Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
ACR Open Rheumatol ; 2(9): 512-524, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32869536

ABSTRACT

OBJECTIVE: RasGTPases are master regulators of multiple intracellular signaling cascades. Perturbation of this pathway has been implicated in the pathogenesis of rheumatoid arthritis (RA). In this study we aimed to define the therapeutic potential of a novel RasGTPases inhibitor, farnesylthiosalicylate (FTS), in the preclinical mouse model of collagen-induced arthritis (CIA) and better delineate its immunomodulatory effects both ex vivo and in the mouse. METHODS: We analyzed in vitro the immunomodulatory effects of FTS on various CD4+ T-cell functions such as activation, proliferation, T-helper polarization, and production of proinflammatory cytokines. Using the CIA model, we further determined the efficacy of FTS to inhibit clinical, histopathologic, and diverse immunological outcomes of arthritis. RESULTS: FTS treatment of CD4+ T cells in vitro effectively targeted distinct kinases (extracellular signal-regulated kinase 1/2, p38, protein kinase B/AKT, and mammalian target of rapamycin), the production of interleukin (IL)-17A, IL-22, and granulocyte-macrophage colony-stimulating factor, and Th17 polarization. FTS therapy in the mouse CIA model significantly reduced clinical disease severity and joint inflammation/damage by histology. Importantly, FTS suppressed the in vivo induction of splenic IL-17+ IL-22+ Th17 cells and the secretion of proinflammatory cytokines. The production of pathogenic autoantibodies and their abnormal hyposialylation was significantly attenuated by FTS therapy. Importantly, in vivo generation of collagen type-II specific effector CD4+ T cells was likewise repressed by FTS therapy. CONCLUSION: The RasGTPases inhibitor FTS attenuates the production of proinflammatory cytokines by in vitro-activated T cells and is a potent immunomodulatory compound in the CIA model, primarily targeting the generation of autoreactive Th17 cells and the production of autoantibodies and their subsequent pathogenic hyposialylation.

2.
Oncogenesis ; 8(9): 48, 2019 Sep 02.
Article in English | MEDLINE | ID: mdl-31477684

ABSTRACT

Pleuropulmonary blastoma (PPB) is a rare pediatric lung neoplasm that recapitulates developmental pathways of early embryonic lungs. As lung development proceeds with highly regulated mesenchymal-epithelial interactions, a DICER1 mutation in PPB generates a faulty lung differentiation program with resultant biphasic tumors composed of a primitive epithelial and mesenchymal stroma with early progenitor blastomatous cells. Deciphering of PPB progression has been hampered by the difficulty of culturing PPB cells, and specifically progenitor blastomatous cells. Here, we show that in contrast with in-vitro culture, establishment of PPB patient-derived xenograft (PDX) in NOD-SCID mice selects for highly proliferating progenitor blastoma overexpressing critical regulators of lung development and multiple imprinted genes. These stem-like tumors were sequentially interrogated by gene profiling to show a FGF module that is activated alongside Neural cell adhesion molecule 1 (NCAM1). Targeting the progenitor blastoma and these transitions with an anti-NCAM1 immunoconjugate (Lorvotuzumab mertansine) inhibited tumor growth and progression providing new paradigms for PPB therapeutics. Altogether, our novel in-vivo PPB xenograft model allowed us to enrich for highly proliferating stem-like cells and to identify FGFR and NCAM1 as two key players that can serve as therapeutic targets in this poorly understood and aggressive disease.

3.
Stem Cell Reports ; 11(3): 795-810, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30122444

ABSTRACT

Cancer stem cell (CSC) identification relies on transplantation assays of cell subpopulations sorted from fresh tumor samples. Here, we attempt to bypass limitations of abundant tumor source and predetermined immune selection by in vivo propagating patient-derived xenografts (PDX) from human malignant rhabdoid tumor (MRT), a rare and lethal pediatric neoplasm, to an advanced state in which most cells behave as CSCs. Stemness is then probed by comparative transcriptomics of serial PDXs generating a gene signature of epithelial to mesenchymal transition, invasion/motility, metastasis, and self-renewal, pinpointing putative MRT CSC markers. The relevance of these putative CSC molecules is analyzed by sorting tumorigenic fractions from early-passaged PDX according to one such molecule, deciphering expression in archived primary tumors, and testing the effects of CSC molecule inhibition on MRT growth. Using this platform, we identify ALDH1 and lysyl oxidase (LOX) as relevant targets and provide a larger framework for target and drug discovery in rare pediatric cancers.


Subject(s)
Carcinogenesis/pathology , Neoplasm Invasiveness/pathology , Neoplastic Stem Cells/pathology , Rhabdoid Tumor/pathology , Aldehyde Dehydrogenase 1 Family , Animals , Epithelial-Mesenchymal Transition , Female , Humans , Isoenzymes/analysis , Mice, Inbred NOD , Mice, SCID , Protein-Lysine 6-Oxidase/analysis , Retinal Dehydrogenase/analysis , Tumor Cells, Cultured
5.
Mol Cancer Ther ; 16(11): 2462-2472, 2017 11.
Article in English | MEDLINE | ID: mdl-28729402

ABSTRACT

Cancer stem cells (CSC) form a specific population within the tumor that has been shown to have self-renewal and differentiation properties, increased ability to migrate and form metastases, and increased resistance to chemotherapy. Consequently, even a small number of cells remaining after therapy can repopulate the tumor and cause recurrence of the disease. CSCs in Wilms tumor, a pediatric renal cancer, were previously shown to be characterized by neural cell adhesion molecule (NCAM) expression. Therefore, NCAM provides a specific biomarker through which the CSC population in this tumor can be targeted. We have recently developed an NCAM-targeted nanosized conjugate of paclitaxel bound to a biodegradable polyglutamic acid polymer. In this work, we examined the ability of the conjugate to inhibit Wilms tumor by targeting the NCAM-expressing CSCs. Results show that the conjugate selectively depleted the CSC population of the tumors and effectively inhibited tumor growth without causing toxicity. We propose that the NCAM-targeted conjugate could be an effective therapeutic for Wilms tumor. Mol Cancer Ther; 16(11); 2462-72. ©2017 AACR.


Subject(s)
Nanoconjugates/administration & dosage , Neural Cell Adhesion Molecules/genetics , Paclitaxel/administration & dosage , Wilms Tumor/drug therapy , Cell Differentiation/drug effects , Cell Self Renewal , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Nanoconjugates/chemistry , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Paclitaxel/chemistry , Polymers/administration & dosage , Polymers/chemistry , Wilms Tumor/genetics , Wilms Tumor/pathology
6.
Front Immunol ; 8: 799, 2017.
Article in English | MEDLINE | ID: mdl-28736556

ABSTRACT

The Ras family of GTPases plays an important role in signaling nodes downstream to T cell receptor and CD28 activation, potentially lowering the threshold for T-cell receptor activation by autoantigens. Somatic mutation in NRAS or KRAS may cause a rare autoimmune disorder coupled with abnormal expansion of lymphocytes. T cells from rheumatoid arthritis (RA) patients show excessive activation of Ras/MEK/ERK pathway. The small molecule farnesylthiosalicylic acid (FTS) interferes with the interaction between Ras GTPases and their prenyl-binding chaperones to inhibit proper plasma membrane localization. In the present study, we tested the therapeutic and immunomodulatory effects of FTS and its derivative 5-fluoro-FTS (F-FTS) in the rat adjuvant-induced arthritis model (AIA). We show that AIA severity was significantly reduced by oral FTS and F-FTS treatment compared to vehicle control treatment. FTS was as effective as the mainstay anti-rheumatic drug methotrexate, and combining the two drugs significantly increased efficacy compared to each drug alone. We also discovered that FTS therapy inhibited both the CFA-driven in vivo induction of Th17 and IL-17/IFN-γ producing "double positive" as well as the upregulation of serum levels of the Th17-associated cytokines IL-17A and IL-22. By gene microarray analysis of effector CD4+ T cells from CFA-immunized rats, re-stimulated in vitro with the mycobacterium tuberculosis heat-shock protein 65 (Bhsp65), we determined that FTS abrogated the Bhsp65-induced transcription of a large list of genes (e.g., Il17a/f, Il22, Ifng, Csf2, Lta, and Il1a). The functional enrichment bioinformatics analysis showed significant overlap with predefined gene sets related to inflammation, immune system processes and autoimmunity. In conclusion, FTS and F-FTS display broad immunomodulatory effects in AIA with inhibition of the Th17-type response to a dominant arthritogenic antigen. Hence, targeting Ras signal-transduction cascade is a potential novel therapeutic approach for RA.

7.
EMBO Mol Med ; 9(4): 508-530, 2017 04.
Article in English | MEDLINE | ID: mdl-28275008

ABSTRACT

Angiomyolipoma (AML), the most common benign renal tumor, can result in severe morbidity from hemorrhage and renal failure. While mTORC1 activation is involved in its growth, mTORC1 inhibitors fail to eradicate AML, highlighting the need for new therapies. Moreover, the identity of the AML cell of origin is obscure. AML research, however, is hampered by the lack of in vivo models. Here, we establish a human AML-xenograft (Xn) model in mice, recapitulating AML at the histological and molecular levels. Microarray analysis demonstrated tumor growth in vivo to involve robust PPARγ-pathway activation. Similarly, immunostaining revealed strong PPARγ expression in human AML specimens. Accordingly, we demonstrate that while PPARγ agonism accelerates AML growth, PPARγ antagonism is inhibitory, strongly suppressing AML proliferation and tumor-initiating capacity, via a TGFB-mediated inhibition of PDGFB and CTGF. Finally, we show striking similarity between AML cell lines and mesenchymal stem cells (MSCs) in terms of antigen and gene expression and differentiation potential. Altogether, we establish the first in vivo human AML model, which provides evidence that AML may originate in a PPARγ-activated renal MSC lineage that is skewed toward adipocytes and smooth muscle and away from osteoblasts, and uncover PPARγ as a regulator of AML growth, which could serve as an attractive therapeutic target.


Subject(s)
Angiomyolipoma/pathology , PPAR gamma/metabolism , Animals , Cell Line, Tumor , Connective Tissue Growth Factor/metabolism , Gene Expression Profiling , Humans , Mesenchymal Stem Cells , Mice , Proto-Oncogene Proteins c-sis/metabolism , Therapeutics , Transforming Growth Factor beta/metabolism
8.
Oncotarget ; 7(34): 54370-54379, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27494846

ABSTRACT

Adenosine deaminase acting on RNA (ADAR) 1 is the master editor of the transcriptome, catalyzing the conversion of adenosine to inosine (A-to-I). RNA transcripts fold into a variety of secondary structures including long intramolecular RNA duplexes that are the major substrate of ADAR1. Most A-to-I editing sites occur within RNA duplexes formed by complementary pairing of inverted retrotransposable elements interspersed within noncoding regions of transcripts. This catalytic activity of ADAR1 most likely prevents the abnormal activation of cytosolic nucleic acid sensors by self-dsRNAs. Homozygous disruption of mouse Adar is embryonic lethal due to a toxic type-I interferons response and correspondingly biallelic missense mutations in human ADAR1 cause a severe congenital interferonopathy. Here, we report that Cd19-Cre-mediated Adar gene ablation in the mouse causes a significant defect in the final stages of B cell development with an almost complete absence of newly formed immature and CD23+ mature recirculating B cells in the BM. Adar ablation in pre-B cells induced upregulation of typical interferon-stimulated genes (ISGs) and apoptosis upon further maturation. ADAR1 deficiency also inhibited the in vitro, IL-7-mediated, differentiation of BM-derived B cell precursors. In summary, ADAR1 is required, non-redundantly, for normal B lymphopoiesis in the BM and peripheral maintenance.


Subject(s)
Adenosine Deaminase/physiology , B-Lymphocytes/physiology , Bone Marrow/physiology , Cell Lineage/physiology , Lymphopoiesis , Animals , Antigens, CD19/physiology , Apoptosis , Interleukin-7/physiology , Mice , Mice, Inbred C57BL
9.
Sci Rep ; 6: 23562, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-27020553

ABSTRACT

When assembling a nephron during development a multipotent stem cell pool becomes restricted as differentiation ensues. A faulty differentiation arrest in this process leads to transformation and initiation of a Wilms' tumor. Mapping these transitions with respective surface markers affords accessibility to specific cell subpopulations. NCAM1 and CD133 have been previously suggested to mark human renal progenitor populations. Herein, using cell sorting, RNA sequencing, in vitro studies with serum-free media and in vivo xenotransplantation we demonstrate a sequential map that links human kidney development and tumorigenesis; In nephrogenesis, NCAM1(+)CD133(-) marks SIX2(+) multipotent renal stem cells transiting to NCAM1(+)CD133(+) differentiating segment-specific SIX2(-) epithelial progenitors and NCAM1(-)CD133(+) differentiated nephron cells. In tumorigenesis, NCAM1(+)CD133(-) marks SIX2(+) blastema that includes the ALDH1(+) WT cancer stem/initiating cells, while NCAM1(+)CD133(+) and NCAM1(-)CD133(+) specifying early and late epithelial differentiation, are severely restricted in tumor initiation capacity and tumor self-renewal. Thus, negative selection for CD133 is required for defining NCAM1(+) nephron stem cells in normal and malignant nephrogenesis.


Subject(s)
Biomarkers/metabolism , Carcinogenesis/genetics , Kidney/metabolism , Neoplastic Stem Cells/metabolism , Nephrons/metabolism , Stem Cells/metabolism , AC133 Antigen/genetics , AC133 Antigen/metabolism , Animals , CD56 Antigen/genetics , CD56 Antigen/metabolism , Carcinogenesis/metabolism , Cells, Cultured , Child , Child, Preschool , Female , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Infant , Kidney/embryology , Male , Mice, Inbred NOD , Neoplastic Stem Cells/pathology , Nephrons/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organogenesis/genetics , Prospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Cells, Cultured
10.
Stem Cell Reports ; 3(1): 24-33, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25068119

ABSTRACT

An open question remains in cancer stem cell (CSC) biology whether CSCs are by definition at the top of the differentiation hierarchy of the tumor. Wilms' tumor (WT), composed of blastema and differentiated renal elements resembling the nephrogenic zone of the developing kidney, is a valuable model for studying this question because early kidney differentiation is well characterized. WT neural cell adhesion molecule 1-positive (NCAM1(+)) aldehyde dehydrogenase 1-positive (ALDH1(+)) CSCs have been recently isolated and shown to harbor early renal progenitor traits. Herein, by generating pure blastema WT xenografts, composed solely of cells expressing the renal developmental markers SIX2 and NCAM1, we surprisingly show that sorted ALDH1(+) WT CSCs do not correspond to earliest renal stem cells. Rather, gene expression and proteomic comparative analyses disclose a cell type skewed more toward epithelial differentiation than the bulk of the blastema. Thus, WT CSCs are likely to dedifferentiate to propagate WT blastema.


Subject(s)
Neoplastic Stem Cells/pathology , Wilms Tumor/metabolism , Wilms Tumor/pathology , Aldehyde Dehydrogenase 1 Family , Animals , CD56 Antigen/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Female , Humans , Isoenzymes/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, SCID , Models, Biological , Neoplastic Stem Cells/metabolism , Retinal Dehydrogenase/metabolism
11.
PLoS One ; 9(3): e90879, 2014.
Article in English | MEDLINE | ID: mdl-24621570

ABSTRACT

The development of the mammalian kidney is a highly complex process dependent upon the interplay of various cell types, secreted morphogens, and the extra-cellular matrix (ECM). Although integrins are the most important receptors for ECM proteins and are ubiquitously expressed during kidney development, mice lacking expression of integrin α3 (Itga3) do not demonstrate a reduced number of nephrons, but mostly a disorganized GBM (glomerular basement membrane) leading to proteinuria. Thus, ITGA3 is considered mostly a passive GBM stabilizer and not an active player in nephrogenesis. Recently, mutations in the human ITGA3 were shown to cause congenital nephrotic syndrome, epidermolysis bullosa and interstitial lung disease, otherwise termed NEP syndrome (Nephrotic syndrome, Epidermolysis bullosa and Pulmonary disease). Herein, we performed histological and molecular analysis on the kidneys of a single patient from the initial cohort harboring an ITGA3 mutation, to illuminate the role of ITGA3 in human renal development. We show the patient to harbor a unique phenotype at birth, including severe unilateral renal hypodysplasia. Interrogation of global gene expression in the hypodysplastic kidney versus three controls (fetal, child and adult kidneys) revealed perturbed expression in several renal developmental pathways implicated in hypodysplasia, including the Wnt, BMP (bone morphogenetic protein) and TGF (transforming growth factor) pathways. Moreover, the affected kidney showed upregulation of early embryonic genes (e.g. OCT4 and PAX8) concomitant with downregulated kidney differentiation markers, implying a defect in proper renal differentiation. In conclusion, we show for the first time that ITGA3 is not merely a passive anchor for renal ECM proteins, as predicted by mouse models. Instead, our results may suggest it plays a central role in the interplay of cells, morphogens and ECM, required for proper nephrogenesis, thus adding ITGA3 to the list of CAKUT (congenital anomalies of the kidney and urinary tract)-causing genes.


Subject(s)
Integrin alpha3/genetics , Kidney/growth & development , Mutation , Adult , Animals , Epidermolysis Bullosa/genetics , Gene Expression Regulation, Developmental , Humans , Integrin alpha3/metabolism , Lung Diseases/genetics , Mice , Nephrotic Syndrome/genetics , Phenotype , Protein Transport
12.
Am J Pathol ; 183(5): 1621-1633, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24055371

ABSTRACT

The nephron is composed of a monolayer of epithelial cells that make up its various compartments. In development, these cells begin as mesenchyme. NCAM1, abundant in the mesenchyme and early nephron lineage, ceases to express in mature kidney epithelia. We show that, once placed in culture and released from quiescence, adult human kidney epithelial cells (hKEpCs), uniformly positive for CD24/CD133, re-express NCAM1 in a specific cell subset that attains a stem/progenitor state. Immunosorted NCAM1(+) cells overexpressed early nephron progenitor markers (PAX2, SALL1, SIX2, WT1) and acquired a mesenchymal fate, indicated by high vimentim and reduced E-cadherin levels. Gene expression and microarray analysis disclosed both a proximal tubular origin of these cells and molecules regulating epithelial-mesenchymal transition. NCAM1(+) cells generated clonal progeny when cultured in the presence of fetal kidney conditioned medium, differentiated along mesenchymal lineages but retained the unique propensity to generate epithelial kidney spheres and produce epithelial renal tissue on single-cell grafting in chick CAM and mouse. Depletion of NCAM1(+) cells from hKEpCs abrogated stemness traits in vitro. Eliminating these cells during the regenerative response that follows glycerol-induced acute tubular necrosis worsened peak renal injury in vivo. Thus, higher clone-forming and developmental capacities characterize a distinct subset of adult kidney-derived cells. The ability to influence an endogenous regenerative response via NCAM1 targeting may lead to novel therapeutics for renal diseases.


Subject(s)
CD56 Antigen/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Kidney/pathology , Stem Cells/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Adult , Animals , Antibodies/metabolism , Biomarkers/metabolism , Blood Urea Nitrogen , Cell Differentiation/genetics , Cell Proliferation , Chickens , Clone Cells , Down-Regulation/genetics , Gene Ontology , HEK293 Cells , Humans , Mesoderm/pathology , Mice , Molecular Sequence Annotation , Nephrons/metabolism , Nephrons/pathology , Oligonucleotide Array Sequence Analysis , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcriptome/genetics , Up-Regulation/genetics
13.
J Hepatol ; 55(5): 1086-94, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21354232

ABSTRACT

BACKGROUND & AIMS: The identification of the cellular and molecular pathways that mediate the development of non-alcoholic steatohepatitis is of crucial importance. Cytokines produced by liver-resident and infiltrating inflammatory cells, play a pivotal role in liver inflammation. The role of the proinflammatory cytokines IL-1α and IL-1ß in steatohepatitis remains elusive. METHODS: We employed IL-1α and IL-1ß-deficient mice and transplanted marrow cells to study the role of liver-resident and bone marrow-derived IL-1 in steatosis and its progression to steatohepatitis. RESULTS: Atherogenic diet-induced steatohepatitis in wild-type mice was associated with 16 and 4.6 fold-elevations in mRNA levels of hepatic IL-1α and IL-1ß, respectively. In mice deficient in either IL-1α or IL-1ß the transformation of steatosis to steatohepatitis and liver fibrosis was markedly reduced. This protective effect in IL-1α-deficient mice was noted despite increased liver cholesterol levels. Deficiency of IL-1α markedly reduced plasma serum amyloid A and steady-state levels of mRNA coding for inflammatory genes (P-selectin, CXCL1, IL-6, and TNFα) as well as pro-fibrotic genes (MMP-9 and Collagen) and particularly a 50% decrease in TGFß levels (p = 0.004). IL-1α mRNA levels were two-folds lower in IL-1ß-deficient mice, and IL-1ß transcripts were three-folds lower in IL-1α-deficient compared to wild-type mice. Hepatic cell derived IL-1α rather than from recruited bone marrow-derived cells was required for steatohepatitis development. CONCLUSIONS: These data demonstrate the critical role of IL-1α and IL-1ß in the transformation of steatosis to steatohepatitis and liver fibrosis in hypercholesterolemic mice. Therefore, the potential of neutralizing IL-1α and/or IL-1ß to inhibit the development of steatohepatitis should be explored.


Subject(s)
Fatty Liver/metabolism , Hepatitis/metabolism , Interleukin-1alpha/deficiency , Interleukin-1beta/deficiency , Liver Cirrhosis/metabolism , RNA, Messenger/metabolism , Analysis of Variance , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Collagen/genetics , Collagen/metabolism , Diet, Atherogenic , Disease Progression , Fatty Liver/pathology , Gene Expression , Hepatitis/pathology , Hypercholesterolemia/complications , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Liver Cirrhosis/pathology , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/genetics , P-Selectin/metabolism , Serum Amyloid A Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
Biochem Biophys Res Commun ; 405(2): 197-203, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21219852

ABSTRACT

OBJECTIVE: Interleukin (IL)-1α and IL-1ß are products of macrophages, endothelial cells and vascular smooth muscle cells; moreover, each of these cell types is affected by the pro-inflammatory properties of both IL-1's. Whereas several studies demonstrate the proatherogenic properties of IL-1ß, the role of IL-1α in atherogenesis remains unclear. We assessed whether IL-1α and IL-1ß from tissue resident vascular cells or emigrating bone marrow-derived cells promote the development of atherosclerosis in apoE-/- mice and determined the effect of selective macrophage IL-1α or IL-1ß deficiency on degradation of LDL and cytokine production. METHODS: We generated strains of double knock-out (KO) mice (apoE-/-/IL-1α-/- and apoE-/-/IL-1ß-/-) and created chimeras consisting of apoE-/- mice reconstituted with bone marrow-derived cells from apoE-/-/IL-1+/+, apoE-/-/IL-1α-/- and apoE-/-/IL-1ß-/-. RESULTS: The areas of aortic sinus lesions were lower in either double KO mice compared to solely apoE-/- mice, despite higher non-HDL cholesterol levels. Importantly, selective deficiency of IL-1α or IL-1ß in bone marrow-derived cells inhibited atherogenesis to the same extent as in double KO mice without affecting plasma lipids. Aortic sinus lesions in apoE-/- mice transplanted with IL-1ß-/- or IL-1α-/- cells were 32% and 52% lower, respectively, than in IL-1+/+ transplanted mice. Ex vivo, isolated IL-1α-/- macrophages from atherosclerotic mice degraded LDL and secreted IL-6, TNFα and IL-12 similarly to IL-1+/+ macrophages; however, IL-1α deficient macrophages secreted reduced levels of IL-1ß (-50%) and 2-3-fold higher levels of the anti-inflammatory cytokine IL-10. CONCLUSION: We show for the first time that it is IL-1α from bone marrow-derived cells that accelerates atherogenesis in apoE-deficient mice rather than constitutive IL-1α in vascular cells, possibly by increasing the inflammatory cytokine profile of macrophages.


Subject(s)
Atherosclerosis/metabolism , Bone Marrow/metabolism , Cytokines/antagonists & inhibitors , Interleukin-1alpha/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Foam Cells/metabolism , Interleukin-1alpha/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...