Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 14(3): 217, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36977701

ABSTRACT

Atypically expressed transglutaminase 2 (TG2) has been identified as a poor prognostic factor in a variety of cancers. In this study, we evaluated the contribution of TG2 to the prolonged cell survival of differentiated acute promyelocytic leukaemia (APL) cells in response to the standard treatment with combined retinoic acid (ATRA) and arsenic trioxide (ATO). We report that one advantage of ATRA + ATO treatment compared to ATRA alone diminishes the amount of activated and non-activated CD11b/CD18 and CD11c/CD18 cell surface integrin receptors. These changes suppress ATRA-induced TG2 docking on the cytosolic part of CD18 ß2-integrin subunits and reduce cell survival. In addition, TG2 overexpresses and hyperactivates the phosphatidylinositol-3-kinase (PI3K), phospho-AKT S473, and phospho-mTOR S2481 signalling axis. mTORC2 acts as a functional switch between cell survival and death by promoting the full activation of AKT. We show that TG2 presumably triggers the formation of a signalosome platform, hyperactivates downstream mTORC2-AKT signalling, which in turn phosphorylates and inhibits the activity of FOXO3, a key pro-apoptotic transcription factor. In contrast, the absence of TG2 restores basic phospho-mTOR S2481, phospho-AKT S473, PI3K, and PTEN expression and activity, thereby sensitising APL cells to ATO-induced cell death. We conclude, that atypically expressed TG2 may serve as a hub, facilitating signal transduction via signalosome formation by the CD18 subunit with both PI3K hyperactivation and PTEN inactivation through the PI3K-PTEN cycle in ATRA-treated APL cells.


Subject(s)
Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinase , Proto-Oncogene Proteins c-akt/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Arsenic Trioxide , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Tretinoin/pharmacology , TOR Serine-Threonine Kinases , Cell Death , Mechanistic Target of Rapamycin Complex 2 , Integrins , Arsenicals/pharmacology , PTEN Phosphohydrolase/genetics
2.
Int J Mol Sci ; 21(11)2020 May 29.
Article in English | MEDLINE | ID: mdl-32485850

ABSTRACT

The multifunctional tissue transglutaminase has been demonstrated to act as α1-adrenergic receptor-coupled G protein with GTPase activity in several cell types. To explore further the pathophysiological significance of this function we investigated the in vivo effects of the α1-adrenergic receptor agonist phenylephrine comparing responses in wild type and TG2-/- mice. Injection of phenylephrine, but not a beta3-adrenergic agonist (CL-316,243), resulted in the long-term decline of the respiratory exchange ratio and lower lactate concentration in TG2-/- mice indicating they preferred to utilize fatty acids instead of glucose as fuels. Measurement of tail blood pressure revealed that the vasoconstrictive effect of phenylephrine was milder in TG2-/- mice leading to lower levels of lactate dehydrogenase (LDH) isoenzymes in blood. LDH isoenzyme patterns indicated more damage in lung, liver, kidney, skeletal, and cardiac muscle of wild type mice; the latter was confirmed by a higher level of heart-specific CK-MB. Our data suggest that TG2 as an α1-adrenergic receptor-coupled G protein has important regulatory functions in alpha1-adrenergic receptor-mediated metabolic processes and vascular functions.


Subject(s)
Blood Vessels/drug effects , GTP-Binding Proteins/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Transglutaminases/metabolism , Vasoconstriction , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , Blood Vessels/metabolism , Blood Vessels/physiology , Dioxoles/pharmacology , Fatty Acids/metabolism , GTP-Binding Proteins/genetics , Glucose/metabolism , Kidney/metabolism , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Liver/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Muscles/metabolism , Phenylephrine/pharmacology , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/genetics
3.
J Immunol ; 203(6): 1532-1547, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31405954

ABSTRACT

The infiltration and subsequent in situ subtype specification of monocytes to effector/inflammatory and repair macrophages is indispensable for tissue repair upon acute sterile injury. However, the chromatin-level mediators and regulatory events controlling this highly dynamic macrophage phenotype switch are not known. In this study, we used a murine acute muscle injury model to assess global chromatin accessibility and gene expression dynamics in infiltrating macrophages during sterile physiological inflammation and tissue regeneration. We identified a heme-binding transcriptional repressor, BACH1, as a novel regulator of this process. Bach1 knockout mice displayed impaired muscle regeneration, altered dynamics of the macrophage phenotype transition, and transcriptional deregulation of key inflammatory and repair-related genes. We also found that BACH1 directly binds to and regulates distal regulatory elements of these genes, suggesting a novel role for BACH1 in controlling a broad spectrum of the repair response genes in macrophages upon injury. Inactivation of heme oxygenase-1 (Hmox1), one of the most stringently deregulated genes in the Bach1 knockout in macrophages, impairs muscle regeneration by changing the dynamics of the macrophage phenotype switch. Collectively, our data suggest the existence of a heme-BACH1--HMOX1 regulatory axis, that controls the phenotype and function of the infiltrating myeloid cells upon tissue damage, shaping the overall tissue repair kinetics.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Muscle, Skeletal/metabolism , Regeneration/physiology , Animals , Inflammation/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Transcription, Genetic/physiology
4.
Article in English | MEDLINE | ID: mdl-31275241

ABSTRACT

With thymic senescence the epithelial network shrinks to be replaced by adipose tissue. Transcription factor TBX-1 controls thymus organogenesis, however, the same TBX-1 has also been reported to orchestrate beige adipose tissue development. Given these different roles of TBX-1, we have assessed if thymic TBX-1 expression persists and demonstrates this dualism during adulthood. We have also checked whether thymic adipose involution could yield beige adipose tissue. We have used adult mouse and human thymus tissue from various ages to evaluate the kinetics of TBX-1 expression, as well as mouse (TEP1) and human (1889c) thymic epithelial cells (TECs) for our studies. Electron micrographs show multi-locular lipid deposits typical of beige adipose cells. Histology staining shows the accumulation of neutral lipid deposits. qPCR measurements show persistent and/or elevating levels of beige-specific and beige-indicative markers (TBX-1, EAR-2, UCP-1, PPAR-gamma). We have performed miRNome profiling using qPCR-based QuantStudio platform and amplification-free NanoString platform. We have observed characteristic alterations, including increased miR21 level (promoting adipose tissue development) and decreased miR34a level (bias toward beige adipose tissue differentiation). Finally, using the Seahorse metabolic platform we have recorded a metabolic profile (OCR/ECAR ratio) indicative of beige adipose tissue. In summary, our results support that thymic adipose tissue emerging with senescence is bona fide beige adipose tissue. Our data show how the borders blur between a key immune tissue (the thymus) and a key metabolic tissue (beige adipose tissue) with senescence. Our work contributes to the understanding of cross talk between the immune system and metabolism.

5.
J Neurosci ; 38(35): 7683-7700, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30054395

ABSTRACT

Aging contributes to cellular stress and neurodegeneration. Our understanding is limited regarding the tissue-restricted mechanisms providing protection in postmitotic cells throughout life. Here, we show that spinal cord motoneurons exhibit a high abundance of asymmetric dimethyl arginines (ADMAs) and the presence of this posttranslational modification provides protection against environmental stress. We identify protein arginine methyltransferase 8 (PRMT8) as a tissue-restricted enzyme responsible for proper ADMA level in postmitotic neurons. Male PRMT8 knock-out mice display decreased muscle strength with aging due to premature destabilization of neuromuscular junctions. Mechanistically, inhibition of methyltransferase activity or loss of PRMT8 results in accumulation of unrepaired DNA double-stranded breaks and decrease in the cAMP response-element-binding protein 1 (CREB1) level. As a consequence, the expression of CREB1-mediated prosurvival and regeneration-associated immediate early genes is dysregulated in aging PRMT8 knock-out mice. The uncovered role of PRMT8 represents a novel mechanism of stress tolerance in long-lived postmitotic neurons and identifies PRMT8 as a tissue-specific therapeutic target in the prevention of motoneuron degeneration.SIGNIFICANCE STATEMENT Although most of the cells in our body have a very short lifespan, postmitotic neurons must survive for many decades. Longevity of a cell within the organism depends on its ability to properly regulate signaling pathways that counteract perturbations, such as DNA damage, oxidative stress, or protein misfolding. Here, we provide evidence that tissue-specific regulators of stress tolerance exist in postmitotic neurons. Specifically, we identify protein arginine methyltransferase 8 (PRMT8) as a cell-type-restricted arginine methyltransferase in spinal cord motoneurons (MNs). PRMT8-dependent arginine methylation is required for neuroprotection against age-related increased of cellular stress. Tissue-restricted expression and the enzymatic activity of PRMT8 make it an attractive target for drug development to delay the onset of neurodegenerative disorders.


Subject(s)
DNA Damage/physiology , Motor Neurons/enzymology , Protein-Arginine N-Methyltransferases/physiology , Aging/metabolism , Amino Acid Sequence , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Cell Line , Cyclic AMP Response Element-Binding Protein/physiology , DNA Breaks, Double-Stranded , DNA Repair , Isometric Contraction , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle Cells/enzymology , Muscle Cells/physiology , Neuromuscular Junction/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/deficiency , Protein-Arginine N-Methyltransferases/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/metabolism , Reflex, Abnormal , Rotarod Performance Test , Spinal Cord/cytology , Spinal Cord/growth & development
6.
J Nutr Biochem ; 57: 238-245, 2018 07.
Article in English | MEDLINE | ID: mdl-29800810

ABSTRACT

Dietary supplementation with conjugated linoleic acid (CLA) has been proposed for weight management and to prevent gut inflammation. However, some animal studies suggest that supplementation with CLA leads to the development of nonalcoholic fatty liver disease. The aims of this study were to test the efficiency of CLA in preventing dextran sulfate sodium (DSS)-induced colitis, to analyze the effects of CLA in the liver function, and to access putative liver alterations upon CLA supplementation during colitis. So, C57BL/6 mice were supplemented for 3 weeks with either control diet (AIN-G) or 1% CLA-supplemented diet. CLA content in the diet and in the liver of mice fed CLA containing diet were accessed by gas chromatography. On the first day of the third week of dietary treatment, mice received ad libitum a 1.5%-2.5% DSS solution for 7 days. Disease activity index score was evaluated; colon and liver samples were stained by hematoxylin and eosin for histopathology analysis and lamina propria cells were extracted to access the profile of innate cell infiltrate. Metabolic alterations before and after colitis induction were accessed by an open calorimetric circuit. Serum glucose, cholesterol, triglycerides and alanine aminotransaminase were measured; the content of fat in liver and feces was also accessed. CLA prevented weight loss, histopathologic and macroscopic signs of colitis, and inflammatory infiltration. Mice fed CLA-supplemented without colitis induction diet developed steatosis, which was prevented in mice with colitis probably due to the higher lipid consumption as energy during gut inflammation. This result suggests that CLA is safe for use during gut inflammation but not at steady-state conditions.


Subject(s)
Colitis/diet therapy , Linoleic Acids, Conjugated/pharmacology , Non-alcoholic Fatty Liver Disease/chemically induced , Alanine Transaminase/blood , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colitis/chemically induced , Colitis/prevention & control , Colon/drug effects , Colon/immunology , Colon/pathology , Dextran Sulfate/toxicity , Dietary Supplements , Female , Immunity, Innate/drug effects , Immunity, Innate/physiology , Linoleic Acid/metabolism , Linoleic Acids, Conjugated/adverse effects , Liver/cytology , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL
7.
Mol Cell Biol ; 38(10)2018 05 15.
Article in English | MEDLINE | ID: mdl-29507185

ABSTRACT

The liver X receptors (LXRs) are ligand-activated nuclear receptors with established roles in the maintenance of lipid homeostasis in multiple tissues. LXRs exert additional biological functions as negative regulators of inflammation, particularly in macrophages. However, the transcriptional responses controlled by LXRs in other myeloid cells, such as dendritic cells (DCs), are still poorly understood. Here we used gain- and loss-of-function models to characterize the impact of LXR deficiency on DC activation programs. Our results identified an LXR-dependent pathway that is important for DC chemotaxis. LXR-deficient mature DCs are defective in stimulus-induced migration in vitro and in vivo Mechanistically, we show that LXRs facilitate DC chemotactic signaling by regulating the expression of CD38, an ectoenzyme important for leukocyte trafficking. Pharmacological or genetic inactivation of CD38 activity abolished the LXR-dependent induction of DC chemotaxis. Using the low-density lipoprotein receptor-deficient (LDLR-/-) LDLR-/- mouse model of atherosclerosis, we also demonstrated that hematopoietic CD38 expression is important for the accumulation of lipid-laden myeloid cells in lesions, suggesting that CD38 is a key factor in leukocyte migration during atherogenesis. Collectively, our results demonstrate that LXRs are required for the efficient emigration of DCs in response to chemotactic signals during inflammation.


Subject(s)
Chemotaxis/physiology , Dendritic Cells/physiology , Liver X Receptors/physiology , ADP-ribosyl Cyclase 1/metabolism , Animals , Cells, Cultured , Dendritic Cells/cytology , Inflammation , Lipid Metabolism , Liver X Receptors/genetics , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear , Signal Transduction
8.
Front Immunol ; 8: 1515, 2017.
Article in English | MEDLINE | ID: mdl-29163553

ABSTRACT

Thymic senescence contributes to increased incidence of infection, cancer and autoimmunity at senior ages. This process manifests as adipose involution. As with other adipose tissues, thymic adipose involution is also controlled by PPARgamma. This is supported by observations reporting that systemic PPARgamma activation accelerates thymic adipose involution. Therefore, we hypothesized that decreased PPARgamma activity could prevent thymic adipose involution, although it may trigger metabolic adverse effects. We have confirmed that both human and murine thymic sections show marked staining for PPARgamma at senior ages. We have also tested the thymic lobes of PPARgamma haplo-insufficient and null mice. Supporting our working hypothesis both adult PPARgamma haplo-insufficient and null mice show delayed thymic senescence by thymus histology, thymocyte mouse T-cell recombination excision circle qPCR and peripheral blood naive T-cell ratio by flow-cytometry. Delayed senescence showed dose-response with respect to PPARgamma deficiency. Functional immune parameters were also evaluated at senior ages in PPARgamma haplo-insufficient mice (null mice do not reach senior ages due to metabolic adverse affects). As expected, sustained and elevated T-cell production conferred oral tolerance and enhanced vaccination efficiency in senior PPARgamma haplo-insufficient, but not in senior wild-type littermates according to ELISA IgG measurements. Of note, humans also show increased oral intolerance issues and decreased protection by vaccines at senior ages. Moreover, PPARgamma haplo-insufficiency also exists in human known as a rare disease (FPLD3) causing metabolic adverse effects, similar to the mouse. When compared to age- and metabolic disorder-matched other patient samples (FPLD2 not affecting PPARgamma activity), FPLD3 patients showed increased human Trec (hTrec) values by qPCR (within healthy human range) suggesting delayed thymic senescence, in accordance with mouse results and supporting our working hypothesis. In summary, our experiments prove that systemic decrease of PPARgamma activity prevents thymic senescence, albeit with metabolic drawbacks. However, thymic tissue-specific PPARgamma antagonism would likely solve the issue.

9.
Proc Natl Acad Sci U S A ; 114(40): 10725-10730, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28923935

ABSTRACT

Retinoid X receptor (RXR) regulates several key functions in myeloid cells, including inflammatory responses, phagocytosis, chemokine secretion, and proangiogenic activity. Its importance, however, in tumor-associated myeloid cells is unknown. In this study, we demonstrate that deletion of RXR in myeloid cells enhances lung metastasis formation while not affecting primary tumor growth. We show that RXR deficiency leads to transcriptomic changes in the lung myeloid compartment characterized by increased expression of prometastatic genes, including important determinants of premetastatic niche formation. Accordingly, RXR-deficient myeloid cells are more efficient in promoting cancer cell migration and invasion. Our results suggest that the repressive activity of RXR on prometastatic genes is mediated primarily through direct DNA binding of the receptor along with nuclear receptor corepressor (NCoR) and silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) corepressors and is largely unresponsive to ligand activation. In addition, we found that expression and transcriptional activity of RXRα is down-modulated in peripheral blood mononuclear cells of patients with lung cancer, particularly in advanced and metastatic disease. Overall, our results identify RXR as a regulator in the myeloid cell-assisted metastatic process and establish lipid-sensing nuclear receptors in the microenvironmental regulation of tumor progression.


Subject(s)
Carcinoma, Lewis Lung/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Myeloid Cells/pathology , Retinoid X Receptors/physiology , Transcription, Genetic , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Cells, Cultured , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism
10.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(12): 1575-1586, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28774822

ABSTRACT

During cold-exposure 'beige' adipocytes with increased mitochondrial content are activated in white adipose tissue (WAT). These cells, similarly to brown adipose tissue (BAT), dissipate stored chemical energy in the form of heat with the help of uncoupling protein 1 (UCP1). We investigated the effect of tissue transglutaminase (TG2) ablation on the function of ATs in mice. Although TG2+/+ and TG2-/- mice had the same amount of WAT and BAT, we found that TG2+/+ animals could tolerate acute cold exposure for 4h, whereas TG2-/- mice only for 3h. Both TG2-/- and TG2+/+ animals used up half of the triacylglycerol content of subcutaneous WAT (SCAT) after 3h treatment; however, TG2-/- mice still possessed markedly whiter and higher amount of gonadal WAT (GONAT) as reflected in the larger size of adipocytes and lower free fatty acid levels in serum. Furthermore, lower expression of 'beige' marker genes such as UCP1, TBX1 and TNFRFS9 was observed after cold exposure in GONAT of TG2-/- mice, paralleled with a lower level of UCP1 protein and a decreased mitochondrial content. The detected changes in gene expression of Resistin and Adiponectin did not provoke glucose intolerance in the investigated TG2-/- mice, and TG2 deletion did not influence adrenaline, noradrenaline, glucagon and insulin production. Our data suggest that TG2 has a tissue-specific role in GONAT function and browning, which becomes apparent under acute cold exposure.


Subject(s)
Acclimatization , Adipose Tissue, White/metabolism , Cold Temperature , Fatty Acids/metabolism , GTP-Binding Proteins/deficiency , Testis/metabolism , Transglutaminases/deficiency , Adiponectin/biosynthesis , Adiponectin/genetics , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Animals , Fatty Acids/genetics , Male , Mice , Mice, Knockout , Protein Glutamine gamma Glutamyltransferase 2 , Resistin/biosynthesis , Resistin/genetics , Testis/cytology
11.
J Physiol ; 595(17): 5815-5842, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28714082

ABSTRACT

KEY POINTS: The in situ phenotypic switch of macrophages is delayed in acute injury following irradiation. The combination of bone marrow transplantation and local muscle radiation protection allows for the identification of a myeloid cell contribution to tissue repair. PET-MRI allows monitoring of myeloid cell invasion and metabolism. Altered cellular composition prior to acute sterile injury affects the in situ phenotypic transition of invading myeloid cells to repair macrophages. There is reciprocal intercellular communication between local muscle cell compartments, such as PAX7 positive cells, and recruited macrophages during skeletal muscle regeneration. ABSTRACT: Skeletal muscle regeneration is a complex interplay between various cell types including invading macrophages. Their recruitment to damaged tissues upon acute sterile injuries is necessary for clearance of necrotic debris and for coordination of tissue regeneration. This highly dynamic process is characterized by an in situ transition of infiltrating monocytes from an inflammatory (Ly6Chigh ) to a repair (Ly6Clow ) macrophage phenotype. The importance of the macrophage phenotypic shift and the cross-talk of the local muscle tissue with the infiltrating macrophages during tissue regeneration upon injury are not fully understood and their study lacks adequate methodology. Here, using an acute sterile skeletal muscle injury model combined with irradiation, bone marrow transplantation and in vivo imaging, we show that preserved muscle integrity and cell composition prior to the injury is necessary for the repair macrophage phenotypic transition and subsequently for proper and complete tissue regeneration. Importantly, by using a model of in vivo ablation of PAX7 positive cells, we show that this radiosensitive skeletal muscle progenitor pool contributes to macrophage phenotypic transition following acute sterile muscle injury. In addition, local muscle tissue radioprotection by lead shielding during irradiation preserves normal macrophage transition dynamics and subsequently muscle tissue regeneration. Taken together, our data suggest the existence of a more extensive and reciprocal cross-talk between muscle tissue compartments, including satellite cells, and infiltrating myeloid cells upon tissue damage. These interactions shape the macrophage in situ phenotypic shift, which is indispensable for normal muscle tissue repair dynamics.


Subject(s)
Macrophages/immunology , Muscle, Skeletal , Radiation Injuries, Experimental/immunology , Animals , Bone Marrow Transplantation , Cardiotoxins , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/immunology , Muscle, Skeletal/injuries , Muscle, Skeletal/radiation effects , Phenotype , Positron Emission Tomography Computed Tomography , Radiation Injuries, Experimental/diagnostic imaging , Regeneration
12.
J Immunol ; 198(1): 239-248, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27852743

ABSTRACT

Application of dendritic cells (DCs) to prime responses to tumor Ags provides a promising approach to immunotherapy. However, only a limited number of DCs can be manufactured from adult precursors. In contrast, pluripotent embryonic stem (ES) cells represent an inexhaustible source for DC production, although it remains a major challenge to steer directional differentiation because ES cell-derived cells are typically immature with impaired functional capacity. Consistent with this notion, we found that mouse ES cell-derived DCs (ES-DCs) represented less mature cells compared with bone marrow-derived DCs. This finding prompted us to compare the gene expression profile of the ES cell- and adult progenitor-derived, GM-CSF-instructed, nonconventional DC subsets. We quantified the mRNA level of 17 DC-specific transcription factors and observed that 3 transcriptional regulators (Irf4, Spi-B, and Runx3) showed lower expression in ES-DCs than in bone marrow-derived DCs. In light of this altered gene expression, we probed the effects of these transcription factors in developing mouse ES-DCs with an isogenic expression screen. Our analysis revealed that forced expression of Irf4 repressed ES-DC development, whereas, in contrast, Runx3 improved the ES-DC maturation capacity. Moreover, LPS-treated and Runx3-activated ES-DCs exhibited enhanced T cell activation and migratory potential. In summary, we found that ex vivo-generated ES-DCs had a compromised maturation ability and immunogenicity. However, ectopic expression of Runx3 enhances cytokine-driven ES-DC development and acts as an instructive tool for the generation of mature DCs with enhanced immunogenicity from pluripotent stem cells.


Subject(s)
Cell Differentiation/physiology , Core Binding Factor Alpha 3 Subunit/biosynthesis , Dendritic Cells/cytology , Ectopic Gene Expression/physiology , Embryonic Stem Cells/cytology , Animals , Blotting, Western , Cell Separation , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/immunology , Pluripotent Stem Cells/metabolism , Real-Time Polymerase Chain Reaction , Transcriptome
13.
Immunity ; 45(5): 1038-1051, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27836432

ABSTRACT

Tissue regeneration requires inflammatory and reparatory activity of macrophages. Macrophages detect and eliminate the damaged tissue and subsequently promote regeneration. This dichotomy requires the switch of effector functions of macrophages coordinated with other cell types inside the injured tissue. The gene regulatory events supporting the sensory and effector functions of macrophages involved in tissue repair are not well understood. Here we show that the lipid activated transcription factor, PPARγ, is required for proper skeletal muscle regeneration, acting in repair macrophages. PPARγ controls the expression of the transforming growth factor-ß (TGF-ß) family member, GDF3, which in turn regulates the restoration of skeletal muscle integrity by promoting muscle progenitor cell fusion. This work establishes PPARγ as a required metabolic sensor and transcriptional regulator of repair macrophages. Moreover, this work also establishes GDF3 as a secreted extrinsic effector protein acting on myoblasts and serving as an exclusively macrophage-derived regeneration factor in tissue repair.


Subject(s)
Growth Differentiation Factor 3/metabolism , Muscle, Skeletal/physiology , Myoblasts/metabolism , PPAR gamma/metabolism , Regeneration/physiology , Animals , Blotting, Western , Cell Separation , Chromatin Immunoprecipitation , Disease Models, Animal , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/injuries , Oligonucleotide Array Sequence Analysis , Wound Healing/physiology
14.
Int J Mol Sci ; 17(8)2016 Jul 30.
Article in English | MEDLINE | ID: mdl-27483259

ABSTRACT

With the increasing number of patients affected with metabolic diseases such as type 2 diabetes, obesity, atherosclerosis and insulin resistance, academic researchers and pharmaceutical companies are eager to better understand metabolic syndrome and develop new drugs for its treatment. Many studies have focused on the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ), which plays a crucial role in adipogenesis and lipid metabolism. These studies have been able to connect this transcription factor to several human metabolic diseases. Due to obvious limitations concerning experimentation in humans, animal models-mainly mouse models-have been generated to investigate the role of PPARγ in different tissues. This review focuses on the metabolic features of human and mouse PPARγ-related diseases and the utility of the mouse as a model.


Subject(s)
Disease Models, Animal , Metabolic Diseases/physiopathology , PPAR gamma/metabolism , Animals , Humans , Mice
16.
Cardiovasc Diabetol ; 13: 150, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25361524

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor-γ (PPARγ) agonists, which have been used as insulin sensitizers in diabetic patients, may improve functions of endothelial cells (ECs). We investigated the effect of PPARγ on angiogenic activities of murine ECs and bone marrow-derived proangiogenic cells (PACs). METHODS: PACs were isolated from bone marrow of 10-12 weeks old, wild type, db/db and PPARγ heterozygous animals. Cells were cultured on fibronectin and gelatin coated dishes in EGM-2MV medium. For in vitro stimulations, rosiglitazone (10 µmol/L) or GW9662 (10 µmol/L) were added to 80% confluent cell cultures for 24 hours. Angiogenic potential of PACs and ECs was tested in vitro and in vivo in wound healing assay and hind limb ischemia model. RESULTS: ECs and PACs isolated from diabetic db/db mice displayed a reduced angiogenic potential in ex vivo and in vitro assays, the effect partially rescued by incubation of cells with rosiglitazone (PPARγ activator). Correction of diabetes by administration of rosiglitazone in vivo did not improve angiogenic potential of isolated PACs or ECs. In a hind limb ischemia model we demonstrated that local injection of conditioned media harvested from wild type PACs improved the blood flow restoration in db/db mice, confirming the importance of paracrine action of the bone marrow-derived cells. CONCLUSIONS: In summary, activation of PPARγ by rosiglitazone improves angiogenic potential of diabetic ECs and PACs, but decreased expression of PPARγ in diabetes does not impair angiogenesis.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow/drug effects , Endothelial Cells/drug effects , PPAR gamma/metabolism , Stem Cells/metabolism , Animals , Bone Marrow Cells/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Hypoglycemic Agents/pharmacology , Ischemia/drug therapy , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , PPAR gamma/genetics , Rosiglitazone , Stem Cells/cytology , Stem Cells/drug effects , Thiazolidinediones/pharmacology , Wound Healing/drug effects
17.
J Lipid Res ; 54(9): 2458-74, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23833249

ABSTRACT

All-trans retinoic acid (ATRA) has a key role in dendritic cells (DCs) and affects T cell subtype specification and gut homing. However, the identity of the permissive cell types and the required steps of conversion of vitamin A to biologically active ATRA bringing about retinoic acid receptor-regulated signaling remains elusive. Here we present that only a subset of murine and human DCs express the necessary enzymes, including RDH10, RALDH2, and transporter cellular retinoic acid binding protein (CRABP)2, to produce ATRA and efficient signaling. These permissive cell types include CD103(+) DCs, granulocyte-macrophage colony-stimulating factor, and interleukin-4-treated bone marrow-derived murine DCs and human monocyte-derived DCs (mo-DCs). Importantly, in addition to RDH10 and RALDH2, CRABP2 also appears to be regulated by the fatty acid-sensing nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) and colocalize in human gut-associated lymphoid tissue DCs. In our model of human mo-DCs, all three proteins (RDH10, RALDH2, and CRABP2) appeared to be required for ATRA production induced by activation of PPARγ and therefore form a linear pathway. This now functionally validated PPARγ-regulated ATRA producing and signaling axis equips the cells with the capacity to convert precursors to active retinoids in response to receptor-activating fatty acids and is potentially amenable to intervention in diseases involving or affecting mucosal immunity.


Subject(s)
Alcohol Oxidoreductases/metabolism , Dendritic Cells/metabolism , PPAR gamma/metabolism , Receptors, Retinoic Acid/metabolism , Retinal Dehydrogenase/metabolism , Signal Transduction , Tretinoin/metabolism , Alcohol Oxidoreductases/deficiency , Alcohol Oxidoreductases/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Dendritic Cells/cytology , Dendritic Cells/enzymology , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Intestines/cytology , Male , Mice , Monocytes/cytology , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Protein Transport , Receptors, Retinoic Acid/deficiency , Receptors, Retinoic Acid/genetics , Retinal Dehydrogenase/deficiency , Retinal Dehydrogenase/genetics , Transglutaminases/metabolism
18.
J Leukoc Biol ; 91(1): 127-36, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22028334

ABSTRACT

GCs are powerful anti-inflammatory compounds inhibiting inflammatory cell recruitment and production of proinflammatory cytokines. We have recently found that DCs, the key players of T cell priming and polarization, respond to allogeneic apoptotic neutrophils with proinflammatory cytokine release and Th1 cell activation. Here, we show that monocyte-derived human DCs develop their capacity to engulf apoptotic cells by up-regulating a set of apoptophagocytic genes. This gene expression pattern was reprogrammed when differentiation took place in the presence of the synthetic GC Dex, which increased the expression of phagocytosis receptors MERTK and CD14, the bridging molecule C1QA, DNASE2, and ADORA3. The increased phagocytosis was attenuated by the addition of ADORA3 antagonist and could not be observed when bone marrow-derived DCs of ADORA3 KO mice were treated with Dex. The GC-treated human DCs loaded with allogeneic apoptotic neutrophils secreted, in response to LPS and IFN-γ, the inflammatory cytokine TNF-α. Furthermore, the Dex-treated DCs could activate autologous T lymphocytes toward Th1 effector cells, and this was enhanced by their exposure to allogeneic apoptotic neutrophils.


Subject(s)
Dendritic Cells/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Neutrophils/immunology , Phagocytosis/drug effects , Animals , Apoptosis/immunology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Neutrophils/cytology , Phagocytosis/immunology
19.
Immunity ; 33(5): 699-712, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21093321

ABSTRACT

Peroxisome proliferator-activated receptor γ (PPARγ) is a lipid-activated transcription factor regulating lipid metabolism and inflammatory response in macrophages and dendritic cells (DCs). These immune cells exposed to distinct inflammatory milieu show cell type specification as a result of altered gene expression. We demonstrate here a mechanism how inflammatory molecules modulate PPARγ signaling in distinct subsets of cells. Proinflammatory molecules inhibited whereas interleukin-4 (IL-4) stimulated PPARγ activity in macrophages and DCs. Furthermore, IL-4 signaling augmented PPARγ activity through an interaction between PPARγ and signal transducer and activators of transcription 6 (STAT6) on promoters of PPARγ target genes, including FABP4. Thus, STAT6 acts as a facilitating factor for PPARγ by promoting DNA binding and consequently increasing the number of regulated genes and the magnitude of responses. This interaction, underpinning cell type-specific responses, represents a unique way of controlling nuclear receptor signaling by inflammatory molecules in immune cells.


Subject(s)
Dendritic Cells/metabolism , Gene Expression Regulation , Macrophages/metabolism , PPAR gamma/metabolism , STAT6 Transcription Factor/metabolism , Animals , Fatty Acid-Binding Proteins/metabolism , Humans , Inflammation Mediators/metabolism , Interleukin-4/metabolism , Mice , Promoter Regions, Genetic
20.
Mol Endocrinol ; 24(11): 2218-31, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20861222

ABSTRACT

Retinoid X receptors (RXRs) are heterodimerization partners for many nuclear receptors and also act as homodimers. Heterodimers formed by RXR and a nonpermissive partner, e.g. retinoic acid receptor (RAR) and vitamin D receptor (VDR), can be activated only by the agonist of the partner receptor. In contrast, heterodimers that contain permissive partners, e.g. liver X receptor (LXR) and peroxisome proliferator-activated receptor (PPAR), can be activated by agonists for either the partner receptor or RXR, raising the possibility of pleiotropic RXR signaling. However, it is not known to what extent the receptor's activation results in triggering mechanisms dependent or independent of permissive heterodimers. In this study, we systematically and quantitatively characterized all probable RXR-signaling pathways in differentiating human monocyte-derived dendritic cells (Mo-DCs). Using pharmacological, microarray and quantitative RT-PCR techniques, we identified and characterized gene sets regulated by RXR agonists (LG100268 and 9-cis retinoic acid) and agonists for LXRs, PPARs, RARα, and VDR. Our results demonstrated that permissiveness was partially impaired in Mo-DCs, because a large number of genes regulated by PPAR or LXR agonists was not affected by RXR-specific agonists or was regulated to a lesser extent. As expected, we found that RXR agonists regulated only small portions of RARα or VDR targets. Importantly, we could identify and characterize PPAR- and LXR-independent pathways in Mo-DCs most likely mediated by RXR homodimers. These data suggested that RXR signaling in Mo-DCs was mediated via multiple permissive heterodimers and also by mechanism(s) independent of permissive heterodimers, and it was controlled in a cell-type and gene-specific manner.


Subject(s)
Cell Differentiation/genetics , Dendritic Cells/cytology , Gene Expression Profiling , Gene Expression Regulation , Monocytes/cytology , Receptors, Retinoic Acid/metabolism , Benzoates/pharmacology , Cell Differentiation/drug effects , Cell Line , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Humans , Ligands , Liver X Receptors , Monocytes/drug effects , Monocytes/metabolism , Nicotinic Acids/pharmacology , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/metabolism , PPAR delta/agonists , PPAR delta/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , Phenotype , Receptors, Calcitriol/agonists , Receptors, Calcitriol/metabolism , Receptors, Retinoic Acid/agonists , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Tetrahydronaphthalenes/pharmacology , Tretinoin/pharmacology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...