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










Publication year range
1.
Int J Mol Sci ; 22(21)2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34769441

ABSTRACT

Notch signaling has been identified as a critical regulator of cartilage development and homeostasis. Its pivotal role was established by both several joint specific Notch signaling loss of function mouse models and transient or sustained overexpression. NOTCH1 is the most abundantly expressed NOTCH receptors in normal cartilage and its expression increases in osteoarthritis (OA), when chondrocytes exit from their healthy "maturation arrested state" and resume their natural route of proliferation, hypertrophy, and terminal differentiation. The latter are hallmarks of OA that are easily evaluated in vitro in 2-D or 3-D culture models. The aim of our study was to investigate the effect of NOTCH1 knockdown on proliferation (cell count and Picogreen mediated DNA quantification), cell cycle (flow cytometry), hypertrophy (gene and protein expression of key markers such as RUNX2 and MMP-13), and terminal differentiation (viability measured in 3-D cultures by luminescence assay) of human OA chondrocytes. NOTCH1 silencing of OA chondrocytes yielded a healthier phenotype in both 2-D (reduced proliferation) and 3-D with evidence of decreased hypertrophy (reduced expression of RUNX2 and MMP-13) and terminal differentiation (increased viability). This demonstrates that NOTCH1 is a convenient therapeutic target to attenuate OA progression.


Subject(s)
Chondrocytes/pathology , Core Binding Factor Alpha 1 Subunit/metabolism , Hypertrophy/pathology , Matrix Metalloproteinase 13/metabolism , Osteoarthritis/pathology , Receptor, Notch1/metabolism , Aged , Cell Culture Techniques, Three Dimensional , Cells, Cultured , Chondrocytes/metabolism , Female , Humans , Hypertrophy/etiology , Hypertrophy/metabolism , Male , Osteoarthritis/etiology , Osteoarthritis/metabolism , Signal Transduction
2.
Sci Rep ; 11(1): 21697, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34737366

ABSTRACT

IKKα and IKKß are essential kinases for activating NF-κB transcription factors that regulate cellular differentiation and inflammation. By virtue of their small size, chemokines support the crosstalk between cartilage and other joint compartments and contribute to immune cell chemotaxis in osteoarthritis (OA). Here we employed shRNA retroviruses to stably and efficiently ablate the expression of each IKK in primary OA chondrocytes to determine their individual contributions for monocyte chemotaxis in response to chondrocyte conditioned media. Both IKKα and IKKß KDs blunted both the monocyte chemotactic potential and the protein levels of CCL2/MCP-1, the chemokine with the highest concentration and the strongest association with monocyte chemotaxis. These findings were mirrored by gene expression analysis indicating that the lowest levels of CCL2/MCP-1 and other monocyte-active chemokines were in IKKαKD cells under both basal and IL-1ß stimulated conditions. We find that in their response to IL-1ß stimulation IKKαKD primary OA chondrocytes have reduced levels of phosphorylated NFkappaB p65pSer536 and H3pSer10. Confocal microscopy analysis revealed co-localized p65 and H3pSer10 nuclear signals in agreement with our findings that IKKαKD effectively blunts their basal level and IL-1ß dependent increases. Our results suggest that IKKα could be a novel OA disease target.


Subject(s)
I-kappa B Kinase/metabolism , Interleukin-1beta/metabolism , Monocytes/metabolism , Cells, Cultured , Chemokine CCL2/metabolism , Chemokines/immunology , Chemokines/metabolism , Chemotaxis/physiology , Chondrocytes/metabolism , Female , Humans , I-kappa B Kinase/physiology , Inflammation , Interleukin-1beta/physiology , Male , Middle Aged , NF-kappa B/metabolism , Osteoarthritis/metabolism , Phosphorylation , Protein Serine-Threonine Kinases , Signal Transduction/physiology , Transcription Factor RelA
3.
Free Radic Biol Med ; 166: 212-225, 2021 04.
Article in English | MEDLINE | ID: mdl-33636333

ABSTRACT

During osteoarthritis development, chondrocytes are subjected to a functional derangement. This increases their susceptibility to stressful conditions such as oxidative stress, a characteristic of the aging tissue, which can further provoke extrinsic senescence by DNA damage responses. It was previously observed that IκB kinase α knockdown increases the replicative potential of primary human OA chondrocytes cultured in monolayer and the survival of the same cells undergoing hypertrophic-like differentiation in 3-D. In this paper we investigated whether IKKα knockdown could modulate oxidative stress-induced senescence of OA chondrocytes undergoing a DDR and particularly the involvement in this process of the DNA mismatch repair system, the principal mechanism for repair of replicative and recombinational errors, devoted to genomic stability maintenance in actively replicating cells. This repair system is also implicated in oxidative stress-mediated DNA damage repair. We analyzed microsatellite instability and expression of the mismatch repair components in human osteoarthritis chondrocytes after IKKα knockdown and H2O2 exposure. Only low MSI levels and incidence were detected and exclusively in IKKα proficient cells. Moreover, we found that IKKα proficient and deficient chondrocytes differently regulated MMR proteins after oxidative stress, both at mRNA and protein level, suggesting a reduced susceptibility of IKKα deficient cells. Our data suggest an involvement of the MMR system in the response to oxidative stress that tends to be more efficient in IKKαKD cells. This argues for a partial contribution of the MMR system to the better ability to recover DNA damage already observed in these cells.


Subject(s)
Chondrocytes , Osteoarthritis , Chondrocytes/metabolism , DNA Damage , DNA Mismatch Repair/genetics , DNA Repair/genetics , Humans , Hydrogen Peroxide/pharmacology , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Osteoarthritis/genetics , Oxidative Stress/genetics
4.
Free Radic Biol Med ; 153: 159-172, 2020 06.
Article in English | MEDLINE | ID: mdl-32305648

ABSTRACT

Oxidative stress (OS) contributes to Osteoarthritis (OA) pathogenesis and its effects are worsened by the impairment of homeostatic mechanisms such as autophagy in OA chondrocytes. Rescue of an efficient autophagic flux could therefore reduce the bulk of damaged molecules, and at the same time improve cell function and viability. As a promising dietary or intra-articular supplement to rescue autophagy in OA chondrocytes, we tested spermidine (SPD), known to induce autophagy and to reduce OS in several other cellular models. Chondrocytes were obtained from OA cartilage and seeded at high-density to keep their differentiated phenotype. The damaging effects of OS and the chondroprotective activity of SPD were assessed by evaluating the extent of cell death, oxidative DNA damage and caspase 3 activation. The autophagy promoting activity of SPD was evaluated by assessing pivotal autophagic effectors, i.e. Beclin-1 (BECN-1), microtubule-associated protein 1 light chain 3 II (LC3-II) and p62. BECN-1 protein expression was significantly increased by SPD and reduced by H2O2 treatment. SPD also rescued the impaired autophagic flux consequent to H2O2 exposure by increasing mRNA and protein expression of LC3-II and p62. SPD induction of mitophagy was revealed by immunofluorescent co-localization of LC3-II and TOM20. The key protective role of autophagy was confirmed by the loss of SPD chondroprotection upon autophagy-related gene 5 (ATG5) silencing. Significant SPD tuning of the H2O2-dependent induction of degradative (MMP-13), inflammatory (iNOS, COX-2) and hypertrophy markers (RUNX2 and VEGF) was revealed by Real Time PCR and pointed at the SPD ability of reducing NF-κB activation through autophagy induction. Conversely, blockage of autophagy led to parallel increases of oxidative markers and p65 nuclear translocation. SPD also increased the proliferation of slow-proliferating primary cultures. Taken together, our findings highlight the chondroprotective, anti-oxidant and anti-inflammatory activity of SPD and suggest that the protection afforded by SPD against OS is exerted through the rescue of the autophagic flux.


Subject(s)
Chondrocytes , Spermidine , Autophagy , Hydrogen Peroxide/toxicity , Oxidative Stress , Spermidine/pharmacology
5.
Sci Rep ; 9(1): 14269, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31582764

ABSTRACT

According to previous research, natural polyamines exert a role in regulating cell committment and differentiation from stemness during skeletal development. In order to assess whether distinct polyamine patterns are associated with different skeletal cell types, primary cultures of stem cells, chondrocytes or osteoblasts were dedicated for HPLC analysis of intracellular polyamines. Spermine (SPM) and Spermidine (SPD) levels were higher in adipose derived stem cells (ASC) compared to mature skeletal cells, i.e. chondrocytes and osteoblasts, confirming the connection of polyamine content with stemness. To establish whether polyamines can protect ASC against oxidative DNA damage in a 3-D differentiation model, the level of γH2AX was measured by western blot, and found to correlate with age and BMI of patients. Addition of either polyamine to ASC was able to hinder DNA damage in the low micromolecular range, with marked reduction of γH2AX level at 10 µM SPM and 5 µM SPD. Molecular analysis of the mechanisms that might underlie the protective effect of polyamine supplementation evidences a possible involvement of autophagy. Altogether, these results support the idea that polyamines are able to manage both stem cell differentiation and cell oxidative damage, and therefore represent appealing tools for regenerative and cell based applications.


Subject(s)
DNA Damage , Mesenchymal Stem Cells/metabolism , Spermidine/metabolism , Spermine/metabolism , Adult , Aged , Cells, Cultured , DNA Damage/drug effects , Histones/analysis , Histones/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Middle Aged , Spermidine/pharmacology , Spermine/pharmacology
6.
Sci Rep ; 9(1): 13603, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31537813

ABSTRACT

Osteoarthritis (OA), the most prevalent degenerative joint disease, still lacks a true disease-modifying therapy. The involvement of the NF-κB pathway and its upstream activating kinases in OA pathogenesis has been recognized for many years. The ability of the N-acetyl phenylalanine glucosamine derivative (NAPA) to increase anabolism and reduce catabolism via inhibition of IKKα kinase has been previously observed in vitro and in vivo. The present study aims to confirm the chondroprotective effects of NAPA in an in vitro model of joint OA established with primary cells, respecting both the crosstalk between chondrocytes and synoviocytes and their phenotypes. This model satisfactorily reproduces some features of the previously investigated DMM model, such as the prominent induction of ADAMTS-5 upon inflammatory stimulation. Both gene and protein expression analysis indicated the ability of NAPA to counteract key cartilage catabolic enzymes (ADAMTS-5) and effectors (MCP-1). Molecular analysis showed the ability of NAPA to reduce IKKα nuclear translocation and H3Ser10 phosphorylation, thus inhibiting IKKα transactivation of NF-κB signalling, a pivotal step in the NF-κB-dependent gene expression of some of its targets. In conclusion, our data confirm that NAPA could truly act as a disease-modifying drug in OA.


Subject(s)
Chondrocytes/cytology , Glucosamine/pharmacology , I-kappa B Kinase/genetics , Osteoarthritis/immunology , Synoviocytes/cytology , ADAMTS5 Protein/genetics , ADAMTS5 Protein/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chondrocytes/drug effects , Chondrocytes/immunology , Coculture Techniques , Gene Expression Regulation/drug effects , Glucosamine/chemistry , Humans , I-kappa B Kinase/metabolism , Models, Biological , Osteoarthritis/drug therapy , Osteoarthritis/genetics , Phosphorylation/drug effects , Synoviocytes/drug effects , Synoviocytes/immunology
8.
Oxid Med Cell Longev ; 2018: 3075293, 2018.
Article in English | MEDLINE | ID: mdl-29599894

ABSTRACT

The prevalence of Osteoarthritis (OA) is increasing because of the progressive aging and unhealthy lifestyle. These risk factors trigger OA by removing constraints that keep the tightly regulated low turnover of the extracellular matrix (ECM) of articular cartilage, the correct chondrocyte phenotype, and the functionality of major homeostatic mechanisms, such as mitophagy, that allows for the clearance of dysfunctional mitochondria, preventing increased production of reactive oxygen species, oxidative stress, and senescence. After OA onset, the presence of ECM degradation products is perceived as a "danger" signal by the chondrocytes and the synovial macrophages that release alarmins with autocrine/paracrine effects on the same cells. Alarmins trigger innate immunity in the joint, with important systemic crosstalks that explain the beneficial effects of dietary interventions and improved lifestyle. Alarmins also boost low-grade inflammation: the release of inflammatory molecules and chemokines sustained by continuous triggering of NF-κB within an altered cellular setting that allows its higher transcriptional activity. Chemokines exert pleiotropic functions in OA, including the recruitment of inflammatory cells and the induction of ECM remodeling. Some chemokines have been successfully targeted to attenuate structural damage or pain in OA animal models. This represents a promising strategy for the future management of human OA.


Subject(s)
Cellular Senescence/physiology , Chondrocytes/metabolism , Inflammation/physiopathology , Osteoarthritis/physiopathology , Humans , Oxidative Stress
9.
Sci Rep ; 7(1): 7458, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28785079

ABSTRACT

Patients with long-standing ulcerative colitis (UC) have an increased colorectal cancer (CRC) risk. In this pilot study we evaluated the effect of Eicosapentaenoic acid as free fatty acid (EPA-FFA) supplementation on mucosal disease activity, colonic differentiation markers and microbiota composition in UC patients. Twenty long-standing UC patients in stable clinical remission and with fecal calprotectin (FC) > 150 µg/g were enrolled (T0) and supplemented with EPA-FFA 2 g/daily for 90 days (T3). Endoscopic and histologic disease activities were measured by Mayo and Geboes scores, respectively. HES1, KLF4, STAT3, IL-10 and SOCS3 levels were determined using western blotting and qRT-PCR, while phospho-STAT3 levels were assessed by western blotting. Goblet cells were stained by Alcian blue. Microbiota analyses were performed on both fecal and colonic samples. Nineteen patients completed the study; seventeen (89.5%) were compliant. EPA-FFA treatment reduced FC levels at T3. Patients with FC > 150 µg/g at T3 (n = 2) were assumed as non-responders. EPA-FFA improved endoscopic and histological inflammation and induced IL-10, SOCS3, HES1 and KLF4 in compliant and responder patients. Importantly, long-term UC-driven microbiota composition was partially redressed by EPA-FFA. In conclusion, EPA-FFA supplementation reduced mucosal inflammation, promoted goblet cells differentiation and modulated intestinal microbiota composition in long-standing UC patients.


Subject(s)
Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Eicosapentaenoic Acid/administration & dosage , Fatty Acids, Nonesterified/administration & dosage , Microbiota/drug effects , Adult , Aged , Aged, 80 and over , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Nonesterified/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukocyte L1 Antigen Complex/metabolism , Male , Middle Aged , Phosphorylation/drug effects , Pilot Projects , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Treatment Outcome , Young Adult
10.
Oxid Med Cell Longev ; 2017: 3720128, 2017.
Article in English | MEDLINE | ID: mdl-28713485

ABSTRACT

Osteoarthritis (OA) is a debilitating degenerative disease of the articular cartilage with a multifactorial etiology. Aging, the main risk factor for OA development, is associated with a systemic oxidative and inflammatory phenotype. Autophagy is a central housekeeping system that plays an antiaging role by supporting the clearance of senescence-associated alterations of macromolecules and organelles. Autophagy deficiency has been related to OA pathogenesis because of the accumulation of cellular defects in chondrocytes. Microribonucleic acids (microRNAs or miRs) are a well-established class of posttranscriptional modulators belonging to the family of noncoding RNAs that have been identified as key players in the regulation of cellular processes, such as autophagy, by targeting their own cognate mRNAs. Here, we present a state-of-the-art literature review on the role of miRs and autophagy in the scenario of OA pathogenesis. In addition, a comprehensive survey has been performed on the functional connections of the miR network and the autophagy pathway in OA by using "microRNA," "autophagy," and "osteoarthritis" as key words. Discussion of available evidence sheds light on some aspects that need further investigation in order to reach a more comprehensive view of the potential of this topic in OA.


Subject(s)
Autophagy/physiology , Cellular Senescence/physiology , Chondrocytes/metabolism , Homeostasis/physiology , Osteoarthritis/metabolism , Animals , Humans , Mice , MicroRNAs/metabolism
11.
Oncotarget ; 7(26): 39609-39626, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27167202

ABSTRACT

Hepatocellular carcinoma (HCC) ranks fifth in frequency worldwide amongst all human cancers causing one million deaths annually. Despite many promising treatment options, long-term prognosis remains dismal for the majority of patients who develop recurrence or present with advanced disease. Notch signaling is an evolutionarily conserved pathway crucial for the development and homeostasis of many organs including liver. Herein we showed that aberrant Notch1 is linked to HCC development, tumor recurrence and invasion, which might be mediated, at least in part, through the Notch1-E-Cadherin pathway. Collectively, these findings suggest that targeting Notch1 has important therapeutic value in hepatocellular carcinoma. In this regard, comparative analysis of the secretome of HepG2 and HepG2 Notch1 depleted cells identified novel secreted proteins related to Notch1 expression. Soluble E-Cadherin (sE-Cad) and Thrombospondin-1 (Thbs1) were further validated in human serum as potential biomarkers to predict response to Notch1 inhibitors for a tailored individualized therapy.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Proteomics/methods , Receptor, Notch1/metabolism , Aged , Aged, 80 and over , Animals , Antigens, CD , Biomarkers, Tumor , Cadherins/blood , Cell Line, Tumor , Culture Media, Conditioned/chemistry , Female , Genetic Markers/genetics , Hep G2 Cells , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Rats , Rats, Wistar , Signal Transduction , Thrombospondin 1/blood , Wound Healing
12.
PLoS One ; 10(11): e0143865, 2015.
Article in English | MEDLINE | ID: mdl-26618897

ABSTRACT

INTRODUCTION: Recent evidence suggests that GSK3 activity is chondroprotective in osteoarthritis (OA), but at the same time, its inactivation has been proposed as an anti-inflammatory therapeutic option. Here we evaluated the extent of GSK3ß inactivation in vivo in OA knee cartilage and the molecular events downstream GSK3ß inactivation in vitro to assess their contribution to cell senescence and hypertrophy. METHODS: In vivo level of phosphorylated GSK3ß was analyzed in cartilage and oxidative damage was assessed by 8-oxo-deoxyguanosine staining. The in vitro effects of GSK3ß inactivation (using either LiCl or SB216763) were evaluated on proliferating primary human chondrocytes by combined confocal microscopy analysis of Mitotracker staining and reactive oxygen species (ROS) production (2',7'-dichlorofluorescin diacetate staining). Downstream effects on DNA damage and senescence were investigated by western blot (γH2AX, GADD45ß and p21), flow cytometric analysis of cell cycle and light scattering properties, quantitative assessment of senescence associated ß galactosidase activity, and PAS staining. RESULTS: In vivo chondrocytes from obese OA patients showed higher levels of phosphorylated GSK3ß, oxidative damage and expression of GADD45ß and p21, in comparison with chondrocytes of nonobese OA patients. LiCl mediated GSK3ß inactivation in vitro resulted in increased mitochondrial ROS production, responsible for reduced cell proliferation, S phase transient arrest, and increase in cell senescence, size and granularity. Collectively, western blot data supported the occurrence of a DNA damage response leading to cellular senescence with increase in γH2AX, GADD45ß and p21. Moreover, LiCl boosted 8-oxo-dG staining, expression of IKKα and MMP-10. CONCLUSIONS: In articular chondrocytes, GSK3ß activity is required for the maintenance of proliferative potential and phenotype. Conversely, GSK3ß inactivation, although preserving chondrocyte survival, results in functional impairment via induction of hypertrophy and senescence. Indeed, GSK3ß inactivation is responsible for ROS production, triggering oxidative stress and DNA damage response.


Subject(s)
Chondrocytes/drug effects , Glycogen Synthase Kinase 3/metabolism , Lithium Chloride/pharmacology , Obesity/pathology , Osteoarthritis, Knee/pathology , Cell Proliferation/drug effects , Cell Size/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Chondrocytes/cytology , Chondrocytes/pathology , DNA Damage , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Indoles/pharmacology , Maleimides/pharmacology , Obesity/enzymology , Osteoarthritis, Knee/enzymology , Oxidative Stress/drug effects , Phosphorylation/drug effects
13.
Oncotarget ; 5(21): 10607-20, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25431954

ABSTRACT

To successfully target Notch receptors as part of a multidrug anticancer strategy, it will be essential to fully characterize the factors that are modulated by Notch signaling. We recently reported that Notch3 silencing in HCC results in p53 up-regulation in vitro and, therefore, we focused on the mechanisms that associate Notch3 to p53 protein expression. We explored the regulation of p53 by Notch3 signalling in three HCC cell lines HepG2, SNU398 and Hep3B.We found that Notch3 regulates p53 at post-transcriptional level controlling both Cyclin G1 expression and the feed-forward circuit involving p53, miR-221 and MDM2. Moreover, our results were validated in human HCCs and in a rat model of HCC treated with Notch3 siRNAs. Our findings are becoming an exciting area for further in-depth research toward targeted inactivation of Notch3 receptor as a novel therapeutic approach for increasing the drug-sensitivity, and thereby improving the treatment outcome of patients affected by HCC. Indeed, we proved that Notch3 silencing strongly increases the effects of Nutilin-3.With regard to therapeutic implications, Notch3-specific drugs could represent a valuable strategy to limit Notch signaling in the context of hepatocellular carcinoma over-expressing this receptor.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cyclin G1/metabolism , Liver Neoplasms/metabolism , MicroRNAs/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, Notch/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Apoptosis , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Movement , Cell Proliferation , Cyclin G1/antagonists & inhibitors , Cyclin G1/genetics , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Liver Neoplasms/genetics , Male , Mutation/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptor, Notch3 , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
14.
PLoS One ; 9(10): e109724, 2014.
Article in English | MEDLINE | ID: mdl-25279550

ABSTRACT

Hydroxytyrosol (HT), a phenolic compound mainly derived from olives, has been proposed as a nutraceutical useful in prevention or treatment of degenerative diseases. In the present study we have evaluated the ability of HT to counteract the appearance of osteoarthritis (OA) features in human chondrocytes. Pre-treatment of monolayer cultures of chondrocytes with HT was effective in preventing accumulation of reactive oxidant species (ROS), DNA damage and cell death induced by H2O2 exposure, as well as the increase in the mRNA level of pro-inflammatory, matrix-degrading and hypertrophy marker genes, such as iNOS, COX-2, MMP-13, RUNX-2 and VEGF. HT alone slightly enhanced ROS production, but did not enhance cell damage and death or the expression of OA-related genes. Moreover HT was tested in an in vitro model of OA, i.e. three-dimensional micromass cultures of chondrocytes stimulated with growth-related oncogene α (GROα), a chemokine involved in OA pathogenesis and known to promote hypertrophy and terminal differentiation of chondrocytes. In micromass constructs, HT pre-treatment inhibited the increases in caspase activity and the level of the messengers for iNOS, COX-2, MMP-13, RUNX-2 and VEGF elicited by GROα. In addition, HT significantly increased the level of SIRT-1 mRNA in the presence of GROα. In conclusion, the present study shows that HT reduces oxidative stress and damage, exerts pro-survival and anti-apoptotic actions and favourably influences the expression of critical OA-related genes in human chondrocytes treated with stressors promoting OA-like features.


Subject(s)
Chemokine CXCL1/metabolism , Chondrocytes/drug effects , Hydrogen Peroxide/pharmacology , Inflammation Mediators/metabolism , Osteoarthritis/drug therapy , Phenylethyl Alcohol/analogs & derivatives , Aged , Aged, 80 and over , Antioxidants/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CXCL1/genetics , Chondrocytes/metabolism , Chondrocytes/pathology , Humans , Hypertrophy/drug therapy , Hypertrophy/metabolism , Hypertrophy/pathology , Middle Aged , Osteoarthritis/metabolism , Osteoarthritis/pathology , Oxidants/pharmacology , Oxidative Stress/drug effects , Phenylethyl Alcohol/pharmacology , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
15.
Amino Acids ; 46(3): 717-28, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24248311

ABSTRACT

The first step in skeleton development is the condensation of mesenchymal precursors followed by any of two different types of ossification, depending on the type of bone segment: in intramembranous ossification, the bone is deposed directly in the mesenchymal anlagen, whereas in endochondral ossification, the bone is deposed onto a template of cartilage that is subsequently substituted by bone. Polyamines and polyamine-related enzymes have been implicated in bone development as global regulators of the transcriptional and translational activity of stem cells and pivotal transcription factors. Therefore, it is tempting to investigate their use as a tool to improve regenerative medicine strategies in orthopedics. Growing evidence in vitro suggests a role for polyamines in enhancing differentiation in both adult stem cells and differentiated chondrocytes. Adipose-derived stem cells have recently proved to be a convenient alternative to bone marrow stromal cells, due to their easy accessibility and the high frequency of stem cell precursors per volume unit. State-of-the-art "prolotherapy" approaches for skeleton regeneration include the use of adipose-derived stem cells and platelet concentrates, such as platelet-rich plasma (PRP). Besides several growth factors, PRP also contains polyamines in the micromolar range, which may also exert an anti-apoptotic effect, thus helping to explain the efficacy of PRP in enhancing osteogenesis in vitro and in vivo. On the other hand, spermidine and spermine are both able to enhance hypertrophy and terminal differentiation of chondrocytes and therefore appear to be inducers of endochondral ossification. Finally, the peculiar activity of spermidine as an inducer of autophagy suggests the possibility of exploiting its use to enhance this cytoprotective mechanism to counteract the degenerative changes underlying either the aging or degenerative diseases that affect bone or cartilage.


Subject(s)
Drug Delivery Systems , Muscle, Skeletal/drug effects , Polyamines/pharmacology , Stem Cells/drug effects , Tissue Engineering , Animals , Cell Differentiation/drug effects , Humans , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Polyamines/metabolism , Stem Cells/metabolism
16.
Stem Cells Dev ; 22(10): 1588-601, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23301872

ABSTRACT

The molecular mechanisms underlying spermine osteo-inductive activity on human adipose-derived stem cells (ASCs) grown in 3-dimensional (3D) cultures were investigated. Spermine belongs to the polyamine family, naturally occurring, positively charged polycations that are able to control several cellular processes. Spermine was used at a concentration close to that found in platelet-rich plasma (PRP), an autologous blood product containing growth and differentiation factors, which has recently become popular in in vitro and in vivo bone healing and engineering. Adipose tissue was surgically obtained from the hypodermis of patients undergoing hip arthroplasty. Patient age negatively affected both ASC yield and ASC ability to form 3D constructs. ASC 3D cultures were seeded in either non differentiating or chondrogenic conditions, with or without the addition of 5 µM spermine to evaluate its osteogenic potential across 1, 2, and 3 weeks of maturation. Osteogenic medium was used as a reference. The effects of the addition of spermine on molecular markers of osteogenesis, at both gene and protein level, and mineralization were evaluated. The effects of spermine were temporally defined and responsible for the progression from the early to the mature osteoblast differentiation phases. Spermine initially promoted gene and protein expression of Runx-2, an early marker of the osteoblast lineage; then, it increased ß-catenin expression and activation, which led to the induction of Osterix gene expression, the mature osteoblast commitment factor. The finding that spermine induces ASC to differentiate toward mature osteoblasts supports the use of these easily accessible mesenchymal stem cells coupled with PRP for orthopedic applications.


Subject(s)
Adipose Tissue/cytology , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/drug effects , Spermine/pharmacology , Stem Cells/cytology , beta Catenin/metabolism , Adult , Aged , Apoptosis/drug effects , Cell Aggregation/drug effects , Cell Count , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chondrogenesis/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Flow Cytometry , Humans , Kinetics , Male , Middle Aged , Osteoblasts/drug effects , Platelet-Rich Plasma/metabolism , Protein Transport/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Transcription Factors/metabolism
17.
Am J Pathol ; 181(2): 413-22, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22705236

ABSTRACT

CDKN1C/P57 is a cyclin-dependent kinase inhibitor implicated in different human cancers, including hepatocellular carcinoma (HCC); however, little is known regarding the role of CDKN1C/P57 and its regulation in HCC. In this study, we show that the down-regulation of Notch1 and Notch3 in two HCC cell lines resulted in Hes1 down-regulation, CDKN1C/P57 up-regulation, and reduced cell growth. In line with these data, we report that CDKN1C/P57 is a target of transcriptional repression by the Notch effector, Hes1. We found that the up-regulation of CDKN1C/P57 by cDNA transfection decreased tumor growth, as determined by growth curve, flow cytometry analysis, and cyclin D1 down-regulation, without affecting the apoptosis machinery. Indeed, the expression of Bax, Noxa, PUMA, BNIP(3), and cleaved caspase-3 was not affected by CDKN1C/P57 induction. Morphologically CDKN1C/p57-induced HCC cells became flat and lengthened in shape, accumulated the senescence-associated ß-galactosidase marker, and increased P16 protein expression. Evaluation of senescence in cells depleted both for Hes1 and CDKN1C/P57 revealed that the senescent state really depends on the accumulation of CDKN1C/p57. Finally, we validated our in vitro results in primary HCCs, showing that Hes1 protein expression inversely correlates with CDKN1C/P57 mRNA levels. In addition, reduced Hes1 protein expression is accompanied by a shorter time to recurrence after curative resection, suggesting that Hes1 may represent a biomarker for prediction of patients with poor prognosis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , Homeodomain Proteins/genetics , Receptor, Notch1/metabolism , Receptors, Notch/metabolism , Aged , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Receptor, Notch3 , Recurrence , Signal Transduction , Time Factors , Transcription Factor HES-1
18.
Eur J Med Chem ; 48: 391-401, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22227043

ABSTRACT

We described the optimization, by molecular modelling, of small pyrazole derivatives, as kinase inhibitors, obtained through a 1,3-dipolar cycloaddition between nitrile imines and functionalized acetylenes. The two compounds, selected as potential agents active against hepatocellular carcinoma (HCC) were then evaluated in vitro for their biological activity on HCC-derived cell lines. The compounds show a promising inhibitory growth efficacy (IC(50) 50-100 µM) in SNU449 cell line, as well as block of cell cycle progression and induction of apoptosis, and can be considered as lead compounds for further SAR developments.


Subject(s)
Antineoplastic Agents/chemical synthesis , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Protein Kinase Inhibitors/chemical synthesis , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Blotting, Western , Carcinoma, Hepatocellular/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Dynamics Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Pyrazoles/chemistry , Spectrometry, Mass, Electrospray Ionization
19.
J Hepatol ; 50(5): 969-79, 2009 May.
Article in English | MEDLINE | ID: mdl-19304334

ABSTRACT

BACKGROUND/AIMS: The functional roles of endogenous Notch3 and Notch1 for protecting human hepatocellular carcinoma (HCC) lines against doxorubicin-induced death have been investigated. We previously reported aberrant Notch3 and Notch4 up-regulation in HCC and we have extended these observations to include Notch1. METHODS: Notch1 expression was assessed by immunohistochemistry and immunoblotting. Notch3 and Notch1 expression were ablated in multiple HCC lines by stable retroviral transduction of short hairpin RNAs (shRNAs). Effects on doxorubicin sensitivity were evaluated with respect to cell growth, expression of specific cell cycle effectors and multiple apoptotic parameters. RESULTS: Notch3 depletion increased p53 expression, doxorubicin uptake, DNA damage, the apoptosis inducing effects of doxorubicin and also impeded the cell cycle progression of HCC cells. Ablating p53 expression in Notch3 knockdown (KD) cells largely abolished their enhanced doxorubicin sensitivity; and Notch3 KD in p53(-/-) Hep3B cells failed to influence their response to doxorubicin. Although up-regulated in most HCC, Notch1 (unlike Notch3) did not contribute to the doxorubicin resistance of HCC lines. CONCLUSIONS: Our in vitro results represent the first evidence that Notch3 silencing in combination with chemotherapeutics could conceivably provide a novel strategy for HCC treatment that deserves further exploration.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Apoptosis/physiology , Carcinoma, Hepatocellular/drug therapy , Doxorubicin/therapeutic use , Liver Neoplasms/drug therapy , Receptors, Notch/metabolism , Tumor Suppressor Protein p53/metabolism , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/physiology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/physiology , Female , Gene Deletion , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Notch3 , Receptors, Notch/genetics , Sensitivity and Specificity , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...