeutic outcome.


Subject(s)
Antineoplastic Agents/therapeutic use , Cachexia/drug therapy , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Antineoplastic Agents/pharmacology , Cachexia/etiology , Cachexia/immunology , Cachexia/pathology , Chemokines/administration & dosage , Chemokines/immunology , Humans , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/immunology , Lipolysis/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Neoplasms/complications , Neoplasms/immunology , Neoplasms/pathology , Treatment Outcome , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
12.
PLoS One ; 11(4): e0150434, 2016.
Article in English | MEDLINE | ID: mdl-27100180

ABSTRACT

The leucocyte-specific phosphatase CD45 is present in two main isoforms: the large CD45RA and the short CD45RO. We have recently shown that distinctive expression of these isoforms distinguishes natural killer (NK) populations. For example, co-expression of both isoforms identifies in vivo the anti tumor NK cells in hematological cancer patients. Here we show that low CD45 expression associates with less mature, CD56bright, NK cells. Most NK cells in healthy human donors are CD45RA+CD45RO-. The CD45RA-RO+ phenotype, CD45RO cells, is extremely uncommon in B or NK cells, in contrast to T cells. However, healthy donors possess CD45RAdimRO- (CD45RAdim cells), which show immature markers and are largely expanded in hematopoietic stem cell transplant patients. Blood borne cancer patients also have more CD45RAdim cells that carry several features of immature NK cells. However, and in opposition to their association to NK cell progenitors, they do not proliferate and show low expression of the transferrin receptor protein 1/CD71, suggesting low metabolic activity. Moreover, CD45RAdim cells properly respond to in vitro encounter with target cells by degranulating or gaining CD69 expression. In summary, they are quiescent NK cells, with low metabolic status that can, however, respond after encounter with target cells.


Subject(s)
Killer Cells, Natural/immunology , Leukocyte Common Antigens/immunology , Protein Isoforms/immunology , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Biomarkers/metabolism , Bone Marrow/immunology , Cell Line, Tumor , Hematopoietic Stem Cell Transplantation/methods , Humans , K562 Cells , Lectins, C-Type/immunology
13.
EBioMedicine ; 2(10): 1364-76, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26629531

ABSTRACT

Natural killer (NK) cells, a cytotoxic lymphocyte lineage, are able to kill tumor cells in vitro and in mouse models. However, whether these cells display an anti-tumor activity in cancer patients has not been demonstrated. Here we have addressed this issue in patients with several hematological cancers. We found a population of highly activated CD56(dim)CD16(+) NK cells that have recently degranulated, evidence of killing activity, and it is absent in healthy donors. A high percentage of these cells expressed natural killer cell p46-related protein (NKp46), natural-killer group 2, member D (NKG2D) and killer inhibitory receptors (KIRs) and a low percentage expressed NKG2A and CD94. They are also characterized by a high metabolic activity and active proliferation. Notably, we found that activated NK cells from hematological cancer patients have non-NK tumor cell antigens on their surface, evidence of trogocytosis during tumor cell killing. Finally, we found that these activated NK cells are distinguished by their CD45RA(+)RO(+) phenotype, as opposed to non-activated cells in patients or in healthy donors displaying a CD45RA(+)RO(-) phenotype similar to naïve T cells. In summary, we show that CD45RA(+)RO(+) cells, which resemble a unique NK population, have recognized tumor cells and degranulate in patients with hematological neoplasias.


Subject(s)
Antigens, Surface/metabolism , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Biomarkers , Case-Control Studies , Cell Communication , Cell Degranulation/immunology , Hematologic Neoplasms/virology , Humans , Immunophenotyping , K562 Cells , Leukocyte Common Antigens/metabolism , Mice , Phenotype , Protein Isoforms
14.
Oncotarget ; 6(22): 19228-45, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26231043

ABSTRACT

Manipulation of metabolic pathways in hematological cancers has therapeutic potential. Here, we determined the molecular mechanism of action of the metabolic modulator dichloroacetate (DCA) in leukemic cells. We found that DCA induces the AMP-activated protein kinase (AMPK)/p53 pathway with increased efficacy in tumors expressing wild type (wt p53). Clinically relevant, low concentrations of doxorubicin synergize in vitro and in vivo with DCA to further enhance p53 activation and to block tumor progression. Leukemia cell lines and primary leukemic cells containing mutant p53 are resistant to the above-described combination approach. However, DCA synergized with the Hsp90 inhibitor 17-AAG to specifically eliminate these cells. Our studies strongly indicate that depending on the p53 status, different combination therapies would provide better treatment with decreased side effects in hematological cancers.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Leukemia/drug therapy , Leukemia/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Benzoquinones/administration & dosage , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Dichloroacetic Acid/administration & dosage , Doxorubicin/administration & dosage , Drug Synergism , HCT116 Cells , HL-60 Cells , Humans , Lactams, Macrocyclic/administration & dosage , Leukemia/genetics , Mice , Oxidative Phosphorylation , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/genetics
15.
Int J Biochem Cell Biol ; 49: 42-52, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24440757

ABSTRACT

NK cell is an innate immune system lymphocyte lineage with natural cytotoxicity. Its optimal use in the clinic requires in vitro expansion and activation. Cytokines and encounter with target cells activate NK cells and induce proliferation, and this could depend on the presence of other immune cells. Here we activated PBMCs during 5 days with IL-2, with IL-2 plus the tumor cell line K562 and with the lymphoblastoid cell line R69 and perform integrated analyses of microRNA and mRNA expression profiles of purified NK cells. The samples cluster depending on the stimuli and not on the donor, indicating that the pattern of NK cell stimulation is acutely well conserved between individuals. Regulation of mRNA expression is tighter than that of miRNA expression. All stimuli induce a common preserved genetic remodeling. In addition, encounter with target cells mainly activates pathways related to metabolism. Different target cells induce different NK cell remodeling which affects cytokine response and cytotoxicity, supporting the notion that encounter with different target cells significantly changing the activation pattern. We validate our analysis by showing that activation down regulates miR-23a, which is a negative regulator of cathepsin C (CTSC) mRNA, a gene up regulated by all stimuli. The peptidase CTSC activates the granzymes, the main effector proteases involved in NK cell cytotoxicity. All-trans retinoic acid (ATRA), which induces miR-23a expression, decreases CTSC expression and granzyme B activity leading to impaired NK cell cytotoxicity in an in vivo mouse model.


Subject(s)
Cathepsin C/genetics , Cytotoxicity, Immunologic/drug effects , Granzymes/genetics , Killer Cells, Natural/drug effects , MicroRNAs/genetics , Tretinoin/pharmacology , Animals , Blotting, Western , Cathepsin C/metabolism , Cell Line , Cells, Cultured , Cluster Analysis , Female , Granzymes/metabolism , Humans , Interleukin-2/pharmacology , Jurkat Cells , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome/drug effects , Transcriptome/genetics
16.
Anticancer Agents Med Chem ; 14(2): 223-32, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24237221

ABSTRACT

Tumor cells, including leukemic cells, remodel their bioenergetic system in favor of aerobic glycolysis. This process is called "the Warburg effect" and offers an attractive pharmacological target to preferentially eliminate malignant cells. In addition, recent results show that metabolic changes can be linked to tumor immune evasion. Mouse models demonstrate the importance of this metabolic remodeling in leukemogenesis. Some leukemias, although treatable, remain incurable and resistance to chemotherapy produces an elevated percentage of relapse in most leukemia cases. Several groups have targeted the specific metabolism of leukemia cells in preclinical and clinical studies to improve the prognosis of these patients, i.e. using L-asparaginase to treat pediatric acute lymphocytic leukemia (ALL). Additional metabolic drugs that are currently being used to treat other diseases or tumors could also be exploited for leukemia, based on preclinical studies. Finally, we discuss the potential use of several metabolic drugs in combination therapies, including immunomodulatory drugs (IMiDs) or immune cell-based therapies, to increase their efficacy and reduce side effects in the treatment of hematological cancers.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia/therapy , Animals , Cell Transplantation/methods , Combined Modality Therapy , Glutamine/metabolism , Glycolysis , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histone Deacetylases/metabolism , Humans , Immunomodulation , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Leukemia/drug therapy , Leukemia/immunology , Leukemia/metabolism , Oxidative Phosphorylation , Sirtuins/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation
17.
Int J Biochem Cell Biol ; 45(1): 106-13, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22568930

ABSTRACT

Tumorigenesis implies adaptation of tumor cells to an adverse environment. First, developing tumors must acquire nutrients to ensure their rapid growth. Second, they must escape the attack from the host immune system. Recent studies suggest that these phenomena could be related and that tumor cell metabolism may propel tumor immune escape. Tumor cell metabolism tends to avoid mitochondrial activity and oxidative phosphorylation (OXPHOS), and largely relies on glycolysis to produce energy. This specific metabolism helps tumor cells to avoid the immune attack from the host by blocking or avoiding the immune attack. By changing their metabolism, tumor cells produce or sequester a variety of amino acids, lipids and chemical compounds that directly alter immune function therefore promoting immune evasion. A second group of metabolism-related modification targets the major histocompatibility complex-I (MHC-I) and related molecules. Tumor MHC-I presents tumor-associated antigens (TAAs) to cytotoxic T-cells (CTLs) and hence, sensitizes cancer cells to the cytolytic actions of the anti-tumor adaptive immune response. Blocking tumor mitochondrial activity decreases expression of MHC-I molecules at the tumor cell surface. And peroxynitrite (PNT), produced by tumor-infiltrating myeloid cells, chemically modifies MHC-I avoiding TAA expression in the plasma membrane. These evidences on the role of tumor cell metabolism on tumor immune escape open the possibility of combining drugs designed to control tumor cell metabolism with new procedures of anti-tumor immunotherapy. This article is part of a Directed Issue entitled: Bioenergetic dysfunction, adaptation and therapy.


Subject(s)
Cell Transformation, Neoplastic/immunology , Neoplasms/immunology , Neoplasms/metabolism , Tumor Escape/immunology , Animals , Cell Transformation, Neoplastic/genetics , Humans , Neoplasms/genetics , Tumor Escape/genetics
SELECTION OF CITATIONS
SEARCH DETAIL