Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Molecules ; 27(6)2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35335304

ABSTRACT

Bryophytes produce rare and bioactive compounds with a broad range of therapeutic potential, and many species are reported in ethnomedicinal uses. However, only a few studies have investigated their potential as natural anti-inflammatory drug candidate compounds. The present study investigates the anti-inflammatory effects of thirty-two species of bryophytes, including mosses and liverworts, on Raw 264.7 murine macrophages stimulated with lipopolysaccharide (LPS) or recombinant human peroxiredoxin (hPrx1). The 70% ethanol extracts of bryophytes were screened for their potential to reduce the production of nitric oxide (NO), an important pro-inflammatory mediator. Among the analyzed extracts, two moss species significantly inhibited LPS-induced NO production without cytotoxic effects. The bioactive extracts of Dicranum majus and Thuidium delicatulum inhibited NO production in a concentration-dependent manner with IC50 values of 1.04 and 1.54 µg/mL, respectively. The crude 70% ethanol and ethyl acetate extracts were then partitioned with different solvents in increasing order of polarity (n-hexane, diethyl ether, chloroform, ethyl acetate, and n-butanol). The fractions were screened for their inhibitory effects on NO production stimulated with LPS at 1 ng/mL or 10 ng/mL. The NO production levels were significantly affected by the fractions of decreasing polarity such as n-hexane and diethyl ether ones. Therefore, the potential of these extracts to inhibit the LPS-induced NO pathway suggests their effective properties in attenuating inflammation and could represent a perspective for the development of innovative therapeutic agents.


Subject(s)
Bryophyta , Lipopolysaccharides , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Humans , Lipopolysaccharides/pharmacology , Macrophages , Mice , Plant Extracts/metabolism , Plant Extracts/pharmacology
2.
Lab Invest ; 96(5): 570-80, 2016 05.
Article in English | MEDLINE | ID: mdl-26927515

ABSTRACT

Abnormal Wnt signaling is associated with bone mass disorders. Frizzled-related protein (FRZB, also known as secreted frizzled-related protein-3 (SFRP3)) is a Wnt modulator that contains an amino-terminal cysteine-rich domain (CRD) and a carboxy-terminal Netrin-like (NTN) motif. Frzb(-/-) mice show increased cortical thickness. However, the direct effect of FRZB on osteogenic differentiation and the involvement of the structural domains herein are not fully understood. In this study, we observed that stable overexpression of Frzb in MC3T3-E1 cells increased calcium deposition and osteoblast markers compared with control. Western blot analysis showed that the increased osteogenesis was associated with reduced canonical, but increased non-canonical Wnt signaling. On the contrary, loss of Frzb induced the opposite effects on osteogenesis and Wnt signaling. To translationally validate the positive effects of FRZB on primary human cells, we treated human periosteal and human bone marrow stromal cells with conditioned medium from MC3T3-E1 cells overexpressing Frzb and observed an increase in Alizarin red staining. We further studied the effect of the domains. FrzbNTN overexpression induced similar effects on osteogenesis as full-length Frzb, whereas FrzbCRD overexpressing cells mimicked loss of Frzb experiments. The CRD is considered as the Wnt binding domain, but the NTN domain also has important effects on bone biology. FRZB and other SFRPs or their specific domains may hold surprising potential as therapeutics for bone and joint disorders considering that excess of SFRPs has effects that are not expected under physiological, endogenous expression conditions.


Subject(s)
Glycoproteins/physiology , Osteogenesis/physiology , 3T3 Cells , Animals , Calcification, Physiologic , Cell Differentiation , Cells, Cultured , Culture Media, Conditioned , Gene Knockdown Techniques , Glycoproteins/chemistry , Glycoproteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics , Protein Domains , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology
3.
Ann Rheum Dis ; 74(10): 1861-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-24928840

ABSTRACT

OBJECTIVES: To identify genetic associations with hip osteoarthritis (HOA), we performed a meta-analysis of genome-wide association studies (GWAS) of HOA. METHODS: The GWAS meta-analysis included approximately 2.5 million imputed HapMap single nucleotide polymorphisms (SNPs). HOA cases and controls defined radiographically and by total hip replacement were selected from the Osteoporotic Fractures in Men (MrOS) Study and the Study of Osteoporotic Fractures (SOF) (654 cases and 4697 controls, combined). Replication of genome-wide significant SNP associations (p ≤5×10(-8)) was examined in five studies (3243 cases and 6891 controls, combined). Functional studies were performed using in vitro models of chondrogenesis and osteogenesis. RESULTS: The A allele of rs788748, located 65 kb upstream of the IGFBP3 gene, was associated with lower HOA odds at the genome-wide significance level in the discovery stage (OR 0.71, p=2×10(-8)). The association replicated in five studies (OR 0.92, p=0.020), but the joint analysis of discovery and replication results was not genome-wide significant (p=1×10(-6)). In separate study populations, the rs788748 A allele was also associated with lower circulating IGFBP3 protein levels (p=4×10(-13)), suggesting that this SNP or a variant in linkage disequilibrium could be an IGFBP3 regulatory variant. Results from functional studies were consistent with association results. Chondrocyte hypertrophy, a deleterious event in OA pathogenesis, was largely prevented upon IGFBP3 knockdown in chondrocytes. Furthermore, IGFBP3 overexpression induced cartilage catabolism and osteogenic differentiation. CONCLUSIONS: Results from GWAS and functional studies provided suggestive links between IGFBP3 and HOA.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/genetics , Osteoarthritis, Hip/genetics , Aged , Aged, 80 and over , Case-Control Studies , Chondrogenesis/genetics , Chondrogenesis/physiology , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Insulin-Like Growth Factor Binding Protein 3/physiology , Male , Middle Aged , Osteoarthritis, Hip/physiopathology , Osteogenesis/genetics , Osteogenesis/physiology , Polymorphism, Single Nucleotide
4.
Respir Res ; 16: 38, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25849157

ABSTRACT

BACKGROUND: Effective treatments for fibrotic diseases such as idiopathic pulmonary fibrosis are largely lacking. Transforming growth factor beta (TGFß) plays a central role in the pathophysiology of fibrosis. We hypothesized that bone morphogenetic proteins (BMP), another family within the TGFß superfamily of growth factors, modulate fibrogenesis driven by TGFß. We therefore studied the role of endogenous BMP signaling in bleomycin induced lung fibrosis. METHODS: Lung fibrosis was induced in wild-type or noggin haploinsufficient (Nog +/LacZ ) mice by intratracheal instillation of bleomycin, or phosphate buffered saline as a control. Invasive pulmonary function tests were performed using the flexiVent® SCIREQ system. The mice were sacrificed and lung tissue was collected for analysis using histopathology, collagen quantification, immunohistochemistry and gene expression analysis. RESULTS: Nog +/LacZ mice are a known model of increased BMP signaling and were partially protected from bleomycin-induced lung fibrosis with reduced Ashcroft score, reduced collagen content and preservation of pulmonary compliance. In bleomycin-induced lung fibrosis, TGFß and BMP signaling followed an inverse course, with dynamic activation of TGFß signaling and repression of BMP signaling activity. CONCLUSIONS: Upon bleomycin exposure, active BMP signaling is decreased. Derepression of BMP signaling in Nog +/LacZ mice protects against bleomycin-induced pulmonary fibrosis. Modulating the balance between BMP and TGFß, in particular increasing endogenous BMP signals, may therefore be a therapeutic target in fibrotic lung disease.


Subject(s)
Bleomycin , Bone Morphogenetic Proteins/metabolism , Lung/metabolism , Pulmonary Fibrosis/prevention & control , Animals , Apoptosis , Carrier Proteins/genetics , Cell Proliferation , Collagen/metabolism , Disease Models, Animal , Down-Regulation , Haploinsufficiency , Lung/pathology , Lung/physiopathology , Lung Compliance , Male , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism , Time Factors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
5.
Proc Natl Acad Sci U S A ; 109(21): 8218-23, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22566624

ABSTRACT

Hip osteoarthritis (HOA) is one of the most disabling and common joint disorders with a large genetic component that is, however, still ill-defined. To date, genome-wide association studies (GWAS) in osteoarthritis (OA) and specifically in HOA have yielded only few loci, which is partly explained by heterogeneity in the OA definition. Therefore, we here focused on radiographically measured joint-space width (JSW), a proxy for cartilage thickness and an important underlying intermediate trait for HOA. In a GWAS of 6,523 individuals on hip-JSW, we identified the G allele of rs12982744 on chromosome 19p13.3 to be associated with a 5% larger JSW (P = 4.8 × 10(-10)). The association was replicated in 4,442 individuals from three United Kingdom cohorts with an overall meta-analysis P value of 1.1 × 10(-11). The SNP was also strongly associated with a 12% reduced risk for HOA (P = 1 × 10(-4)). The SNP is located in the DOT1L gene, which is an evolutionarily conserved histone methyltransferase, recently identified as a potentially dedicated enzyme for Wnt target-gene activation in leukemia. Immunohistochemical staining of the DOT1L protein in mouse limbs supports a role for DOT1L in chondrogenic differentiation and adult articular cartilage. DOT1L is also expressed in OA articular chondrocytes. Silencing of Dot1l inhibited chondrogenesis in vitro. Dot1l knockdown reduces proteoglycan and collagen content, and mineralization during chondrogenesis. In the ATDC5 chondrogenesis model system, DOT1L interacts with TCF and Wnt signaling. These data are a further step to better understand the role of Wnt-signaling during chondrogenesis and cartilage homeostasis. DOT1L may represent a therapeutic target for OA.


Subject(s)
Chondrocytes/physiology , Chondrogenesis/genetics , Genome-Wide Association Study , Methyltransferases/genetics , Osteoarthritis, Hip/genetics , Age Factors , Animals , Cartilage, Articular/pathology , Cartilage, Articular/physiology , Cell Line , Chondrocytes/cytology , Genetic Variation , Hepatocyte Nuclear Factor 1-alpha/metabolism , Histone-Lysine N-Methyltransferase , Humans , Methyltransferases/metabolism , Mice , Osteoarthritis, Hip/epidemiology , Osteoarthritis, Hip/pathology , Risk Factors , Wnt Signaling Pathway/physiology
6.
J Biol Chem ; 286(22): 19215-28, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21471198

ABSTRACT

Transforming growth factor (TGF)-ß1 stimulates extracellular PP(i) (ePP(i)) generation and promotes chondrocalcinosis, which also occurs secondary to hyperparathyroidism-induced hypercalcemia. We previously demonstrated that ANK was up-regulated by TGF-ß1 activation of ERK1/2 and Ca(2+)-dependent protein kinase C (PKCα). Thus, we investigated mechanisms by which calcium could affect ePP(i) metabolism, especially its main regulating proteins ANK and PC-1 (plasma cell membrane glycoprotein-1). We stimulated articular chondrocytes with TGF-ß1 under extracellular (eCa(2+)) or cytosolic Ca(2+) (cCa(2+)) modulations. We studied ANK, PC-1 expression (quantitative RT-PCR, Western blotting), ePP(i) levels (radiometric assay), and cCa(2+) input (fluorescent probe). Voltage-operated Ca(2+)-channels (VOC) and signaling pathways involved were investigated with selective inhibitors. Finally, Ank promoter activity was evaluated (gene reporter). TGF-ß1 elevated cCa(2+) and ePP(i) levels (by up-regulating Ank and PC-1 mRNA/proteins) in an eCa(2+) dose-dependent manner. TGF-ß1 effects were suppressed by cCa(2+) chelation or L- and T-VOC blockade while being mostly reproduced by ionomycin. In the same experimental conditions, the activation of Ras, the phosphorylation of ERK1/2 and PKCα, and the stimulation of Ank promoter activity were affected similarly. Activation of SP1 (specific protein 1) and ELK-1 (Ets-like protein-1) transcription factors supported the regulatory role of Ca(2+). SP1 or ELK-1 overexpression or blockade experiments demonstrated a major contribution of ELK-1, which acted synergistically with SP1 to activate Ank promoter in response to TGF-ß1. TGF-ß1 promotes input of eCa(2+) through opening of L- and T-VOCs, to potentiate ERK1/2 and PKCα signaling cascades, resulting in an enhanced activation of Ank promoter and ePP(i) production in chondrocyte.


Subject(s)
Calcium/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Diphosphates/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocalcinosis/genetics , Chondrocalcinosis/metabolism , Chondrocalcinosis/pathology , Chondrocytes/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Ion Transport/drug effects , Ion Transport/genetics , Ionomycin/pharmacology , Ionophores/pharmacology , Male , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphate Transport Proteins/genetics , Phosphate Transport Proteins/metabolism , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Promoter Regions, Genetic/genetics , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , Rats , Rats, Wistar , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transforming Growth Factor beta1/pharmacology , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism , ras Proteins/genetics , ras Proteins/metabolism
7.
Clin Transl Med ; 12(10): e1032, 2022 10.
Article in English | MEDLINE | ID: mdl-36245291

ABSTRACT

Psoriasis is a chronic inflammatory disorder affecting skin and joints that results from immunological dysfunction such as enhanced IL-23 induced Th-17 differentiation. IkappaB-Zeta (IκBζ) is an atypical transcriptional factor of the IκB protein family since, contrary to the other family members, it positively regulates NF-κB pathway by being exclusively localized into the nucleus. IκBζ deficiency reduces visible manifestations of experimental psoriasis by diminishing expression of psoriasis-associated genes. It is thus tempting to consider IκBζ as a potential therapeutic target for psoriasis as well as for other IL23/IL17-mediated inflammatory diseases. In this review, we will discuss the regulation of expression of NFKBIZ and its protein IκBζ, its downstream targets, its involvement in pathogenesis of multiple disorders with emphasis on psoriasis and evidences supporting that inhibition of IκBζ may be a promising alternative to current therapeutic managements of psoriasis.


Subject(s)
NF-kappa B , Psoriasis , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Inflammation/genetics , Inflammation/metabolism , Interleukin-23 , NF-kappa B/genetics , NF-kappa B/metabolism , Psoriasis/genetics , Psoriasis/metabolism
8.
J Biol Chem ; 285(14): 10572-82, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20133941

ABSTRACT

The differentiated phenotype of chondrocyte is lost in pathological situations and after interleukin (IL)-1beta challenge. Wnt proteins and the inorganic pyrophosphate (PP(i)) transporter Ank regulate the differentiation process in many cell types. We investigated the possible contribution of Ank and/or PP(i) to the maintenance of the differentiated chondrocyte phenotype with special care to Wnt signaling. Primary articular chondrocytes lost their phenotype upon IL-1beta challenge, with cessation of type II collagen and Sox-9 expression. Ank expression and PP(i) transport were strongly reduced by IL-1beta, whereas Wnt-5a was the only Wnt protein increased. Transient overexpression of Ank counteracted most of IL-1beta effects on Type II collagen, Sox-9, and Wnt-5a expression. When resting chondrocytes were transfected with a siRNA against Ank, this reproduced the phenotype induced by IL-1beta. In both cases, no markers for hypertrophic chondrocytes were detected. The conditioned supernatant from chondrocytes knocked-down for Ank contained Wnt-5a, which activated Tcf/Lef reporter plasmids and promoted translocation of beta-catenin into the nucleus without activating the c-Jun N-terminal kinase (JNK) pathway. Supplementation with PP(i) compensated for most effects of Ank deficiency on Type II collagen, Sox-9, and Wnt-5 expression, both in IL-1beta and Ank knock-down conditions. Phenotype changes induced by IL-1beta were also supported by activation of the JNK pathway, but this latter was not sensitive to PP(i) supplementation. Altogether our data demonstrate that the transport of PP(i) by ANK contributed to the maintenance of the differentiated phenotype of chondrocyte by controlling the canonical Wnt pathway in a Wnt-5a-dependent manner.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Diphosphates/metabolism , Membrane Proteins/metabolism , Phenotype , Signal Transduction , Animals , Cartilage, Articular/cytology , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Collagen Type II/genetics , Collagen Type II/metabolism , Culture Media, Conditioned/pharmacology , Interleukin-1beta/pharmacology , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Phosphate Transport Proteins , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
9.
JCI Insight ; 6(24)2021 12 22.
Article in English | MEDLINE | ID: mdl-34727094

ABSTRACT

Osteoarthritis is the most prevalent joint disease worldwide, and it is a leading source of pain and disability. To date, this disease lacks curative treatment, as underlying molecular mechanisms remain largely unknown. The histone methyltransferase DOT1L protects against osteoarthritis, and DOT1L-mediated H3K79 methylation is reduced in human and mouse osteoarthritic joints. Thus, restoring DOT1L function seems to be critical to preserve joint health. However, DOT1L-regulating molecules and networks remain elusive, in the joint and beyond. Here, we identified transcription factors and networks that regulate DOT1L gene expression using a potentially novel bioinformatics pipeline. Thereby, we unraveled a possibly undiscovered link between the hypoxia pathway and DOT1L. We provide evidence that hypoxia enhanced DOT1L expression and H3K79 methylation via hypoxia-inducible factor-1 α (HIF1A). Importantly, we demonstrate that DOT1L contributed to the protective effects of hypoxia in articular cartilage and osteoarthritis. Intra-articular treatment with a selective hypoxia mimetic in mice after surgical induction of osteoarthritis restored DOT1L function and stalled disease progression. Collectively, our data unravel a molecular mechanism that protects against osteoarthritis with hypoxia inducing DOT1L transcription in cartilage. Local treatment with a selective hypoxia mimetic in the joint restores DOT1L function and could be an attractive therapeutic strategy for osteoarthritis.


Subject(s)
Cartilage, Articular/immunology , Cell Hypoxia/genetics , Histone-Lysine N-Methyltransferase/metabolism , Osteoarthritis/genetics , Animals , Humans , Mice
10.
FEBS J ; 286(24): 4832-4851, 2019 12.
Article in English | MEDLINE | ID: mdl-31677330

ABSTRACT

Osteo-articular diseases are characterized by a dysregulation of joint and/or bone homeostasis. These include diseases affecting the joints originally, such as osteoarthritis and rheumatoid arthritis, or the bone, such as osteoporosis. Inflammation and the involvement of Wingless-related integration site (Wnt) signaling pathways are key pathophysiological features of these diseases resulting in tissue degradation by matrix-degrading enzymes, namely matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinases with thrombospondin motifs (ADAMTs), secreted by the joint resident cells and/or by infiltrating immune cells. Activation of Wnt signaling pathways is modulated by different families of proteins, including Dickkopfs and the secreted Frizzled-related proteins (sFRPs). The sFRP family is composed of five secreted glycoproteins in mammals that regulate Wnt signaling in the extracellular compartment. Indeed, sFRPs are able to bind both to the soluble Wnt ligands and to their cell membrane receptors, the Frizzled proteins. Their expression profile is altered in osteo-articular diseases, suggesting that they could account for the abnormal activation of Wnt pathways. In the present article, we review how sFRPs are more than simple antagonists of the Wnt signaling pathways and discuss their pathophysiological relevance in the context of osteo-articular diseases. We detail their Wnt-dependent and their Wnt-independent roles, with a particular emphasis on their ability to modulate the inflammatory response and extracellular matrix (ECM) remodeling. We also discuss their potential therapeutic use with a focus on bone remodeling, osteo-articular cancers, and tissue engineering.


Subject(s)
Bone Diseases/metabolism , Osteoarthritis/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Animals , Bone Diseases/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Musculoskeletal Diseases/genetics , Musculoskeletal Diseases/metabolism , Osteoarthritis/genetics , Protein Transport/genetics , Protein Transport/physiology , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
12.
PLoS One ; 13(6): e0198104, 2018.
Article in English | MEDLINE | ID: mdl-29897942

ABSTRACT

Tissue calcification is an important physiological process required for the normal structure and function of bone. However, ectopic or excessive calcification contributes to diseases such as chondrocalcinosis, to calcium deposits in the skin or to vascular calcification. SMOC2 is a member of the BM-40/osteonectin family of calcium-binding secreted matricellular proteins. Using osteoprogenitor MC3T3-E1 cells stably overexpressing SMOC2, we show that SMOC2 inhibits osteogenic differentiation and extracellular matrix mineralization. Stable Smoc2 knockdown in these cells had no effect on mineralization suggesting that endogenous SMOC2 is not essential for the mineralization process. Mineralization in MC3T3-E1 cells overexpressing mutant SMOC2 lacking the extracellular calcium-binding domain was significantly increased compared to cells overexpressing full length SMOC2. When SMOC2 overexpressing cells were cultured in the presence of extracellular calcium supplementation, SMOC2's inhibitory effect on calcification was rescued. Our observations were translationally validated in primary human periosteal-derived cells. Furthermore, SMOC2 was able to impair mineralization in transdifferentiated human umbilical vein endothelial cells. Taken together, our data indicate that SMOC2 can act as an inhibitor of mineralization. We propose a possible role for SMOC2 to prevent calcification disorders.


Subject(s)
Calcification, Physiologic/genetics , Calcium-Binding Proteins/physiology , Cell Differentiation/genetics , Endothelial Cells/physiology , Osteoblasts/physiology , Animals , Calcium-Binding Proteins/genetics , Cells, Cultured , Human Umbilical Vein Endothelial Cells/physiology , Humans , Mice , Osteogenesis/genetics
13.
RMD Open ; 3(2): e000604, 2017.
Article in English | MEDLINE | ID: mdl-29299344

ABSTRACT

OBJECTIVES: Suramin is an old drug used for the treatment of African sleeping sickness. We investigated therapeutic repositioning of suramin to protect against cartilage damage, as suramin may interact with tissue inhibitor of metalloproteinase-3 (TIMP3). METHODS: In vitro extracellular matrix (ECM) accumulation and turnover in the presence or absence of suramin were studied in the ATDC5 micromass model of chondrogenesis and in pellet cultures of human articular chondrocytes from osteoarthritis and control patients, by gene expression, protein analysis, colorimetric staining, immunoprecipitation, fluorimetric analysis and immunohistochemistry. To study suramin in vivo, the drug was injected intra-articularly in the papain model of joint damage. Disease severity was analysed by histology, immunohistochemistry and contrast-enhanced nanofocus CT. RESULTS: In ATDC5 micromasses, suramin increased TIMP3 levels and decreased the activity of matrix metalloproteinases (MMPs) and aggrecanases. Suramin treatment resulted in increased glycosaminoglycans. This effect on the ECM was blocked by an anti-TIMP3 antibody. Direct interaction between suramin and endogenous TIMP3 was demonstrated in immunoprecipitates. Mice treated intra-articularly with suramin injections showed reduced cartilage damage compared with controls, with increased TIMP3 and decreased MMP and aggrecanase activity. Translational validation in human chondrocytes confirmed increased TIMP3 function and reduced cartilage breakdown after suramin treatment. CONCLUSION: Suramin prevented loss of articular cartilage in a mouse model of cartilage damage. The effects appear to be mediated by a functional increase of TIMP3 and a subsequent decrease in the activity of catabolic enzymes. Thus, suramin repositioning could be considered to prevent progressive cartilage damage and avoid evolution toward osteoarthritis.

14.
Nat Commun ; 8: 15889, 2017 06 19.
Article in English | MEDLINE | ID: mdl-28627522

ABSTRACT

Osteoarthritis is the most prevalent and crippling joint disease, and lacks curative treatment, as the underlying molecular basis is unclear. Here, we show that DOT1L, an enzyme involved in histone methylation, is a master protector of cartilage health. Loss of DOT1L disrupts the molecular signature of healthy chondrocytes in vitro and causes osteoarthritis in mice. Mechanistically, the protective function of DOT1L is attributable to inhibition of Wnt signalling, a pathway that when hyper-activated can lead to joint disease. Unexpectedly, DOT1L suppresses Wnt signalling by inhibiting the activity of sirtuin-1 (SIRT1), an important regulator of gene transcription. Inhibition of SIRT1 protects against osteoarthritis triggered by loss of DOT1L activity. Modulating the DOT1L network might therefore be a therapeutic approach to protect the cartilage against osteoarthritis.


Subject(s)
Cartilage/metabolism , Methyltransferases/metabolism , Osteoarthritis/pathology , Animals , Benzimidazoles/pharmacology , Benzimidazoles/toxicity , Cartilage/pathology , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Female , Gene Expression Regulation , Histone-Lysine N-Methyltransferase , Homeostasis , Male , Methylation , Methyltransferases/genetics , Mice, Inbred C57BL , Mice, Knockout , Osteoarthritis/chemically induced , Osteoarthritis/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Wnt Signaling Pathway
15.
Am J Clin Nutr ; 82(2): 467-70, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16087994

ABSTRACT

BACKGROUND: Serotonin (5-hydroxytryptamine; 5-HT) is a key mediator in the control of food intake and is probably involved in the etiology of anorexia nervosa. An association between a polymorphism of the 5-HT receptor (5-HT2A) gene promoter (-1438G/A) and anorexia nervosa has been reported. OBJECTIVE: We investigated the relation between the -1438G/A polymorphism of the 5-HT(2A) gene and the energy and macronutrient intakes of children and adolescents. DESIGN: This cross-sectional study included 370 children and adolescents aged 10-20 y (176 boys and 194 girls from 251 families) drawn from the Stanislas Family Study. Energy and macronutrient intakes were assessed by using 3-d food records. The -1438G/A polymorphism was analyzed by polymerase chain reaction and then by Hpa II digestion. RESULTS: In the overall group, after adjustment for age, sex, weight, height, and family correlation, the A allele was significantly associated with lower energy (P for trend = 0.045) and with total, monounsaturated, and saturated fat intakes expressed in g/d (P for trend = 0.007, 0.005, and 0.006, respectively). Subjects with the GA genotype had intermediate values. In addition, genotype x sex and genotype x age interactions were not significant. CONCLUSIONS: The 5-HT2A gene polymorphism in the promoter region is associated with energy and fat intakes in young people. This could be explained by the role of the serotonergic system as a determinant of food intakes and eating behavior.


Subject(s)
Eating , Polymorphism, Genetic , Receptor, Serotonin, 5-HT2A/genetics , Adolescent , Adult , Child , Cross-Sectional Studies , Female , Humans , Male , Promoter Regions, Genetic
17.
Arthritis Res Ther ; 14(1): R16, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22264237

ABSTRACT

INTRODUCTION: The aim of this research was to study molecular changes in the articular cartilage and subchondral bone of the tibial plateau from mice deficient in frizzled-related protein (Frzb) compared to wild-type mice by transcriptome analysis. METHODS: Gene-expression analysis of the articular cartilage and subchondral bone of three wild-type and three Frzb-/- mice was performed by microarray. Data from three wild-type and two Frzb-/- samples could be used for pathway analysis of differentially expressed genes and were explored with PANTHER, DAVID and GSEA bioinformatics tools. Activation of the wingless-type (WNT) pathway was analysed using Western blot. The effects of Frzb gain and loss of function on chondrogenesis and cell proliferation was examined using ATDC5 micro-masses and mouse ribcage chondrocytes. RESULTS: Extracellular matrix-associated integrin and cadherin pathways, as well as WNT pathway genes were up-regulated in Frzb-/- samples. Several WNT receptors, target genes and other antagonists were up-regulated, but no difference in active ß-catenin was found. Analysis of ATDC5 cell micro-masses overexpressing FRZB indicated an up-regulation of aggrecan and Col2a1, and down-regulation of molecules related to damage and repair in cartilage, Col3a1 and Col5a1. Silencing of Frzb resulted in down-regulation of aggrecan and Col2a1. Pathways associated with cell cycle were down-regulated in this transcriptome analysis. Ribcage chondrocytes derived from Frzb-/- mice showed decreased proliferation compared to wild-type cells. CONCLUSIONS: Our analysis provides evidence for tight regulation of WNT signalling, shifts in extracellular matrix components and effects on cell proliferation and differentiation in the articular cartilage - subchondral bone unit in Frzb-/- mice. These data further support an important role for FRZB in joint homeostasis and highlight the complex biology of WNT signaling in the joint.


Subject(s)
Bone and Bones/metabolism , Cartilage, Articular/metabolism , Gene Expression Profiling , Glycoproteins/genetics , Wnt Signaling Pathway/genetics , Animals , Blotting, Western , Cell Cycle/genetics , Cell Line , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/genetics , Female , Gene Expression Profiling/statistics & numerical data , Glycoproteins/metabolism , Intracellular Signaling Peptides and Proteins , Knee Joint/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
18.
Biomed Mater Eng ; 20(3): 209-15, 2010.
Article in English | MEDLINE | ID: mdl-20930330

ABSTRACT

The balance between extracellular inorganic phosphate (ePi) and extracellular inorganic pyrophosphate (ePPi) is controlled by four membrane proteins: the transporters ANK (exporting PPi outside the cells) and PiT-1 (importing ePi into the cells), and the enzymes PC-1 (generating ePPi from nucleotides) and Tissue Non-specific Alkaline Phosphatase (TNAP, hydrolyzing ePPi into ePi). TGF-ß1 was shown to stimulate ANK and PC-1 expression in articular chondrocytes, and subsequent ePPi level, as well as to increase ePi uptake by inducing PiT-1 expression in a chondrogenic cell line. Thus, we investigated the ability of ePi to modulate the effect of TGF-ß1 on the regulatory proteins of the ePi/ePPi balance in chondrocytes. In the pathophysiological range of 0.01-1 mM, ePi was inactive by itself but potentiated the stimulatory effects of TGF-ß1 on ANK, PC-1 or PiT-1 mRNA (RT-qPCR) and protein (Western blot) levels. PC-1 activity was also increased by TGF-ß1 and further potentiated by ePi supplementation. TNAP mRNA and activity became undetectable in response to TGF-ß1. These data suggest that ePi could increase ePPi level by changing the control of ANK and PC-1 expression by TGF-ß1, further highlighting an adaptative regulation of the Pi/PPi balance to prevent basic calcium phosphate deposition into the joints.


Subject(s)
Chondrocytes/metabolism , Diphosphates/metabolism , Gene Expression Regulation/physiology , Phosphates/metabolism , Transforming Growth Factor beta1/pharmacology , Animals , Cells, Cultured , Chondrocytes/drug effects , Gene Expression Regulation/drug effects , Male , Rats , Rats, Wistar
19.
Atherosclerosis ; 200(2): 286-93, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18280482

ABSTRACT

C-reactive protein (CRP) is an independent predictor of atherosclerosis and its complications. Monocytes/macrophages are implicated in this complex disease which is, among other mechanisms, characterised by angiogenesis. The aim of this study was to analyse whether CRP plays a role in VEGF-A regulation by monocytic cells. Our findings show that CRP up-regulates VEGF-A mRNA expression and protein excretion in THP-1 cells in a concentration- and time-dependent manner. Furthermore, we studied the signaling pathway underlying this effect. CRP increases VEGF-A expression via a PI3-kinase and an extracellular-signal-regulated kinase (ERK) 1/2 dependent pathway. Our results suggest that CRP could play a role in the angiogenesis process via immune cells such as monocytes.


Subject(s)
C-Reactive Protein/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Enzyme Inhibitors/pharmacology , Humans , MAP Kinase Signaling System , Neovascularization, Pathologic , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Arthritis Res Ther ; 9(6): R122, 2007.
Article in English | MEDLINE | ID: mdl-18034874

ABSTRACT

ANK is a multipass transmembrane protein transporter thought to play a role in the export of intracellular inorganic pyrophosphate and so to contribute to the pathophysiology of chondrocalcinosis. As transforming growth factor-beta-1 (TGF-beta1) was shown to favor calcium pyrophosphate dihydrate deposition, we investigated the contribution of ANK to the production of extracellular inorganic pyrophosphate (ePPi) by chondrocytes and the signaling pathways involved in the regulation of Ank expression by TGF-beta1. Chondrocytes were exposed to 10 ng/mL of TGF-beta1, and Ank expression was measured by quantitative polymerase chain reaction and Western blot. ePPi was quantified in cell supernatants. RNA silencing was used to define the respective roles of Ank and PC-1 in TGF-beta1-induced ePPi generation. Finally, selective kinase inhibitors and dominant-negative/overexpression plasmid strategies were used to explore the contribution of several signaling pathways to Ank induction by TGF-beta1. TGF-beta1 strongly increased Ank expression at the mRNA and protein levels, as well as ePPi production. Using small interfering RNA technology, we showed that Ank contributed approximately 60% and PC-1 nearly 20% to TGF-beta1-induced ePPi generation. Induction of Ank by TGF-beta1 required activation of the extracellular signal-regulated kinase (ERK) pathway but not of p38-mitogen-activated protein kinase or of protein kinase A. In line with the general protein kinase C (PKC) inhibitor calphostin C, Gö6976 (a Ca2+-dependent PKC inhibitor) diminished TGF-beta1-induced Ank expression by 60%, whereas a 10% inhibition was observed with rottlerin (a PKCdelta inhibitor). These data suggest a regulatory role for calcium in TGF-beta1-induced Ank expression. Finally, we demonstrated that the stimulatory effect of TGF-beta1 on Ank expression was inhibited by the suppression of the Ras/Raf-1 pathway, while being enhanced by their constitutive activation. Transient overexpression of Smad 7, an inhibitory Smad, failed to affect the inducing effect of TGF-beta1 on Ank mRNA level. These data show that TGF-beta1 increases ePPi levels, mainly by the induction of the Ank gene, which requires activation of Ras, Raf-1, ERK, and Ca2+-dependent PKC pathways in chondrocytes.


Subject(s)
Chondrocytes/drug effects , Chondrocytes/metabolism , Diphosphates/metabolism , Membrane Proteins/biosynthesis , Transforming Growth Factor beta1/pharmacology , Animals , Base Sequence , Cells, Cultured , Chondrocalcinosis/etiology , Chondrocalcinosis/genetics , Chondrocalcinosis/metabolism , DNA Primers/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Kinetics , MAP Kinase Signaling System/drug effects , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Phosphate Transport Proteins , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Wistar , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL