Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell ; 172(1-2): 147-161.e12, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29328910

ABSTRACT

Trained innate immunity fosters a sustained favorable response of myeloid cells to a secondary challenge, despite their short lifespan in circulation. We thus hypothesized that trained immunity acts via modulation of hematopoietic stem and progenitor cells (HSPCs). Administration of ß-glucan (prototypical trained-immunity-inducing agonist) to mice induced expansion of progenitors of the myeloid lineage, which was associated with elevated signaling by innate immune mediators, such as IL-1ß and granulocyte-macrophage colony-stimulating factor (GM-CSF), and with adaptations in glucose metabolism and cholesterol biosynthesis. The trained-immunity-related increase in myelopoiesis resulted in a beneficial response to secondary LPS challenge and protection from chemotherapy-induced myelosuppression in mice. Therefore, modulation of myeloid progenitors in the bone marrow is an integral component of trained immunity, which to date, was considered to involve functional changes of mature myeloid cells in the periphery.


Subject(s)
Immunity, Innate , Immunologic Memory , Myeloid Progenitor Cells/immunology , Animals , Cells, Cultured , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/drug effects , Myelopoiesis/immunology , beta-Glucans/pharmacology
2.
Nat Immunol ; 20(1): 40-49, 2019 01.
Article in English | MEDLINE | ID: mdl-30455459

ABSTRACT

Resolution of inflammation is essential for tissue homeostasis and represents a promising approach to inflammatory disorders. Here we found that developmental endothelial locus-1 (DEL-1), a secreted protein that inhibits leukocyte-endothelial adhesion and inflammation initiation, also functions as a non-redundant downstream effector in inflammation clearance. In human and mouse periodontitis, waning of inflammation was correlated with DEL-1 upregulation, whereas resolution of experimental periodontitis failed in DEL-1 deficiency. This concept was mechanistically substantiated in acute monosodium-urate-crystal-induced inflammation, where the pro-resolution function of DEL-1 was attributed to effective apoptotic neutrophil clearance (efferocytosis). DEL-1-mediated efferocytosis induced liver X receptor-dependent macrophage reprogramming to a pro-resolving phenotype and was required for optimal production of at least certain specific pro-resolving mediators. Experiments in transgenic mice with cell-specific overexpression of DEL-1 linked its anti-leukocyte-recruitment action to endothelial cell-derived DEL-1 and its efferocytic/pro-resolving action to macrophage-derived DEL-1. Thus, the compartmentalized expression of DEL-1 facilitates distinct homeostatic functions in an appropriate context that can be harnessed therapeutically.


Subject(s)
Carrier Proteins/metabolism , Inflammation/immunology , Macrophages/physiology , Neutrophils/immunology , Periodontitis/immunology , Adult , Animals , Calcium-Binding Proteins , Carrier Proteins/genetics , Cell Adhesion Molecules , Cellular Reprogramming , Cytokines/metabolism , Gene Expression Regulation , Humans , Inflammation/chemically induced , Intercellular Signaling Peptides and Proteins , K562 Cells , Liver X Receptors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis
3.
FASEB J ; 35 Suppl 12021 05.
Article in English | MEDLINE | ID: mdl-34318951

ABSTRACT

Withdrawal: Valeria Lopez Salazar, Rhoda Anane Karikari, Lun Li, Rabih El-Merahbi, Maria Troullinaki, Moya Wu, Tobias Wiedemann, Alina Walth, Manuel Gil Lozano, Maria Rohm, Stephan Herzig, Anastasia Georgiadi. Adipocyte Deletion of ADAM17 Leads to Insulin Resistance in Association with Age and HFD in Mice (2021). The FASEB Journal. 35:s1. doi: 10.1096/fasebj.2021.35.S1.00447. The above abstract, published online on May 14, 2021 in Wiley Online Library (wileyonlinelibrary.com), has been withdrawn by agreement between the authors, FASEB, and Wiley Periodicals Inc. The withdrawal is due to a request made by the authors prior to publication. The Publisher apologizes that this abstract was published in error.

4.
J Immunol ; 204(5): 1214-1224, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31980574

ABSTRACT

Leukocytes are rapidly recruited to sites of inflammation via interactions with the vascular endothelium. The steroid hormone dehydroepiandrosterone (DHEA) exerts anti-inflammatory properties; however, the underlying mechanisms are poorly understood. In this study, we show that an anti-inflammatory mechanism of DHEA involves the regulation of developmental endothelial locus 1 (DEL-1) expression. DEL-1 is a secreted homeostatic factor that inhibits ß2-integrin-dependent leukocyte adhesion, and the subsequent leukocyte recruitment and its expression is downregulated upon inflammation. Similarly, DHEA inhibited leukocyte adhesion to the endothelium in venules of the inflamed mouse cremaster muscle. Importantly, in a model of lung inflammation, DHEA limited neutrophil recruitment in a DEL-1-dependent manner. Mechanistically, DHEA counteracted the inhibitory effect of inflammation on DEL-1 expression. Indeed, whereas TNF reduced DEL-1 expression and secretion in endothelial cells by diminishing C/EBPß binding to the DEL-1 gene promoter, DHEA counteracted the inhibitory effect of TNF via activation of tropomyosin receptor kinase A (TRKA) and downstream PI3K/AKT signaling that restored C/EBPß binding to the DEL-1 promoter. In conclusion, DHEA restrains neutrophil recruitment by reversing inflammation-induced downregulation of DEL-1 expression. Therefore, the anti-inflammatory DHEA/DEL-1 axis could be harnessed therapeutically in the context of inflammatory diseases.


Subject(s)
Calcium-Binding Proteins/immunology , Cell Adhesion Molecules/immunology , Dehydroepiandrosterone/pharmacology , Leukocytes/immunology , Signal Transduction/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/immunology , CD18 Antigens/immunology , Cell Adhesion/immunology , Endothelium, Vascular/immunology , Female , Gene Expression Regulation/immunology , Leukocytes/cytology , Mice , Phosphatidylinositol 3-Kinases/immunology , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptor, trkA/immunology
5.
FASEB J ; 34(2): 3336-3346, 2020 02.
Article in English | MEDLINE | ID: mdl-31916652

ABSTRACT

In Type 1 Diabetes Mellitus (T1DM), leukocyte infiltration of the pancreatic islets and the resulting immune-mediated destruction of beta cells precede hyperglycemia and clinical disease symptoms. In this context, the role of the pancreatic endothelium as a barrier for autoimmunity- and inflammation-related destruction of the islets is not well studied. Here, we identified Robo4, expressed on endothelial cells, as a regulator of pancreatic vascular endothelial permeability during autoimmune diabetes. Circulating levels of Robo4 were upregulated in mice subjected to the Multiple Low-Dose Streptozotocin (MLDS) model of diabetes. Upon MLDS induction, Robo4-deficiency resulted in increased pancreatic vascular permeability, leukocyte infiltration to the islets and islet apoptosis, associated with reduced insulin levels and faster diabetes development. On the contrary, in vivo administration of Slit2 in mice modestly delayed the emergence of hyperglycaemia and ameliorated islet inflammation in MLDS-induced diabetes. Thus, Robo4-mediated endothelial barrier integrity reduces insulitis and islet destruction in autoimmune diabetes. Our findings highlight the importance of the endothelium as gatekeeper of pancreatic inflammation during T1DM development and may pave the way for novel Robo4-related therapeutic approaches for autoimmune diabetes.


Subject(s)
Capillary Permeability , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Endothelial Cells/metabolism , Insulin-Secreting Cells/metabolism , Receptors, Cell Surface/metabolism , Animals , Apoptosis , Cell Line , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Endothelial Cells/pathology , Humans , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/blood , Receptors, Cell Surface/genetics
6.
Exp Cell Res ; 377(1-2): 10-16, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30817930

ABSTRACT

Microglia, the parenchymal immune cells of the central nervous system, orchestrate neuroinflammation in response to infection or damage, and promote tissue repair. However, aberrant microglial responses are integral to neurodegenerative diseases and critically contribute to disease progression. Thus, it is important to elucidate how microglia - mediated neuroinflammation is regulated by endogenous factors. Here, we explored the effect of Nerve Growth Factor (NGF), an abundant neurotrophin, on microglial inflammatory responses. NGF, via its high affinity receptor TrkA, downregulated LPS - induced production of pro-inflammatory cytokines and NO in primary mouse microglia and inhibited TLR4 - mediated activation of the NF-κB and JNK pathways. Furthermore, NGF attenuated the LPS - enhanced glycolytic activity in microglia, as suggested by reduced glucose uptake and decreased expression of the glycolytic enzymes Pfkß3 and Ldhα. Consistently, 2DG - mediated glycolysis inhibition strongly downregulated LPS - induced cytokine production in microglial cells. Our findings demonstrate that NGF attenuates pro-inflammatory responses in microglia and may thereby contribute to regulation of microglia - mediated neuroinflammation.


Subject(s)
Glycolysis/drug effects , Inflammation/drug therapy , Lipopolysaccharides/toxicity , Microglia/drug effects , Nerve Growth Factor/pharmacology , Animals , Cytokines/genetics , Cytokines/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction
7.
J Cell Mol Med ; 23(4): 2362-2371, 2019 04.
Article in English | MEDLINE | ID: mdl-30680928

ABSTRACT

The mechanism underlying vasoproliferative retinopathies like retinopathy of prematurity (ROP) is hypoxia-triggered neovascularisation. Nerve growth factor (NGF), a neurotrophin supporting survival and differentiation of neuronal cells may also regulate endothelial cell functions. Here we studied the role of NGF in pathological retinal angiogenesis in the course of the ROP mouse model. Topical application of NGF enhanced while intraocular injections of anti-NGF neutralizing antibody reduced pathological retinal vascularization in mice subjected to the ROP model. The pro-angiogenic effect of NGF in the retina was mediated by inhibition of retinal endothelial cell apoptosis. In vitro, NGF decreased the intrinsic (mitochondria-dependent) apoptosis in hypoxia-treated human retinal microvascular endothelial cells and preserved the mitochondrial membrane potential. The anti-apoptotic effect of NGF was associated with increased BCL2 and reduced BAX, as well as with enhanced ERK and AKT phosphorylation, and was abolished by inhibition of the AKT pathway. Our findings reveal an anti-apoptotic role of NGF in the hypoxic retinal endothelium, which is involved in promoting pathological retinal vascularization, thereby pointing to NGF as a potential target for proliferative retinopathies.


Subject(s)
Antibodies, Neutralizing/pharmacology , Neovascularization, Pathologic/therapy , Nerve Growth Factor/antagonists & inhibitors , Retinopathy of Prematurity/therapy , Apoptosis/drug effects , Endothelial Cells , Humans , Injections, Intraocular , Membrane Potential, Mitochondrial/drug effects , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Nerve Growth Factor/genetics , Neurons/drug effects , Neurons/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Retina/drug effects , Retina/pathology , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/pathology , bcl-2-Associated X Protein/genetics
8.
Mol Metab ; 60: 101487, 2022 06.
Article in English | MEDLINE | ID: mdl-35378329

ABSTRACT

OBJECTIVE: Fibrotic organ responses have recently been identified as long-term complications in diabetes. Indeed, insulin resistance and aberrant hepatic lipid accumulation represent driving features of progressive non-alcoholic fatty liver disease (NAFLD), ranging from simple steatosis and non-alcoholic steatohepatitis (NASH) to fibrosis. Effective pharmacological regimens to stop progressive liver disease are still lacking to-date. METHODS: Based on our previous discovery of transforming growth factor beta-like stimulated clone (TSC)22D4 as a key driver of insulin resistance and glucose intolerance in obesity and type 2 diabetes, we generated a TSC22D4-hepatocyte specific knockout line (TSC22D4-HepaKO) and exposed mice to control or NASH diet models. Mechanistic insights were generated by metabolic phenotyping and single-nuclei RNA sequencing. RESULTS: Hepatic TSC22D4 expression was significantly correlated with markers of liver disease progression and fibrosis in both murine and human livers. Indeed, hepatic TSC22D4 levels were elevated in human NASH patients as well as in several murine NASH models. Specific genetic deletion of TSC22D4 in hepatocytes led to reduced liver lipid accumulation, improvements in steatosis and inflammation scores and decreased apoptosis in mice fed a lipogenic MCD diet. Single-nuclei RNA sequencing revealed a distinct TSC22D4-dependent gene signature identifying an upregulation of mitochondrial-related processes in hepatocytes upon loss of TSC22D4. An enrichment of genes involved in the TCA cycle, mitochondrial organization, and triglyceride metabolism underscored the hepatocyte-protective phenotype and overall decreased liver damage as seen in mouse models of hepatocyte-selective TSC22D4 loss-of-function. CONCLUSIONS: Together, our data uncover a new connection between targeted depletion of TSC22D4 and intrinsic metabolic processes in progressive liver disease. Hepatocyte-specific reduction of TSC22D4 improves hepatic steatosis and promotes hepatocyte survival via mitochondrial-related mechanisms thus paving the way for targeted therapies.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Non-alcoholic Fatty Liver Disease , Animals , Diabetes Mellitus, Type 2/metabolism , Fibrosis , Hepatocytes/metabolism , Humans , Lipids , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Transcription Factors/metabolism
9.
J Cell Biochem ; 112(12): 3786-96, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21826705

ABSTRACT

Recently, our knowledge on estrogen receptor alpha (ERα) functions and fate has progressed: ERα enters in repeated transcription-modulating cycles (nucleus/cytoplasm/membrane trafficking processes and proteasomal degradation) that are governed by specific protein-protein interactions. Receptor fragments, especially those resulting from the proteolysis of its ligand binding domain, as well as corresponding synthetic peptides, have been studied with respect to their estrogenic/antiestrogenic potency. A peptide, corresponding to the human ERα P(295) -T(311) sequence (ERα17p) has been shown to alter breast cancer cell fate, triggering proliferation, or apoptosis. The aim of this work was to explore the effect of ERα17p on breast cancer cell migration and actin cytoskeleton dynamics and further analyze the mechanism of its membrane action. We show that ERα17p increases (MCF-7 and SK-BR-3 cells) or decreases (T47D and MDA-MB-231 cells) migration of breast cancer cells, in an ERα-independent manner, by mechanism(s) depending on Rho/ROCK and PI3K/Akt signaling pathways. Moreover, the peptide enhances the association of both estrogens and androgens to membranes and modifies cell migration, induced by E(2) -BSA. Additionally, initial evidence of a possible agonistic action of the peptide on GPR30 is also provided. ERα17p can be considered as a cell migration-modulator and could therefore constitute a therapeutic challenge, even in anti-estrogen-resistant tumors.


Subject(s)
Actins/metabolism , Breast Neoplasms/pathology , Cytoskeleton/metabolism , Estrogen Receptor alpha/physiology , Neoplasm Metastasis/pathology , Biopolymers/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Female , Humans , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
10.
Cell Metab ; 33(8): 1685-1700.e9, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34237252

ABSTRACT

Liver fibrosis is a strong predictor of long-term mortality in individuals with metabolic-associated fatty liver disease; yet, the mechanisms underlying the progression from the comparatively benign fatty liver state to advanced non-alcoholic steatohepatitis (NASH) and liver fibrosis are incompletely understood. Using cell-type-resolved genomics, we show that comprehensive alterations in hepatocyte genomic and transcriptional settings during NASH progression, led to a loss of hepatocyte identity. The hepatocyte reprogramming was under tight cooperative control of a network of fibrosis-activated transcription factors, as exemplified by the transcription factor Elf-3 (ELF3) and zinc finger protein GLIS2 (GLIS2). Indeed, ELF3- and GLIS2-controlled fibrosis-dependent hepatokine genes targeting disease-associated hepatic stellate cell gene programs. Thus, interconnected transcription factor networks not only promoted hepatocyte dysfunction but also directed the intra-hepatic crosstalk necessary for NASH and fibrosis progression, implying that molecular "hub-centered" targeting strategies are superior to existing mono-target approaches as currently used in NASH therapy.


Subject(s)
Gene Regulatory Networks , Non-alcoholic Fatty Liver Disease , Communication , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver Cirrhosis/metabolism , Non-alcoholic Fatty Liver Disease/metabolism
11.
J Innate Immun ; 12(3): 248-256, 2020.
Article in English | MEDLINE | ID: mdl-31574508

ABSTRACT

Mononuclear phagocytes, such as macrophages and microglia, are key regulators of organ homeostasis including vascularization processes. Here, we investigated the role of the suppressor of cytokine signaling 3 (SOCS3) in myeloid cells as a regulator of mononuclear phagocyte function and their interaction with endothelial cells in the context of sprouting angiogenesis. As compared to SOCS3-sufficient counterparts, SOCS3-deficient microglia and macrophages displayed an increased phagocytic activity toward primary apoptotic endothelial cells, which was associated with an enhanced expression of the opsonin growth arrest-specific 6 (Gas6), a major prophagocytic molecule. Furthermore, we found that myeloid SOCS3 deficiency significantly reduced angiogenesis in an ex vivo mouse aortic ring assay, which could be reversed by the inhibition of the Gas6 receptor Mer. Together, SOCS3 in myeloid cells regulates the Gas6/Mer-dependent phagocytosis of endothelial cells, and thereby angiogenesis-related processes. Our findings provide novel insights into the complex crosstalk between mononuclear phagocytes and endothelial cells, and may therefore provide a new platform for the development of new antiangiogenic therapies.


Subject(s)
Apoptosis/immunology , Endothelial Cells/immunology , Myeloid Cells/immunology , Neovascularization, Physiologic/immunology , Suppressor of Cytokine Signaling 3 Protein/deficiency , Animals , Apoptosis/genetics , Mice , Mice, Transgenic , Neovascularization, Physiologic/genetics , Phagocytosis , Suppressor of Cytokine Signaling 3 Protein/immunology
12.
Thromb Haemost ; 119(3): 439-448, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30620991

ABSTRACT

The replication stress inflicted on retinal endothelial cells (ECs) in the context of hypoxia-induced pathological neovascularization during proliferative retinopathy is linked with activation of the deoxyribonucleic acid (DNA) repair response. Here, we studied the effect of deficiency of the DNA damage response adaptor 53BP1, which is an antagonist of homologous recombination (HR), in the context of proliferative retinopathy. In the model of retinopathy of prematurity (ROP), 53BP1-deficient mice displayed increased hypoxia-driven pathological neovascularization and tuft formation, accompanied by increased EC proliferation and reduced EC apoptosis, as compared with 53BP1-sufficient mice. In contrast, physiological retina angiogenesis was not affected by 53BP1 deficiency. Knockdown of 53BP1 in ECs in vitro also resulted in enhanced proliferation and reduced apoptosis of the cells under hypoxic conditions. Additionally, upon 53BP1 knockdown, ECs displayed increased HR rate in hypoxia. Consistently, treatment with an HR inhibitor reversed the hyper-proliferative angiogenic phenotype associated with 53BP1 deficiency in ROP. Thus, by unleashing HR, 53BP1 deletion increases pathological EC proliferation and neovascularization in the context of ROP. Our data shed light to a previously unknown interaction between the DNA repair response and pathological neovascularization in the retina.


Subject(s)
Cell Proliferation , Endothelial Cells/metabolism , Homologous Recombination , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Retinopathy of Prematurity/metabolism , Tumor Suppressor p53-Binding Protein 1/deficiency , Tumor Suppressor p53-Binding Protein 1/metabolism , Animals , Apoptosis , Cell Hypoxia , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Genetic Predisposition to Disease , Homologous Recombination/drug effects , Humans , Mice, Knockout , Morpholines/pharmacology , Phenotype , Pyrroles/pharmacology , Retinal Neovascularization/genetics , Retinal Neovascularization/pathology , Retinal Neovascularization/prevention & control , Retinal Vessels/drug effects , Retinal Vessels/pathology , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/prevention & control , Signal Transduction , Tumor Suppressor p53-Binding Protein 1/genetics
13.
Sci Rep ; 8(1): 11335, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054579

ABSTRACT

Diabetes mellitus is a group of disorders characterized by prolonged high levels of circulating blood glucose. Type 1 diabetes is caused by decreased insulin production in the pancreas whereas type 2 diabetes may develop due to obesity and lack of exercise; it begins with insulin resistance whereby cells fail to respond properly to insulin and it may also progress to decreased insulin levels. The brain is an important target for insulin, and there is great interest in understanding how diabetes affects the brain. In addition to the direct effects of insulin on the brain, diabetes may also impact the brain through modulation of the inflammatory system. Here we investigate how perturbation of circulating insulin levels affects the expression of Hes3, a transcription factor expressed in neural stem and progenitor cells that is involved in tissue regeneration. Our data show that streptozotocin-induced ß-cell damage, high fat diet, as well as metformin, a common type 2 diabetes medication, regulate Hes3 levels in the brain. This work suggests that Hes3 is a valuable biomarker helping to monitor the state of endogenous neural stem and progenitor cells in the context of diabetes mellitus.


Subject(s)
Aging/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/metabolism , Diet, High-Fat , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Metformin/administration & dosage , Nerve Tissue Proteins/metabolism , Streptozocin/toxicity , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/drug effects , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Phenotype , Repressor Proteins
14.
Thromb Haemost ; 117(6): 1150-1163, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28447099

ABSTRACT

We have recently identified endothelial cell-secreted developmental endothelial locus-1 (Del-1) as an endogenous inhibitor of ß2-integrin-dependent leukocyte infiltration. Del-1 was previously also implicated in angiogenesis. Here, we addressed the role of endogenously produced Del-1 in ischaemia-related angiogenesis. Intriguingly, Del-1-deficient mice displayed increased neovascularisation in two independent ischaemic models (retinopathy of prematurity and hind-limb ischaemia), as compared to Del-1-proficient mice. On the contrary, angiogenic sprouting in vitro or ex vivo (aortic ring assay) and physiological developmental retina angiogenesis were not affected by Del-1 deficiency. Mechanistically, the enhanced ischaemic neovascularisation in Del-1-deficiency was linked to higher infiltration of the ischaemic tissue by CD45+ haematopoietic and immune cells. Moreover, Del-1-deficiency promoted ß2-integrin-dependent adhesion of haematopoietic cells to endothelial cells in vitro, and the homing of hematopoietic progenitor cells and of immune cell populations to ischaemic muscles in vivo. Consistently, the increased hind limb ischaemia-related angiogenesis in Del-1 deficiency was completely reversed in mice lacking both Del-1 and the ß2-integrin LFA-1. Additionally, enhanced retinopathy-associated neovascularisation in Del-1-deficient mice was reversed by LFA-1 blockade. Our data reveal a hitherto unrecognised function of endogenous Del-1 as a local inhibitor of ischaemia-induced angiogenesis by restraining LFA-1-dependent homing of pro-angiogenic haematopoietic cells to ischaemic tissues. Our findings are relevant for the optimisation of therapeutic approaches in the context of ischaemic diseases.


Subject(s)
Carrier Proteins/metabolism , Endothelium, Vascular/physiology , Hematopoietic Stem Cells/physiology , Inflammation/metabolism , Ischemia/metabolism , Leukocytes/physiology , Retinopathy of Prematurity/metabolism , Animals , Calcium-Binding Proteins , Carrier Proteins/genetics , Cell Adhesion , Cell Adhesion Molecules , Cell Movement , Disease Models, Animal , Extremities/pathology , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/immunology , Intercellular Signaling Peptides and Proteins , Ischemia/immunology , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Knockout , Neovascularization, Physiologic , RNA, Small Interfering/genetics , Retinopathy of Prematurity/immunology
15.
J Clin Invest ; 127(10): 3624-3639, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28846069

ABSTRACT

Hematopoietic stem cells (HSCs) remain mostly quiescent under steady-state conditions but switch to a proliferative state following hematopoietic stress, e.g., bone marrow (BM) injury, transplantation, or systemic infection and inflammation. The homeostatic balance between quiescence, self-renewal, and differentiation of HSCs is strongly dependent on their interactions with cells that constitute a specialized microanatomical environment in the BM known as the HSC niche. Here, we identified the secreted extracellular matrix protein Del-1 as a component and regulator of the HSC niche. Specifically, we found that Del-1 was expressed by several cellular components of the HSC niche, including arteriolar endothelial cells, CXCL12-abundant reticular (CAR) cells, and cells of the osteoblastic lineage. Del-1 promoted critical functions of the HSC niche, as it regulated long-term HSC (LT-HSC) proliferation and differentiation toward the myeloid lineage. Del-1 deficiency in mice resulted in reduced LT-HSC proliferation and infringed preferentially upon myelopoiesis under both steady-state and stressful conditions, such as hematopoietic cell transplantation and G-CSF- or inflammation-induced stress myelopoiesis. Del-1-induced HSC proliferation and myeloid lineage commitment were mediated by ß3 integrin on hematopoietic progenitors. This hitherto unknown Del-1 function in the HSC niche represents a juxtacrine homeostatic adaptation of the hematopoietic system in stress myelopoiesis.


Subject(s)
Carrier Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Myelopoiesis , Stem Cell Niche , Stress, Physiological , Animals , Calcium-Binding Proteins , Carrier Proteins/genetics , Cell Adhesion Molecules , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Endothelial Cells/metabolism , Humans , Integrin beta3/genetics , Integrin beta3/metabolism , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout
16.
Brain Res ; 1642: 124-130, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27018293

ABSTRACT

Hes3 is a component of the STAT3-Ser/Hes3 Signaling Axis controlling the growth and survival of neural stem cells and other plastic cells. Pharmacological activation of this pathway promotes neuronal rescue and behavioral recovery in models of ischemic stroke and Parkinson's disease. Here we provide initial observations implicating Hes3 in the cuprizone model of demyelination and remyelination. We focus on the subpial motor cortex of mice because we detected high Hes3 expression. This area is of interest as it is impacted both in human demyelinating diseases and in the cuprizone model. We report that Hes3 expression is reduced at peak demyelination and is partially restored within 1 week after cuprizone withdrawal. This raises the possibility of Hes3 involvement in demyelination/remyelination that may warrant additional research. Supporting a possible role of Hes3 in the maintenance of oligodendrocyte markers, a Hes3 null mouse strain shows lower levels of myelin basic protein in undamaged adult mice, compared to wild-type controls. We also present a novel method for culturing the established oligodendrocyte progenitor cell line oli-neu in a manner that maintains Hes3 expression as well as its self-renewal and differentiation potential, offering an experimental tool to study Hes3. Based upon this approach, we identify a Janus kinase inhibitor and dbcAMP as powerful inducers of Hes3 gene expression. We provide a new biomarker and cell culture method that may be of interest in demyelination/remyelination research.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Demyelinating Diseases/genetics , Gene Expression Regulation , Motor Cortex/metabolism , Myelin Sheath/genetics , Nerve Tissue Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Culture Techniques , Culture Media, Conditioned , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , Repressor Proteins
17.
Thromb Haemost ; 114(6): 1241-9, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26311310

ABSTRACT

In proliferative retinopathies, like proliferative diabetic retinopathy and retinopathy of prematurity (ROP), the hypoxia response is sustained by the failure of the retina to revascularise its ischaemic areas. Non-resolving retina ischaemia/hypoxia results in upregulation of pro-angiogenic factors and pathologic neovascularisation with ectopic, fragile neovessels. Promoting revascularisation of the retinal avascular area could interfere with this vicious cycle and lead to vessel normalisation. Here, we examined the function of endothelial junctional adhesion molecule-C (JAM-C) in the context of ROP. Endothelial-specific JAM-C-deficient (EC-JAM-C KO) mice and littermate JAM-C-proficient (EC-JAM-C WT) mice were subjected to the ROP model. An increase in total retinal vascularisation was found at p17 owing to endothelial JAM-C deficiency, which was the result of enhanced revascularisation and vessel normalisation, thereby leading to significantly reduced avascular area in EC-JAM-C KO mice. In contrast, pathologic neovessel formation was not affected by endothelial JAM-C deficiency. Consistent with improved vessel normalisation, tip cell formation at the interface between vascular and avascular area was higher in EC-JAM-C KO mice, as compared to their littermate controls. Consistently, JAM-C inactivation in endothelial cells resulted in increased spreading on fibronectin and enhanced sprouting in vitro in a manner dependent on ß1-integrin and on the activation of the small GTPase RAP1. Together, endothelial deletion of JAM-C promoted endothelial cell sprouting, and consequently vessel normalisation and revascularisation of the hypoxic retina without altering pathologic neovascularisation. Thus, targeting endothelial JAM-C may provide a novel therapeutic strategy for promoting revascularisation and vessel normalisation in the treatment of proliferative retinopathies.


Subject(s)
Endothelium, Vascular/physiopathology , Junctional Adhesion Molecule C/deficiency , Neovascularization, Pathologic/physiopathology , Retinal Vessels/physiopathology , Retinopathy of Prematurity/physiopathology , Vitreoretinopathy, Proliferative/physiopathology , Animals , Cell Adhesion , Cell Hypoxia , Cell Line , Cell Size , Cell Surface Extensions , Disease Models, Animal , Endothelial Cells , Endothelium, Vascular/pathology , Fibronectins , Human Umbilical Vein Endothelial Cells , Humans , Integrin beta1/physiology , Ischemia/physiopathology , Junctional Adhesion Molecule C/physiology , Mice , Mice, Knockout , Neovascularization, Pathologic/etiology , Organ Specificity , Platelet Endothelial Cell Adhesion Molecule-1/analysis , RNA Interference , RNA, Small Interfering/genetics , Retinal Vessels/ultrastructure , rap1 GTP-Binding Proteins/physiology
18.
Mol Oncol ; 7(3): 595-610, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23474223

ABSTRACT

ERα17p is a peptide corresponding to the sequence P295LMIKRSKKNSLALSLT311 of the estrogen receptor alpha (ERα) and initially found to interfere with ERα-related calmodulin binding. ERα17p was subsequently found to elicit estrogenic responses in E2-deprived ERα-positive breast cancer cells, increasing proliferation and ERE-dependent gene transcription. Surprisingly, in E2-supplemented media, ERα17p-induced apoptosis and modified the actin network, influencing cell motility. Here, we report that ERα17p internalizes in breast cancer cells (T47D, MDA-MB-231, SKBR3) and induces a massive early (3 h) transcriptional activity. Remarkably, about 75% of significantly modified transcripts were also modified by E2, confirming the pro-estrogenic profile of ERα17p. The different ER spectra of the used cell lines allowed us to identify a specific ERα17p signature related to ERα as well as its variant ERα36. With respect to ERα, the peptide activates nuclear (cell cycle, cell proliferation, nucleic acid and protein synthesis) and extranuclear signaling pathways. In contrast, through ERα36, it mainly triggers inhibitory actions on inflammation. This is the first work reporting a detailed ERα36-specific transcriptional signature. In addition, we report that ERα17p-induced transcripts related to apoptosis and actin modifying effects of the peptide are independent from its estrogen receptor(s)-related actions. We discuss our findings in view of the potential use of ERα17p as a selective peptidomimetic estrogen receptor modulator (PERM).


Subject(s)
Breast Neoplasms/genetics , Breast/metabolism , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Gene Expression Profiling , Amino Acid Sequence , Apoptosis , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Movement , Estradiol/metabolism , Female , Humans , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcriptional Activation
19.
Steroids ; 77(10): 959-67, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22406407

ABSTRACT

The complexity of estrogen actions mainly relies to the presence of different identified receptors (ERα, ERß, their isoforms, and GPR30/GPER) and their discrete cellular distribution. Depending on the localization of the receptor that mediates estrogen effects, nuclear and extra-nuclear actions have been described. The latter can trigger a number of signaling events leading also to transcriptional modifications. In an attempt to clarify the nature of the receptor(s) involved in the membrane initiated effect of estrogens on gene expression, we performed a whole transcriptome analysis of breast cancer cell lines with different receptor profiles (T47D, MCF7, MDA-MB-231, SK-BR-3). A pharmacological approach was conducted with the use of estradiol (E(2)) or membrane-impermeable E(2)-BSA in the absence or presence of a specific ERα-ß or GPR30/GPER antagonist. Our results clearly show that in addition to the ERα isoforms and/or GPR30/GPER that mainly mediate the transcriptional effect of E(2)-BSA, there is a specific transcriptional signature (found in T47D and MCF-7 cells) suggesting the presence of an unidentified membrane ER element (ERx). Analysis of its signature and phenotypic verification revealed that important cell function such as apoptosis, transcriptional regulation, and growth factor signaling are associated with ERx.


Subject(s)
Estradiol/pharmacology , Estrogens/pharmacology , Receptors, Estrogen/metabolism , Serum Albumin, Bovine/pharmacology , Transcriptome , Apoptosis , Breast Neoplasms , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Estradiol/analogs & derivatives , Female , Fulvestrant , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Oligonucleotide Array Sequence Analysis , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Estrogen/antagonists & inhibitors , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL