Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Nature ; 629(8011): 417-425, 2024 May.
Article En | MEDLINE | ID: mdl-38658748

Cancer-specific TCF1+ stem-like CD8+ T cells can drive protective anticancer immunity through expansion and effector cell differentiation1-4; however, this response is dysfunctional in tumours. Current cancer immunotherapies2,5-9 can promote anticancer responses through TCF1+ stem-like CD8+ T cells in some but not all patients. This variation points towards currently ill-defined mechanisms that limit TCF1+CD8+ T cell-mediated anticancer immunity. Here we demonstrate that tumour-derived prostaglandin E2 (PGE2) restricts the proliferative expansion and effector differentiation of TCF1+CD8+ T cells within tumours, which promotes cancer immune escape. PGE2 does not affect the priming of TCF1+CD8+ T cells in draining lymph nodes. PGE2 acts through EP2 and EP4 (EP2/EP4) receptor signalling in CD8+ T cells to limit the intratumoural generation of early and late effector T cell populations that originate from TCF1+ tumour-infiltrating CD8+ T lymphocytes (TILs). Ablation of EP2/EP4 signalling in cancer-specific CD8+ T cells rescues their expansion and effector differentiation within tumours and leads to tumour elimination in multiple mouse cancer models. Mechanistically, suppression of the interleukin-2 (IL-2) signalling pathway underlies the PGE2-mediated inhibition of TCF1+ TIL responses. Altogether, we uncover a key mechanism that restricts the IL-2 responsiveness of TCF1+ TILs and prevents anticancer T cell responses that originate from these cells. This study identifies the PGE2-EP2/EP4 axis as a molecular target to restore IL-2 responsiveness in anticancer TILs to achieve cancer immune control.


CD8-Positive T-Lymphocytes , Cell Proliferation , Dinoprostone , Lymphocytes, Tumor-Infiltrating , Neoplasms , Stem Cells , Tumor Escape , Animals , Female , Humans , Male , Mice , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Line, Tumor , Dinoprostone/metabolism , Disease Models, Animal , Hepatocyte Nuclear Factor 1-alpha/metabolism , Interleukin-2 , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/prevention & control , Receptors, Prostaglandin E, EP2 Subtype/deficiency , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/deficiency , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Tumor Escape/immunology
2.
Nature ; 590(7844): 122-128, 2021 02.
Article En | MEDLINE | ID: mdl-33473210

Ageing is characterized by the development of persistent pro-inflammatory responses that contribute to atherosclerosis, metabolic syndrome, cancer and frailty1-3. The ageing brain is also vulnerable to inflammation, as demonstrated by the high prevalence of age-associated cognitive decline and Alzheimer's disease4-6. Systemically, circulating pro-inflammatory factors can promote cognitive decline7,8, and in the brain, microglia lose the ability to clear misfolded proteins that are associated with neurodegeneration9,10. However, the underlying mechanisms that initiate and sustain maladaptive inflammation with ageing are not well defined. Here we show that in ageing mice myeloid cell bioenergetics are suppressed in response to increased signalling by the lipid messenger prostaglandin E2 (PGE2), a major modulator of inflammation11. In ageing macrophages and microglia, PGE2 signalling through its EP2 receptor promotes the sequestration of glucose into glycogen, reducing glucose flux and mitochondrial respiration. This energy-deficient state, which drives maladaptive pro-inflammatory responses, is further augmented by a dependence of aged myeloid cells on glucose as a principal fuel source. In aged mice, inhibition of myeloid EP2 signalling rejuvenates cellular bioenergetics, systemic and brain inflammatory states, hippocampal synaptic plasticity and spatial memory. Moreover, blockade of peripheral myeloid EP2 signalling is sufficient to restore cognition in aged mice. Our study suggests that cognitive ageing is not a static or irrevocable condition but can be reversed by reprogramming myeloid glucose metabolism to restore youthful immune functions.


Aging/metabolism , Cognitive Dysfunction/prevention & control , Myeloid Cells/metabolism , Adult , Aged , Aging/drug effects , Aging/genetics , Animals , Cell Respiration , Cells, Cultured , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/genetics , Dinoprostone/metabolism , Energy Metabolism , Glucose/metabolism , Glycogen/biosynthesis , Glycogen/metabolism , Humans , Inflammation/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Memory Disorders/drug therapy , Mice , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Mitochondria/metabolism , Myeloid Cells/immunology , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/deficiency , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Signal Transduction/drug effects , Spatial Memory/drug effects
3.
CNS Neurosci Ther ; 26(6): 628-635, 2020 06.
Article En | MEDLINE | ID: mdl-31617678

AIMS: The goal was to explore the signaling pathways of PGE2 to investigate therapeutic effects against secondary injuries following TBI. METHODS: Young (4.9 ± 1.0 months) and aged (20.4 ± 1.4 months) male wild type (WT) C57BL/6 and PGE2 EP1, 2, and 3 receptor knockout mice were selected to either receive sham or repetitive concussive head injury. Immunohistochemistry protocols with Iba1 and GFAP were performed to evaluate microgliosis and astrogliosis in the hippocampus, two critical components of neuroinflammation. Passive avoidance test measured memory function associated with the hippocampus. RESULTS: No differences in hippocampal microgliosis were found when aged EP2-/- and EP3-/- mice were compared with aged WT mice. However, the aged EP1-/- mice had 69.2 ± 7.5% less hippocampal microgliosis in the contralateral hemisphere compared with WT aged mice. Compared with aged EP2-/- and EP3-/- , EP1-/- aged mice had 78.9 ± 5.1% and 74.7 ± 6.2% less hippocampal microgliosis in the contralateral hemisphere. Within the EP1-/- mice, aged mice had 90.7 ± 2.7% and 81.1 ± 5.6% less hippocampal microgliosis compared with EP1-/- young mice in the contralateral and ipsilateral hemispheres, respectively. No differences were noted in all groups for astrogliosis. There was a significant difference in latency time within EP1-/- , EP2-/- , and EP3-/- on day 1 and day 2 in aged and young mice. CONCLUSION: These findings demonstrate that the PGE2 EP receptors may be potential therapeutic targets to treat repetitive concussions and other acute brain injuries.


Brain Injuries, Traumatic/metabolism , Receptors, Prostaglandin E, EP1 Subtype/deficiency , Receptors, Prostaglandin E, EP2 Subtype/deficiency , Receptors, Prostaglandin E, EP3 Subtype/deficiency , Animals , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Prostaglandin E/deficiency , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP1 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP3 Subtype/genetics
4.
Sci Rep ; 6: 20505, 2016 Feb 08.
Article En | MEDLINE | ID: mdl-26852804

Prostaglandins E2 (PGE2) has been shown to enhance IgE production by B cells in vitro. The physiological and pathological relevance of this phenomenon and the underlying molecular mechanism, however, remain to be elucidated. B cells from wild type and EP2-deficient mice were compared in culture for their responses to PGE2 in terms of IgE class switching and production. Ovalbumin (OVA)-induced asthma models were used to evaluate the impact of EP2-deficiency on IgE responses and the development of asthma. PGE2 promoted IgE class switching, generation of IgE(+) cells and secretion of IgE by B cells stimulated with LPS+IL4. These effects were much attenuated as a consequence of EP2 deficiency. Consistent with the in vitro data, EP2-deficient mice showed a markedly suppressed IgE antibody response and developed less pronounced airway inflammation in the OVA-induced asthma model. Mechanistic studies demonstrated that PGE2, in an EP2-depedent manner, enhanced STAT6 activation induced by IL-4, thereby promoting the expression of IgE germline and post switch transcripts and the transcription of activation-induced cytidine deaminase (AID). Collectively, these data support an important regulatory role of the PGE2-EP2-STAT6 signaling pathway in IgE response and allergic diseases.


Asthma/pathology , Dinoprostone/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Animals , Antibody Formation/drug effects , Asthma/immunology , Asthma/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Cytidine Deaminase/metabolism , Disease Models, Animal , Immunoglobulin E/analysis , Immunoglobulin E/metabolism , Interleukin-4/pharmacology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Phosphorylation/drug effects , Receptors, IgE/antagonists & inhibitors , Receptors, IgE/metabolism , Receptors, Prostaglandin E, EP2 Subtype/deficiency , Receptors, Prostaglandin E, EP2 Subtype/genetics , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects
5.
ASN Neuro ; 7(2)2015.
Article En | MEDLINE | ID: mdl-25873308

Intracerebral hemorrhage (ICH) is a devastating type of stroke characterized by bleeding into the brain parenchyma and secondary brain injury resulting from strong neuroinflammatory responses to blood components. Production of prostaglandin E2 (PGE2) is significantly upregulated following ICH and contributes to this inflammatory response in part through its E prostanoid receptor subtype 2 (EP2). Signaling through the EP2 receptor has been shown to affect outcomes of many acute and chronic neurological disorders; although, not yet explored in the context of ICH. Wildtype (WT) and EP2 receptor knockout (EP2(-/-)) mice were subjected to ICH, and various anatomical and functional outcomes were assessed by histology and neurobehavioral testing, respectively. When compared with age-matched WT controls, EP2(-/-) mice had 41.9 ± 4.7% smaller ICH-induced brain lesions and displayed significantly less ipsilateral hemispheric enlargement and incidence of intraventricular hemorrhage. Anatomical outcomes correlated with improved functional recovery as identified by neurological deficit scoring. Histological staining was performed to begin investigating the mechanisms involved in EP2-mediated neurotoxicity after ICH. EP2(-/-) mice exhibited 45.5 ± 5.8% and 41.4 ± 8.1% less blood and ferric iron accumulation, respectively. Furthermore, significantly less striatal and cortical microgliosis, striatal and cortical astrogliosis, blood-brain barrier breakdown, and peripheral neutrophil infiltration were seen in EP2(-/-) mice. This study is the first to suggest a deleterious role for the PGE2-EP2 signaling axis in modulating brain injury, inflammation, and functional recovery following ICH. Targeting the EP2 G protein-coupled receptor may represent a new therapeutic avenue for the treatment of hemorrhagic stroke.


Brain/physiopathology , Cerebral Hemorrhage/physiopathology , Receptors, Prostaglandin E, EP2 Subtype/deficiency , Animals , Astrocytes/pathology , Astrocytes/physiology , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain/pathology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/pathology , Disease Models, Animal , Gliosis/pathology , Gliosis/physiopathology , Iron/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Microglia/physiology , Neuroimmunomodulation/physiology , Neutrophil Infiltration/physiology , Receptors, Prostaglandin E, EP2 Subtype/genetics , Recovery of Function/physiology
6.
Bone ; 56(1): 31-41, 2013 Sep.
Article En | MEDLINE | ID: mdl-23639875

Intermittent PTH is the major anabolic therapy for osteoporosis while continuous PTH causes bone loss. PTH acts on the osteoblast (OB) lineage to regulate bone resorption and formation. PTH also induces cyclooxygenase-2 (COX-2), producing prostaglandin E2 (PGE(2)) that can act on both OBs and osteoclasts (OCs). Because intermittent PTH is more anabolic in Cox-2 knockout (KO) than wild type (WT) mice, we hypothesized COX-2 might contribute to the effects of continuous PTH by suppressing PTH-stimulated differentiation of mesenchymal stem cells into OBs. We compared effects of continuous PTH on bone marrow stromal cells (BMSCs) and primary OBs (POBs) from Cox-2 KO mice, mice with deletion of PGE(2) receptors (Ptger(4) and Ptger(2) KO mice), and WT controls. PTH increased OB differentiation in BMSCs only in the absence of COX-2 expression or activity. In the absence of COX-2, PTH stimulated differentiation if added during the first week of culture. In Cox-2 KO BMSCs, PTH-stimulated differentiation was prevented by adding PGE(2) to cultures. Co-culture of POBs with M-CSF-expanded bone marrow macrophages (BMMs) showed that the inhibition of PTH-stimulated OB differentiation required not only COX-2 or PGE(2) but also BMMs. Sufficient PGE(2) to mediate the inhibitory effect was made by either WT POBs or WT BMMs. The inhibitory effect mediated by COX-2/PGE(2) was transferred by conditioned media from RANKL-treated BMMs and could be blocked by osteoprotegerin, which interferes with RANKL binding to its receptor on OC lineage cells. Deletion of Ptger(4), but not Ptger(2), in BMMs prevented the inhibition of PTH-stimulated OB differentiation. As expected, PGE(2) also stimulated OB differentiation, but when given in combination with PTH, the stimulatory effects of both were abrogated. These data suggest that PGE(2), acting via EP4R on BMMs committed to the OC lineage, stimulated secretion of a factor or factors that acted to suppress PTH-stimulated OB differentiation. This suppression of OB differentiation could contribute to the bone loss seen with continuous PTH in vivo.


Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Dinoprostone/pharmacology , Macrophages/metabolism , Osteoblasts/cytology , Parathyroid Hormone/pharmacology , Adipogenesis/drug effects , Adipogenesis/genetics , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cattle , Cell Differentiation/genetics , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cyclooxygenase 2/metabolism , Gene Expression Regulation/drug effects , Hematopoietic System/cytology , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Knockout , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteocalcin/genetics , Osteocalcin/metabolism , RANK Ligand/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prostaglandin E, EP2 Subtype/deficiency , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/deficiency , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/enzymology
7.
Am J Physiol Renal Physiol ; 303(2): F209-19, 2012 Jul 15.
Article En | MEDLINE | ID: mdl-22573380

Pharmacological blockade of cyclooxygenase-2 (COX-2) causes impairment of kidney development. The present study was aimed at determining temporal expression pattern and activity of the PGE(2) synthetic pathway during postnatal nephrogenesis in mice and its association to the time window sensitive to COX-2 inhibition. During the first 10 days after birth, we observed transient induction of mRNA and protein for microsomal PGE synthase (mPGES)-1 between postnatal days 4 (P4) and P8, but not for mPGES-2 or cytosolic PGE synthase (cPGES). PGE(2) synthetic activity using arachidonic acid and PGH(2) as substrates and also urinary excretion of PGE(2) were enhanced during this time frame. In parallel to the PGE(2) system, COX-2 but not COX-1 expression was also transiently induced. Studying glomerulogenesis in EP receptor knockout mice revealed a reduction in glomerular size in EP1(-/-), EP2(-/-), and EP4(-/-) mice, supporting the developmental role of PGE(2). The most vulnerable time window to COX-2 inhibition by SC-236 was found closely related to the temporal expression of COX-2 and mPGES-1. The strongest effects of COX-2 inhibition were achieved following 8 days of drug administration. Similar developmental damage was caused by application of rofecoxib, but not by the COX-1-selective inhibitor SC-560. COX-2 inhibition starting after P10 has had no effect on the size of glomeruli or on the relative number of superficial glomeruli; however, growth of the renal cortex was significantly diminished, indicating the requirement of COX-2 activity after P10. Effects of COX-2 inhibition on renal cell differentiation and on renal fibrosis needed a prolonged time of exposition of at least 10 days. In conclusion, temporal expression of the PGE(2) synthetic system coincides with the most vulnerable age interval for the induction of irreversible renal abnormalities. We assume that mPGES-1 is coregulated with COX-2 for PGE(2) synthesis to orchestrate postnatal kidney development and growth.


Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/drug effects , Dinoprostone/metabolism , Intramolecular Oxidoreductases/metabolism , Kidney/growth & development , Kidney/metabolism , Animals , Cyclooxygenase 1/drug effects , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Female , Kidney/drug effects , Lactones/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Prostaglandin-E Synthases , Pyrazoles/pharmacology , Receptors, Prostaglandin E, EP1 Subtype/deficiency , Receptors, Prostaglandin E, EP1 Subtype/genetics , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Receptors, Prostaglandin E, EP2 Subtype/deficiency , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/deficiency , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Sulfones/pharmacology , Time Factors
8.
Arterioscler Thromb Vasc Biol ; 31(8): 1739-47, 2011 Aug.
Article En | MEDLINE | ID: mdl-21636806

OBJECTIVE: Restenosis after angioplasty remains a major clinical problem. Prostaglandin E(2) (PGE(2)) plays an important role in vascular homeostasis. The PGE(2) receptor E-prostanoid 2 (EP2) is involved in the proliferation and migration of various cell types. We aimed to determine the role of EP2 in the pathogenesis of neointimal formation after vascular injury. METHODS AND RESULTS: Wire-mediated vascular injury was induced in the femoral arteries of male wild-type (EP2+/+) and EP2 gene-deficient (EP2-/-) mice. In EP2+/+ mice, EP2 mRNA expression was increased in injured vessels for at least 4 weeks after vascular injury. Neointimal hyperplasia was markedly accelerated in EP2-/- mice, which was associated with increased proliferation and migration of vascular smooth muscle cells (VSMCs) and increased cyclin D1 expression in the neointima layer. Platelet-derived growth factor-BB (PDGF-BB) treatment resulted in more significant cell proliferation and migration in VSMCs of EP2-/- mice than in those of EP2+/+ mice. Activation and overexpression of EP2 attenuated PDGF-BB-elicited cell proliferation and migration, induced G(1)→S-phase arrest and reduced PDGF-BB-stimulated extracellular signal-regulated kinase phosphorylation in EP2+/+ VSMCs. CONCLUSIONS: These findings reveal a novel role of the EP2 receptor in neointimal hyperplasia after arterial injury. The EP2 receptor may represent a potential therapeutic target for restenosis after angioplasty.


Neointima/etiology , Receptors, Prostaglandin E, EP2 Subtype/physiology , Animals , Becaplermin , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coronary Restenosis/etiology , Coronary Restenosis/pathology , Coronary Restenosis/physiopathology , Disease Models, Animal , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , Gene Expression , Humans , Hyperplasia , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/physiology , Neointima/genetics , Neointima/metabolism , Neointima/pathology , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/deficiency , Receptors, Prostaglandin E, EP2 Subtype/genetics
...