Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
J Physiol ; 601(7): 1159-1171, 2023 04.
Article in English | MEDLINE | ID: mdl-36825510

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD), recently also defined as metabolic dysfunction-associated fatty liver disease (MAFLD), is a major health problem, as it affects ∼25% of the population globally and is a major cause of hepatic cirrhosis and thereby liver failure, as well as hepatocellular carcinoma. MALFD comprises a broad range of pathological conditions in the liver, including simple fat accumulation (steatosis) and the more progressive non-alcoholic steatohepatitis (NASH) that can lead to fibrosis development. Cells of innate immunity, and particularly macrophages, comprising the liver resident Kupffer cells and the recruited monocyte-derived macrophages, play complex roles in NASH-related inflammation and disease progression to fibrosis. Here, we discuss the recent developments with regards to the function of liver macrophage subpopulations during MAFLD development and progression.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/metabolism , Liver/metabolism , Macrophages , Kupffer Cells/metabolism , Kupffer Cells/pathology , Fibrosis
3.
Cell Mol Gastroenterol Hepatol ; 15(6): 1277-1292, 2023.
Article in English | MEDLINE | ID: mdl-36828280

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a fast growing, chronic liver disease affecting ∼25% of the global population. Nonalcoholic fatty liver disease severity ranges from the less severe simple hepatic steatosis to the more advanced nonalcoholic steatohepatitis (NASH). The presence of NASH predisposes individuals to liver fibrosis, which can further progress to cirrhosis and hepatocellular carcinoma. This makes hepatic fibrosis an important indicator of clinical outcomes in patients with NASH. Hepatic stellate cell activation dictates fibrosis development during NASH. Here, we discuss recent advances in the analysis of the profibrogenic pathways and mediators of hepatic stellate cell activation and inactivation, which ultimately determine the course of disease in nonalcoholic fatty liver disease/NASH.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/pathology , Hepatic Stellate Cells/pathology , Liver Cirrhosis/pathology , Carcinoma, Hepatocellular/pathology , Fibrosis , Liver Neoplasms/pathology
4.
Int J Mol Sci ; 24(4)2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36834840

ABSTRACT

Though electrical stimulation is used as a therapeutic approach to treat retinal and spinal injuries, many protective mechanisms at cellular level have not been elucidated. We performed a detailed analysis of cellular events in blue light (Li) stressed 661W cells, which were subjected to direct current electric field (EF) stimulation. Our findings revealed that EF stimulation induced protective effects in 661W cells from Li-induced stress by multiple defense mechanisms, such as increase in mitochondrial activity, gain in mitochondrial potential, increase in superoxide levels, and the activation of unfolded protein response (UPR) pathways, all leading to an enhanced cell viability and decreased DNA damage. Here, our genetic screen results revealed the UPR pathway to be a promising target to ameliorate Li-induced stress by EF stimulation. Thus, our study is important for a knowledgeable transfer of EF stimulation into clinical application.


Subject(s)
Retina , Unfolded Protein Response , Cell Line , Mitochondria , Electric Stimulation , Light
5.
Hepatology ; 77(3): E60-E61, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36104065
6.
Int J Mol Sci ; 23(13)2022 Jun 23.
Article in English | MEDLINE | ID: mdl-35805998

ABSTRACT

The prevalence of nonalcoholic fatty liver disease (NAFLD), recently also re-defined as metabolic dysfunction associated fatty liver disease (MAFLD), is rapidly increasing, affecting ~25% of the world population. MALFD/NAFLD represents a spectrum of liver pathologies including the more benign hepatic steatosis and the more advanced non-alcoholic steatohepatitis (NASH). NASH is associated with enhanced risk for liver fibrosis and progression to cirrhosis and hepatocellular carcinoma. Hepatic stellate cells (HSC) activation underlies NASH-related fibrosis. Here, we discuss the profibrogenic pathways, which lead to HSC activation and fibrogenesis, with a particular focus on the intercellular hepatocyte-HSC and macrophage-HSC crosstalk.


Subject(s)
Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Fibrosis , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/metabolism
7.
J Clin Invest ; 132(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35133978

ABSTRACT

The causative role of inflammation in hypertension-related cardiovascular diseases is evident and calls for development of specific immunomodulatory therapies. We tested the therapeutic efficacy and mechanisms of action of developmental endothelial locus-1 (DEL-1), an endogenous antiinflammatory factor, in angiotensin II- (ANGII-) and deoxycorticosterone acetate-salt-induced (DOCA-salt-induced) cardiovascular organ damage and hypertension. By using mice with endothelial overexpression of DEL-1 (EC-Del1 mice) and performing preventive and interventional studies by injecting recombinant DEL-1 in mice, we showed that DEL-1 improved endothelial function and abrogated aortic adventitial fibrosis, medial thickening, and loss of elastin. DEL-1 also protected the mice from cardiac concentric hypertrophy and interstitial and perivascular coronary fibrosis and improved left ventricular function and myocardial coronary perfusion. DEL-1 prevented aortic stiffness and abolished the progression of hypertension. Mechanistically, DEL-1 acted by inhibiting αvß3 integrin-dependent activation of pro-MMP2 in mice and in human isolated aorta. Moreover, DEL-1 stabilized αvß3 integrin-dependent CD25+FoxP3+ Treg numbers and IL-10 levels, which were associated with decreased recruitment of inflammatory cells and reduced production of proinflammatory cytokines in cardiovascular organs. The demonstrated effects and immune-modulating mechanisms of DEL-1 in abrogation of cardiovascular remodeling and progression of hypertension identify DEL-1 as a potential therapeutic factor.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules/metabolism , Hypertension , Ventricular Remodeling , Animals , Cardiomegaly , Fibrosis , Hypertension/complications , Immunomodulation/genetics , Integrins , Mice , Ventricular Remodeling/genetics
8.
Hepatology ; 75(4): 881-897, 2022 04.
Article in English | MEDLINE | ID: mdl-34519101

ABSTRACT

BACKGROUND AND AIMS: NAFLD is initiated by steatosis and can progress through fibrosis and cirrhosis to HCC. The RNA binding protein human antigen R (HuR) controls RNAs at the posttranscriptional level; hepatocyte HuR has been implicated in the regulation of diet-induced hepatic steatosis. The present study aimed to understand the role of hepatocyte HuR in NAFLD development and progression to fibrosis and HCC. APPROACH AND RESULTS: Hepatocyte-specific, HuR-deficient mice and control HuR-sufficient mice were fed either a normal diet or an NAFLD-inducing diet. Hepatic lipid accumulation, inflammation, fibrosis, and HCC development were studied by histology, flow cytometry, quantitative PCR, and RNA sequencing. The liver lipidome was characterized by lipidomics analysis, and the HuR-RNA interactions in the liver were mapped by RNA immunoprecipitation sequencing. Hepatocyte-specific, HuR-deficient mice displayed spontaneous hepatic steatosis and fibrosis predisposition compared to control HuR-sufficient mice. On an NAFLD-inducing diet, hepatocyte-specific HuR deficiency resulted in exacerbated inflammation, fibrosis, and HCC-like tumor development. A multi-omic approach, including lipidomics, transcriptomics, and RNA immunoprecipitation sequencing revealed that HuR orchestrates a protective network of hepatic-metabolic and lipid homeostasis-maintaining pathways. Consistently, HuR-deficient livers accumulated, already at steady state, a triglyceride signature resembling that of NAFLD livers. Moreover, up-regulation of secreted phosphoprotein 1 expression mediated, at least partially, fibrosis development in hepatocyte-specific HuR deficiency on an NAFLD-inducing diet, as shown by experiments using antibody blockade of osteopontin. CONCLUSIONS: HuR is a gatekeeper of liver homeostasis, preventing NAFLD-related fibrosis and HCC, suggesting that the HuR-dependent network could be exploited therapeutically.


Subject(s)
Carcinoma, Hepatocellular , ELAV-Like Protein 1 , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Carcinoma, Hepatocellular/pathology , ELAV-Like Protein 1/metabolism , Homeostasis , Inflammation/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology , RNA , Triglycerides/metabolism
9.
Gut ; 70(5): 940-950, 2021 05.
Article in English | MEDLINE | ID: mdl-32591434

ABSTRACT

OBJECTIVE: The rs641738C>T variant located near the membrane-bound O-acyltransferase domain containing 7 (MBOAT7) locus is associated with fibrosis in liver diseases, including non-alcoholic fatty liver disease (NAFLD), alcohol-related liver disease, hepatitis B and C. We aim to understand the mechanism by which the rs641738C>T variant contributes to pathogenesis of NAFLD. DESIGN: Mice with hepatocyte-specific deletion of MBOAT7 (Mboat7Δhep) were generated and livers were characterised by histology, flow cytometry, qPCR, RNA sequencing and lipidomics. We analysed the association of rs641738C>T genotype with liver inflammation and fibrosis in 846 NAFLD patients and obtained genotype-specific liver lipidomes from 280 human biopsies. RESULTS: Allelic imbalance analysis of heterozygous human liver samples pointed to lower expression of the MBOAT7 transcript on the rs641738C>T haplotype. Mboat7Δhep mice showed spontaneous steatosis characterised by increased hepatic cholesterol ester content after 10 weeks. After 6 weeks on a high fat, methionine-low, choline-deficient diet, mice developed increased hepatic fibrosis as measured by picrosirius staining (p<0.05), hydroxyproline content (p<0.05) and transcriptomics, while the inflammatory cell populations and inflammatory mediators were minimally affected. In a human biopsied NAFLD cohort, MBOAT7 rs641738C>T was associated with fibrosis (p=0.004) independent of the presence of histological inflammation. Liver lipidomes of Mboat7Δhep mice and human rs641738TT carriers with fibrosis showed increased total lysophosphatidylinositol levels. The altered lysophosphatidylinositol and phosphatidylinositol subspecies in MBOAT7Δhep livers and human rs641738TT carriers were similar. CONCLUSION: Mboat7 deficiency in mice and human points to an inflammation-independent pathway of liver fibrosis that may be mediated by lipid signalling and a potentially targetable treatment option in NAFLD.


Subject(s)
Acyltransferases/genetics , Liver Cirrhosis/genetics , Membrane Proteins/genetics , Non-alcoholic Fatty Liver Disease/genetics , Acyltransferases/deficiency , Adult , Aged , Animals , Biopsy , Disease Models, Animal , Disease Progression , Female , Genotype , Haplotypes , Humans , Inflammation/genetics , Male , Membrane Proteins/deficiency , Mice, Inbred C57BL , Middle Aged , Polymorphism, Single Nucleotide
10.
Cell ; 183(3): 771-785.e12, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33125892

ABSTRACT

Trained innate immunity, induced via modulation of mature myeloid cells or their bone marrow progenitors, mediates sustained increased responsiveness to secondary challenges. Here, we investigated whether anti-tumor immunity can be enhanced through induction of trained immunity. Pre-treatment of mice with ß-glucan, a fungal-derived prototypical agonist of trained immunity, resulted in diminished tumor growth. The anti-tumor effect of ß-glucan-induced trained immunity was associated with transcriptomic and epigenetic rewiring of granulopoiesis and neutrophil reprogramming toward an anti-tumor phenotype; this process required type I interferon signaling irrespective of adaptive immunity in the host. Adoptive transfer of neutrophils from ß-glucan-trained mice to naive recipients suppressed tumor growth in the latter in a ROS-dependent manner. Moreover, the anti-tumor effect of ß-glucan-induced trained granulopoiesis was transmissible by bone marrow transplantation to recipient naive mice. Our findings identify a novel and therapeutically relevant anti-tumor facet of trained immunity involving appropriate rewiring of granulopoiesis.


Subject(s)
Granulocytes/immunology , Immunity, Innate , Neoplasms/immunology , Adaptive Immunity , Adoptive Transfer , Animals , Epigenesis, Genetic , Interferon Type I/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Neoplasms/pathology , Neutrophils/metabolism , Phenotype , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/metabolism , Transcription, Genetic , Transcriptome/genetics , beta-Glucans/metabolism
11.
Arterioscler Thromb Vasc Biol ; 40(3): 583-596, 2020 03.
Article in English | MEDLINE | ID: mdl-31996026

ABSTRACT

OBJECTIVE: Inflammatory activation changes the mitochondrial function of macrophages from oxidative phosphorylation to reactive oxygen species production, which may promote necrotic core formation in atherosclerotic lesions. In hypoxic and cancer cells, HIF-1α (hypoxia-inducible factor) promotes oxygen-independent energy production by microRNAs. Therefore, we studied the role of HIF-1α in the regulation of macrophage energy metabolism in the context of atherosclerosis. Approach and Results: Myeloid cell-specific deletion of Hif1a reduced atherosclerosis and necrotic core formation by limiting macrophage necroptosis in apolipoprotein E-deficient mice. In inflammatory bone marrow-derived macrophages, deletion of Hif1a increased oxidative phosphorylation, ATP levels, and the expression of genes encoding mitochondrial proteins and reduced reactive oxygen species production and necroptosis. microRNA expression profiling showed that HIF-1α upregulates miR-210 and downregulates miR-383 levels in lesional macrophages and inflammatory bone marrow-derived macrophages. In contrast to miR-210, which inhibited oxidative phosphorylation and enhanced mitochondrial reactive oxygen species production, miR-383 increased ATP levels and inhibited necroptosis. The effect of miR-210 was due to targeting 2,4-dienoyl-CoA reductase, which is essential in the ß oxidation of unsaturated fatty acids. miR-383 affected the DNA damage repair pathway in bone marrow-derived macrophages by targeting poly(ADP-ribose)-glycohydrolase (Parg), which reduced energy consumption and increased cell survival. Blocking the targeting of Parg by miR-383 prevented the protective effect of Hif1a deletion in macrophages on atherosclerosis and necrotic core formation in mice. CONCLUSIONS: Our findings unveil a new mechanism by which activation of HIF-1α in inflammatory macrophages increases necroptosis through microRNA-mediated ATP depletion, thus increasing atherosclerosis by necrotic core formation.


Subject(s)
Aorta/metabolism , Atherosclerosis/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Necroptosis , Adenosine Triphosphate/metabolism , Animals , Aorta/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Energy Metabolism , Gene Expression Regulation , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation/genetics , Inflammation/pathology , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , MicroRNAs/genetics , Mitochondria/metabolism , Mitochondria/pathology , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
13.
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
14.
Cell Mol Life Sci ; 74(2): 359-372, 2017 01.
Article in English | MEDLINE | ID: mdl-27622243

ABSTRACT

MicroRNAs (miRNAs) coordinate vascular repair by regulating injury-induced gene expression in vascular smooth muscle cells (SMCs) and promote the transition of SMCs from a contractile to a proliferating phenotype. However, the effect of miRNA expression in SMCs on neointima formation is unclear. Therefore, we studied the role of miRNA biogenesis by Dicer in SMCs in vascular repair. Following wire-induced injury to carotid arteries of Apolipoprotein E knockout (Apoe -/-) mice, miRNA microarray analysis revealed that the most significantly regulated miRNAs, such as miR-222 and miR-21-3p, were upregulated. Conditional deletion of Dicer in SMCs increased neointima formation by reducing SMC proliferation in Apoe -/- mice, and decreased mainly the expression of miRNAs, such as miR-147 and miR-100, which were not upregulated following vascular injury. SMC-specific deletion of Dicer promoted growth factor and inflammatory signaling and regulated a miRNA-target interaction network in injured arteries that was enriched in anti-proliferative miRNAs. The most connected miRNA in this network was miR-27a-3p [e.g., with Rho guanine nucleotide exchange factor 26 (ARHGEF26)], which was expressed in medial and neointimal SMCs in a Dicer-dependent manner. In vitro, miR-27a-3p suppresses ARHGEF26 expression and inhibits SMC proliferation by interacting with a conserved binding site in the 3' untranslated region of ARHGEF26 mRNA. We propose that Dicer expression in SMCs plays an essential role in vascular repair by generating anti-proliferative miRNAs, such as miR-27a-3p, to prevent vessel stenosis due to exaggerated neointima formation.


Subject(s)
Gene Regulatory Networks , MicroRNAs/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Neointima/genetics , Ribonuclease III/metabolism , Wound Healing/genetics , Animals , Arteries/metabolism , Arteries/pathology , Cell Proliferation , Female , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice , MicroRNAs/metabolism , Myocytes, Smooth Muscle/pathology , Neointima/metabolism , Organ Specificity/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism
15.
Curr Opin Hematol ; 23(1): 36-43, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26554893

ABSTRACT

PURPOSE OF REVIEW: Neutrophils have traditionally been viewed in the context of acute infection and inflammation forming the first line of defense against invading pathogens. Neutrophil trafficking to the site of inflammation requires adhesion and transmigration through blood vessels, which is orchestrated by adhesion molecules, such as ß2 and ß1-integrins, chemokines, and cytokines. The review focuses on recent advances in understanding the regulators of neutrophil recruitment during inflammation in both acute and chronic settings. RECENT FINDINGS: Recent findings suggest that besides the established pathways of selectin or chemokine-mediated integrin activation, signaling by distinct Toll-like receptors (TLRs) (especially TLR2, TLR4, and TLR5) can activate integrin-dependent neutrophil adhesion. Moreover, the integrin α3ß1 has been vitally implicated as a new player in neutrophil recruitment and TLR-mediated responses in septic inflammation. Furthermore, several endogenous inhibitory mechanisms of leukocyte recruitment have been identified, including the secreted molecules Del-1, PTX3, and GDF-15, which block distinct steps of the leukocyte adhesion cascade, as well as novel regulatory signaling pathways, involving the protein kinase AKT1 and IFN-λ2/IL-28A. SUMMARY: The leukocyte adhesion cascade is a tightly regulated process, subjected to both positive and negative regulators. Dysregulation of this process and hence neutrophil recruitment can lead to the development of inflammatory and autoimmune diseases.


Subject(s)
Neutrophil Infiltration , Neutrophils/physiology , Animals , Cell Adhesion/genetics , Cell Adhesion/immunology , Chemokines/genetics , Chemokines/metabolism , Homeostasis , Humans , Integrin alpha3beta1/genetics , Integrin alpha3beta1/metabolism , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Protein Binding , Selectins/genetics , Selectins/metabolism , Signal Transduction , Transendothelial and Transepithelial Migration/genetics , Transendothelial and Transepithelial Migration/immunology
16.
Hypertension ; 66(6): 1220-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26483345

ABSTRACT

Chemokines mediate monocyte adhesion to dysfunctional endothelial cells (ECs) and promote arterial inflammation during atherosclerosis. Hypoxia-inducible factor (HIF)-1α is expressed in various cell types of atherosclerotic lesions and is associated with lesional inflammation. However, the impact of endothelial HIF-1α in atherosclerosis is unclear. HIF-1α was detectable in the nucleus of ECs covering murine and human atherosclerotic lesions. To study the role of endothelial HIF-1α in atherosclerosis, deletion of the Hif1a gene was induced in ECs from apolipoprotein E knockout mice (EC-Hif1a(-/-)) by Tamoxifen injection. The formation of atherosclerotic lesions, the lesional macrophage accumulation, and the expression of CXCL1 in ECs were reduced after partial carotid ligation in EC-Hif1a(-/-) compared with control mice. Moreover, the lesion area and the lesional macrophage accumulation were decreased in the aortas of EC-Hif1a(-/-) mice compared with control mice during diet-induced atherosclerosis. In vitro, mildly oxidized low-density lipoprotein or lysophosphatidic acid 20:4 increased endothelial CXCL1 expression and monocyte adhesion by inducing HIF-1α expression. Moreover, endothelial Hif1a deficiency resulted in downregulation of miR-19a in atherosclerotic arteries determined by microRNA profiling. In vitro, HIF-1α-induced miR-19a expression mediated the upregulation of CXCL1 in mildly oxidized low-density lipoprotein-stimulated ECs. These results indicate that hyperlipidemia upregulates HIF-1α expression in ECs by mildly oxidized low-density lipoprotein-derived unsaturated lysophosphatidic acid. Endothelial HIF-1α promoted atherosclerosis by triggering miR-19a-mediated CXCL1 expression and monocyte adhesion, indicating that inhibition of the endothelial HIF-1α/miR-19a pathway may be a therapeutic option against atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MicroRNAs/genetics , Monocytes/metabolism , Animals , Atherosclerosis/metabolism , Blotting, Western , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Endothelial Cells/drug effects , Gene Expression/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipoproteins, LDL/pharmacology , Mice, Knockout , Oligonucleotides, Antisense/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
17.
Thromb Haemost ; 107(2): 356-68, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22234341

ABSTRACT

Impaired endothelial recovery after the implantation of drug-eluting stents is a major concern because of the increased risk for late stent thrombosis. The disruption of the chemokine axis CXCL12/CXCR4 inhibits neointima formation by blocking the recruitment of smooth muscle progenitor cells. To directly compare a CXCR4-targeting treatment strategy with drugs that are currently used for stent coating, we studied the effects of the CXCR4 antagonist POL5551 and the drug sirolimus on neointima formation. Apolipoprotein E-deficient mice were treated with POL5551 or sirolimus continuously for 28 days after a carotid wire injury. POL5551 inhibited neointima formation by 63% (for a dosage of 2 mg/kg/day) and by 70% (for a dosage of 20 mg/kg/day). In comparison, sirolimus reduced the neointimal area by 69%. In contrast to treatment with POL5551 during the first three days after injury, injection of POL5551 (20 mg/kg) once per day for 28 days diminished neointimal hyperplasia by 53%. An analysis of the cellular composition of the neointima showed a reduction in the relative smooth muscle cell (SMC) and macrophage content in mice that had been treated with a high dose of POL5551. In contrast, the diminished SMC content after sirolimus treatment was associated with a neointimal enrichment of macrophages. Furthermore, endothelial recovery was impaired by sirolimus, but not by POL5551. Therefore, the inhibition of CXCR4 by POL5551 is equally effective in preventing neointima formation as sirolimus, but POL5551 might be more beneficial because treatment with it results in a more stable lesion phenotype and because it does not impair re-endothelialisation.


Subject(s)
Angioplasty , Blood Vessel Prosthesis Implantation , Endothelium, Vascular/drug effects , Lysophospholipids/administration & dosage , Neointima/prevention & control , Postoperative Complications/prevention & control , Proteins/administration & dosage , Animals , Apolipoproteins E/genetics , Carotid Arteries/pathology , Carotid Arteries/surgery , Cell Movement/drug effects , Coronary Restenosis/etiology , Coronary Restenosis/prevention & control , Disease Models, Animal , Drug-Eluting Stents/statistics & numerical data , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Lysophospholipids/adverse effects , Lysophospholipids/chemistry , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Neointima/etiology , Postoperative Complications/etiology , Proteins/adverse effects , Receptors, CXCR4/antagonists & inhibitors , Sirolimus/administration & dosage , Sirolimus/adverse effects
18.
Cell Metab ; 13(5): 592-600, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21531341

ABSTRACT

Oxidatively modified low-density lipoprotein (oxLDL) plays a key role in the initiation of atherosclerosis by increasing monocyte adhesion. The mechanism that is responsible for the oxLDL-induced atherogenic monocyte recruitment in vivo, however, still remains unknown. Oxidation of LDL generates lysophosphatidylcholine, which is the main substrate for the lysophosphatidic acid (LPA) generating enzyme autotaxin. We show that oxLDL requires endothelial LPA receptors and autotaxin to elicit CXCL1-dependent arterial monocyte adhesion. Unsaturated LPA releases endothelial CXCL1, which is subsequently immobilized on the cell surface and mediates LPA-induced monocyte adhesion. Local and systemic application of LPA accelerates the progression of atherosclerosis in mice. Blocking the LPA receptors LPA(1) and LPA(3) reduced hyperlipidemia-induced arterial leukocyte arrest and atherosclerosis in the presence of functional CXCL1. Thus, atherogenic monocyte recruitment mediated by hyperlipidemia and modified LDL crucially depends on LPA, which triggers endothelial deposition of CXCL1, revealing LPA signaling as a target for cardiovascular disease treatments.


Subject(s)
Atherosclerosis/chemically induced , Atherosclerosis/metabolism , Chemokine CXCL1/metabolism , Endothelium, Vascular/metabolism , Hyperlipidemias , Lipoproteins, LDL/metabolism , Lysophospholipids/pharmacology , Animals , Apolipoproteins E/physiology , Carotid Arteries/cytology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Cells, Cultured , Diet, Atherogenic , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , RNA, Small Interfering/genetics , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism
19.
Circ Res ; 107(1): 96-105, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20360252

ABSTRACT

RATIONALE: The chemokine CXCL12 (CXC motif ligand 12) and its receptor CXCR 4 (CXC motif receptor 4) direct the recruitment of smooth muscle progenitor cells (SPCs) in neointima formation after vascular injury. Lysophosphatidic acid (LPA) induces CXCL12 and neointimal accumulation of smooth muscle cells (SMCs) in uninjured arteries. Thus, we hypothesize that LPA may regulate CXCL12-mediated vascular remodelling. OBJECTIVES: We evaluated the role of LPA receptors in initiating CXCL12-dependent vascular repair by SPCs. METHODS AND RESULTS: Wire-induced carotid injury was performed in apolipoprotein E(-/-) mice on western-type diet. LPA receptor expression was studied by immunostaining and quantitative RT-PCR. LPA receptors LPA(1) and LPA(3) were detected in the media of uninjured arteries and in the injury-induced neointima. LPA(3) mRNA was upregulated and LPA(1) mRNA downregulated at one week after injury. The LPA(1/3) antagonist Ki16425 inhibited neointima formation by 71% and reduced both relative neointimal SMCs and the macrophage content. Additionally, neointimal hypoxia-inducible factor-1alpha and CXCL12 expression, the injury-induced peripheral stem cell antigen-1 (Sca-1)(+)/Lin(-) SPC mobilization, and the neointimal recruitment of Sca-1(+)SMCs were inhibited by Ki16425. In wild type mice, LPA20:4 increased CXCL12 and hypoxia-inducible factor-1alpha expression in carotid arteries as early as 1 day following short-term endoluminal incubation. LPA20:4-induced SPC mobilization and neointima formation were blocked by Ki16425, LPA(1)- and LPA(3)-specific small interfering (si)RNA, and the CXCR4 antagonist POL5551. Ki16425 reduced LPA20:4-mediated neointimal recruitment of SPC as demonstrated by 2-photon microscopy in bone marrow chimeric mice after repopulation with SM22-LacZ transgenic, hematopoietic cells. Moreover, POL5551 decreased the neointimal accumulation of CXCR4(+) SMCs. CONCLUSIONS: LPA(1) and LPA(3) promote neointima formation through activation of CXCL12-mediated mobilization and recruitment of SPCs.


Subject(s)
Cell Movement/physiology , Chemokine CXCL12/physiology , Myocytes, Smooth Muscle/physiology , Receptors, Lysophosphatidic Acid/physiology , Stem Cells/physiology , Tunica Intima/physiology , Animals , Carotid Artery, Common/pathology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/cytology , Stem Cells/cytology , Tunica Intima/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...