Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
J Appl Microbiol ; 134(6)2023 06 01.
Article in English | MEDLINE | ID: mdl-37279902

ABSTRACT

AIMS: The aim of this study was to determine the biofilm-forming ability of a strain belonging to the Pseudomonas fluorescens group isolated from the dairy environment under food-relevant conditions. Moreover, the effects of commercial sanitizers against preformed biofilms were assessed both in terms of viability and structure. METHODS AND RESULTS: The biofilms were formed on polystyrene, stainless steel (SS), and polytetrafluoroethylene (PTFE) in a wide range of temperatures (4-25°C) and were subjected to the action of 10 different sanitizers. The strain under study showed to be a strong biofilm-former regardless of temperature, particularly on polystyrene. The biofilms were mostly sensitive to chlorine and peracetic acid-based sanitizers. For some sanitizers (e.g. amphoteric), a relationship was observed between the material and the tolerance, while the temperature was not statistically significant. The formation of long-term biofilms on SS was also structurally affected by the temperature, showing microcolonies more irregular in shape and with lower cellularity at 4°C compared to 15°C, where the biofilm was more compact and with a high presence of EPS. CONCLUSIONS: The strain belonging to the P. fluorescens group was shown to quickly adhere and form mature biofilm at temperatures and on materials relevant to the food sector; however, biofilms formed under different conditions were differently tolerant to disinfectants. SIGNIFICANCE AND IMPACT OF THE STUDY: Findings from this study could provide a basis for developing targeted sanitation protocols in food plants.


Subject(s)
Disinfectants , Pseudomonas fluorescens , Bacterial Adhesion , Pseudomonas , Polystyrenes , Biofilms , Disinfectants/pharmacology , Stainless Steel
2.
Histochem Cell Biol ; 157(5): 547-556, 2022 May.
Article in English | MEDLINE | ID: mdl-35201398

ABSTRACT

Growth-associated protein 43 (GAP43) is found in skeletal muscle, localized near the calcium release units. In interaction with calmodulin (CaM), it indirectly modulates the activity of dihydropyridine and ryanodine Ca2+ channels. GAP43-CaM interaction plays a key role in intracellular Ca2+ homeostasis and, consequently, in skeletal muscle activity. The control of intracellular Ca2+ signaling is also an important functional requisite in cardiac physiology. The aim of this study is to define the impact of GAP43 on cardiac tissue at macroscopic and cellular levels, using GAP43 knockout (GAP43-/-) newborn C57/BL6 mice. Hearts from newborn GAP43-/- mice were heavier than hearts from wild-type (WT) ones. In these GAP43-/- hearts, histological section analyses revealed a thicker ventricular wall and interventricular septum with a reduced ventricular chamber area. In addition, increased collagen deposits between fibers and increased expression levels of myosin were observed in hearts from GAP43-/- mice. Cardiac tropism and rhythm are controlled by multiple intrinsic and extrinsic factors, including cellular events such those linked to intracellular Ca2+ dynamics, in which GAP43 plays a role. Our data revealed that, in the absence of GAP43, there were cardiac morphological alterations and signs of hypertrophy, suggesting that GAP43 could play a role in the functional processes of the whole cardiac muscle. This paves the way for further studies investigating GAP43 involvement in signaling dynamics at the cellular level.


Subject(s)
Calmodulin , Ventricular Remodeling , Animals , Calcium/metabolism , Calmodulin/genetics , Calmodulin/metabolism , GAP-43 Protein/metabolism , Heart , Hypertrophy/metabolism , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism
3.
Int J Mol Sci ; 23(7)2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35408963

ABSTRACT

This study evaluates the effects of five different peptides, the Epitalon® tetrapeptide, the Vilon® dipeptide, the Thymogen® dipeptide, the Thymalin® peptide complex, and the Chonluten® tripeptide, as regulators of inflammatory and proliferative processes in the human monocytic THP-1, which is a human leukemia monocytic cell line capable of differentiating into macrophages by PMA in vitro. These peptides (Khavinson Peptides®), characterized by Prof. Khavinson from 1973 onwards, were initially isolated from animal tissues and found to be organ specific. We tested the capacity of the five peptides to influence cell cultures in vitro by incubating THP-1 cells with peptides at certain concentrations known for being effective on recipient cells in culture. We found that all five peptides can modulate key proliferative patterns, increasing tyrosine phosphorylation of mitogen-activated cytoplasmic kinases. In addition, the Chonluten tripeptide, derived from bronchial epithelial cells, inhibited in vitro tumor necrosis factor (TNF) production of monocytes exposed to pro-inflammatory bacterial lipopolysaccharide (LPS). The low TNF release by monocytes is linked to a documented mechanism of TNF tolerance, promoting attenuation of inflammatory action. Therefore, all peptides inhibited the expression of TNF and pro-inflammatory IL-6 cytokine stimulated by LPS on terminally differentiated THP-1 cells. Lastly, by incubating the THP1 cells, treated with the peptides, on a layer of activated endothelial cells (HUVECs activated by LPS), we observed a reduction in cell adhesion, a typical pro-inflammatory mechanism. Overall, the results suggest that the Khavinson Peptides® cooperate as natural inducers of TNF tolerance in monocyte, and act on macrophages as anti-inflammatory molecules during inflammatory and microbial-mediated activity.


Subject(s)
Lipopolysaccharides , Monocytes , Cytokines/metabolism , Dipeptides/pharmacology , Endothelial Cells/metabolism , Humans , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Monocytes/metabolism , THP-1 Cells , Tumor Necrosis Factor-alpha/metabolism
4.
J Cell Mol Med ; 25(12): 5381-5390, 2021 06.
Article in English | MEDLINE | ID: mdl-33949765

ABSTRACT

Cardiac stromal cells (CSCs) contain a pool of cells with supportive and paracrine functions. Various types of mesenchymal stromal cells (MSCs) can influence CSCs in the cardiac niche through their paracrine activity. Ischaemia/reperfusion (I/R) leads to cell death and reduction of the paracrine activity of CSCs. The forced co-expression of telomerase reverse transcriptase (TERT) and myocardin (MYOCD), known to potentiate anti-apoptotic, pro-survival and pro-angiogenic activities of MSCs isolated from the adipose tissue (AT-MSCs), may increase CSC survival, favouring their paracrine activities. We aimed at investigating the hypothesis that CSCs feature improved resistance to simulated I/R (SI/R) and increased commitment towards the cardiovascular lineage when preconditioned with conditioned media (CM) or extracellular vesicles (EV) released from AT-MSCs overexpressing TERT and MYOCD (T/M AT-MSCs). Murine CSCs were isolated with the cardiosphere (CSps) isolation technique. T/M AT-MSCs and their secretome improved spontaneous intracellular calcium changes and ryanodine receptor expression in aged CSps. The cytoprotective effect of AT-MSCs was tested in CSCs subjected to SI/R. SI/R induced cell death as compared to normoxia (28 ± 4 vs 10 ± 3%, P = .02). Pre-treatment with CM (15 ± 2, P = .02) or with the EV-enriched fraction (10 ± 1%, P = .02) obtained from mock-transduced AT-MSCs in normoxia reduced cell death after SI/R. The effect was more pronounced with CM (7 ± 1%, P = .01) or the EV-enriched fraction (2 ± 1%, P = .01) obtained from T/M AT-MSCs subjected to SI/R. In parallel, we observed lower expression of the apoptosis marker cleaved caspase-3 and higher expression of cardiac and vascular markers eNOS, sarcomeric α-actinin and cardiac actin. The T/M AT-MSCs secretome exerts a cytoprotective effect and promotes development of CSCs undergoing SI/R towards a cardiovascular phenotype.


Subject(s)
Biomarkers/metabolism , Cardiovascular Diseases/therapy , Heart/growth & development , Mesenchymal Stem Cells/cytology , Nuclear Proteins/metabolism , Reperfusion Injury/complications , Telomerase/metabolism , Trans-Activators/metabolism , Animals , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Telomerase/genetics , Trans-Activators/genetics
5.
Histochem Cell Biol ; 156(5): 423-436, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34370052

ABSTRACT

Periodontitis is a common inflammatory disease that affects the teeth-supporting tissue and causes bone and tooth loss. Moreover, in a worldwide population, periodontal disease is often associated with cardiovascular diseases. Emerging studies have reported that one of the major pathogens related to periodontitis is Porphyromonas gingivalis (P. gingivalis), which triggers the inflammatory intracellular cascade. Here, we hypothesized a possible protective effect of ascorbic acid (AA) in the restoration of the physiological molecular pathway after exposure to lipopolysaccharide derived from P. gingivalis (LPS-G). In particular, human gingiva-derived mesenchymal stem cells (hGMSCs) and endothelial-differentiated hGMSCs (e-hGMSCs) exposed to LPS-G showed upregulation of p300 and downregulation of DNA methyltransferase 1 (DNMT1), proteins associated with DNA methylation and histone acetylation. The co-treatment of AA and LPS-G showed a physiological expression of p300 and DNMT1 in hGMSCs and e-hGMSCs. Moreover, the inflammatory process triggered by LPS-G was demonstrated by evaluation of reactive oxygen species (ROS) and their intracellular localization. AA exposure re-established the physiological ROS levels. Despite the limitations of in vitro study, these findings collectively expand our knowledge regarding the molecular pathways involved in periodontal disease, and suggest the involvement of epigenetic modifications in the development of periodontitis.


Subject(s)
Ascorbic Acid/pharmacology , Endothelial Cells/drug effects , Mesenchymal Stem Cells/drug effects , Porphyromonas gingivalis/drug effects , Protective Agents/pharmacology , Ascorbic Acid/chemistry , Endothelial Cells/metabolism , Epigenesis, Genetic/drug effects , Humans , Mesenchymal Stem Cells/metabolism , Porphyromonas gingivalis/metabolism , Protective Agents/chemistry
6.
Int J Mol Sci ; 22(14)2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34299157

ABSTRACT

Curcumin, a yellow polyphenol extracted from the turmeric root is used as a diet supplement. It exhibits anti-inflammatory, antioxidant, and antitumor properties by modulating different intracellular mechanisms. Due to their low solubility in water, the curcumin molecules must be encapsulated into liposomes to improve the bioavailability and biomedical potential. For the periodontal tissue and systemic health, it is essential to regulate the local inflammatory response. In this study, the possible beneficial effect of liposomes loaded with curcumin (CurLIP) in neural crest-derived human periodontal ligament stem cells (hPDLSCs) and in endothelial-differentiated hPDLSCs (e-hPDLSCs) induced with an inflammatory stimulus (lipopolysaccharide obtained from Porphyromonas gingivalis, LPS-G) was evaluated. The CurLIP formulation exhibited a significant anti-inflammatory effect by the downregulation of Toll-like receptor-4 (TLR4)/Myeloid differentiation primary response 88 (MyD88)/nuclear factor kappa light chain enhancer of activated B cells (NFkB)/NLR Family Pyrin Domain Containing 3 (NLRP3)/Caspase-1/Interleukin (IL)-1ß inflammation cascade and reactive oxygen species (ROS) formation. Moreover, the exposure to LPS-G caused significant alterations in the expression of epigenetic modifiers, such as DNA Methyltransferase 1 (DNMT1) and P300, while the CurLIP treatment showed physiological expression. Overall, our in vitro study provides novel mechanistic insights into the intracellular pathway exert by CurLIP in the regulation of inflammation and epigenetic modifications.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Curcumin/pharmacology , Endothelium, Vascular/drug effects , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , Neural Crest/drug effects , Stem Cells/drug effects , Cell Differentiation , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Liposomes/administration & dosage , Liposomes/chemistry , Neural Crest/cytology , Neural Crest/metabolism , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Porphyromonas gingivalis/chemistry , Reactive Oxygen Species , Stem Cells/cytology , Stem Cells/metabolism
7.
Int J Mol Sci ; 21(10)2020 May 21.
Article in English | MEDLINE | ID: mdl-32455731

ABSTRACT

The effects induced by microgravity on human body functions have been widely described, in particular those on skeletal muscle and bone tissues. This study aims to implement information on the possible countermeasures necessary to neutralize the oxidative imbalance induced by microgravity on osteoblastic cells. Using the model of murine MC3T3-E1 osteoblast cells, cellular morphology, proliferation, and metabolism were investigated during exposure to simulated microgravity on a random positioning machine in the absence or presence of an antioxidant-the 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (Trolox). Our results confirm that simulated microgravity-induced morphological and metabolic alterations characterized by increased levels of reactive oxygen species and a slowdown of the proliferative rate. Interestingly, the use of Trolox inhibited the simulated microgravity-induced effects. Indeed, the antioxidant-neutralizing oxidants preserved cell cytoskeletal architecture and restored cell proliferation rate and metabolism. The use of appropriate antioxidant countermeasures could prevent the modifications and damage induced by microgravity on osteoblastic cells and consequently on bone homeostasis.


Subject(s)
Antioxidants/pharmacology , Chromans/pharmacology , Osteoblasts/drug effects , Weightlessness/adverse effects , Animals , Calcium/metabolism , Cell Line , Cell Proliferation , Cytoskeleton/metabolism , Mice , Osteoblasts/metabolism , Osteoblasts/physiology , Oxidative Stress
8.
Int J Mol Sci ; 20(8)2019 Apr 17.
Article in English | MEDLINE | ID: mdl-30999563

ABSTRACT

The presence of microgravity conditions deeply affects the human body functions at the systemic, organ and cellular levels. This study aimed to investigate the effects induced by simulated-microgravity on non-stimulated Jurkat lymphocytes, an immune cell phenotype considered as a biosensor of the body responses, in order to depict at the cellular level the effects of such a peculiar condition. Jurkat cells were grown at 1 g or on random positioning machine simulating microgravity. On these cells we performed: morphological, cell cycle and proliferation analyses using cytofluorimetric and staining protocols-intracellular Ca2+, reactive oxygen species (ROS), mitochondria membrane potential and O2- measurements using fluorescent probes-aconitase and mitochondria activity, glucose and lactate content using colorimetric assays. After the first exposure days, the cells showed a more homogeneous roundish shape, an increased proliferation rate, metabolic and detoxifying activity resulted in decreased intracellular Ca2+ and ROS. In the late exposure time, the cells adapted to the new environmental condition. Our non-activated proliferating Jurkat cells, even if responsive to altered external forces, adapted to the new environmental condition showing a healthy status. In order to define the cellular mechanism(s) triggered by microgravity, developing standardized experimental approaches and controlled cell culture and simulator conditions is strongly recommended.


Subject(s)
Lymphocytes/cytology , Weightlessness Simulation , Calcium/metabolism , Cell Shape , Glucose/metabolism , Humans , Jurkat Cells , Lymphocytes/metabolism , Membrane Potential, Mitochondrial , Oxidative Stress , Oxygen/metabolism
9.
Molecules ; 24(18)2019 Sep 04.
Article in English | MEDLINE | ID: mdl-31487916

ABSTRACT

Moringin [4-(α-L-rhamnosyloxy) benzyl isothiocyanate] is an isothiocyanate extracted from Moringa oleifera seeds. It is an antioxidant known for several biological properties useful in the treatment of neurodegenerative diseases. Several neurodegenerative disorders such as Parkinson's and Alzheimer's diseases are linked to dysfunctional mitochondria due to the resulting increase of Reactive Oxygen Species (ROS). Stem cell-based therapeutic treatments in neurodegenerative diseases provide an alternative strategy aimed to replace the impaired tissue. In this study were investigated the deregulated genes involved in mitophagy in the human periodontal ligament stem cells pretreated with moringin. The RNA-seq study reveals the downregulation of PINK1, with a fold change (FC) of -0.56, such as the genes involved in the phagophore formation (MAP1LC3B FC: -0.73, GABARAP FC: -0.52, GABARAPL1 FC: -0.70, GABARAPL2 FC: -0.39). The moringin pretreatment downregulates the pro-apoptotic gene BAX (-0.66) and upregulates the anti-apoptotic genes BCL2L12 (FC: 1.35) and MCL1 (FC: 0.36). The downregulation of the most of the caspases (CASP1 FC: -1.43, CASP4 FC: -0.18, CASP6 FC: -1.34, CASP7 FC: -0.46, CASP8 FC: -0.65) implies the inactivation of the apoptotic process. Our results suggest that mitochondrial dysfunctions induced by oxidative stress can be inhibited by moringin pretreatment in human periodontal ligament stem cells (hPDLSCs).


Subject(s)
Gene Expression , Isothiocyanates/pharmacology , Mitophagy/drug effects , Mitophagy/genetics , Periodontal Ligament/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Biomarkers , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Isothiocyanates/chemistry , Membrane Potential, Mitochondrial/drug effects , Models, Biological , Molecular Structure , Stem Cells/cytology , Transcriptome
10.
Molecules ; 24(12)2019 Jun 19.
Article in English | MEDLINE | ID: mdl-31248162

ABSTRACT

Surgical site infections (SSIs) represent the most common nosocomial infections, and surgical sutures are optimal surfaces for bacterial adhesion and biofilm formation. Staphylococcus spp., Enterococcus spp., and Escherichia coli are the most commonly isolated microorganisms. The aim of this research was to evaluate the antibiofilm activity of a medical device (MD) containing TIAB, which is a silver-nanotech patented product. The antibacterial effect was evaluated against Staphylococcus aureus ATCC 29213, Enterococcus faecalis ATCC 29212, and E. coli ATCC 25922 by assessing the minimum inhibitory concentration (MIC) by the Alamar Blue® (AB) assay. The antibiofilm effect was determined by evaluation of the minimum biofilm inhibitory concentration (MBIC) and colony-forming unit (CFU) count. Subsequently, the MD was applied on sutures exposed to the bacterial species. The antimicrobial and antibiofilm effects were evaluated by the agar diffusion test method, confocal laser scanning microscopy (CLSM), and scanning electron microscopy (SEM). The MIC was determined for S. aureus and E. faecalis at 2 mg/mL, while the MBIC was 1.5 mg/mL for S. aureus and 1 mg/mL for E. faecalis. The formation of an inhibition zone around three different treated sutures confirmed the antimicrobial activity, while the SEM and CLSM analysis performed on the MD-treated sutures underlined the presence of a few adhesive cells, which were for the most part dead. The MD showed antimicrobial and antibiofilm activities versus S. aureus and E. faecalis, but a lower efficacy against E. coli. Surgical sutures coated with the MD have the potential to reduce SSIs as well as the risk of biofilm formation post-surgery.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Coated Materials, Biocompatible , Equipment and Supplies/adverse effects , Equipment and Supplies/microbiology , Silver Compounds/chemistry , Surgical Wound Infection/etiology , Bacteria/drug effects , Bacteria/ultrastructure , Humans , Microbial Sensitivity Tests
11.
J Cell Physiol ; 233(10): 6734-6747, 2018 10.
Article in English | MEDLINE | ID: mdl-29600566

ABSTRACT

In the present study we have mimicked, in vitro, an inflammatory process using Lipopolysaccharide derived from Porphyromonas Gingivalis (LPS-G) and human Periodontal Ligament Stem Cells induced to endothelial differentiation (e-hPDLSCs). The research project has been organized into the three following steps: i) induction of hPDLSCs toward endothelial differentiation; ii) evaluation of the molecular signaling pathway involved in the response to the LPS-G, and iii) functional response evaluation of the living construct constituted by porcine decellularized valve/e-hPDLSCs treated with LPS-G. Obtained results showed that 5 µg/ml LPS-G stimulus provokes: a slowdown of cell growth starting from 24 hr and the release of IL6, IL8, and MCP1 molecules. Signaling network analyzed showed the activation of TLR4/ NFkB/ERK1/2/p-ERK1/2 signaling mediated by MyD88 in LPS-G stimulated e-hPDLSCs, moreover a time course put in evidence a nuclear traslocation of ERK1/2 and p-ERK1/2 in differentiated samples. Following, the ability of e-hPDLSCs to expand and colonize the decellularized porcine heart valves was appraised at ultrastructural level. Considering that, the Reactive Oxygen Species (ROS) play an important role in the progression and development of cardiovascular disease (CVD), in LPS-G living construct model e-hPDLSCs/decellularized porcine heart valves (dPHV), ROS production was assessed. Time lapse experiments evidenced that LPS-G provokes in e-hPDLSCs a rapid and sustained increase in ROS generation, negligible on undifferentiated cells. From obtained data, by multiparametric analyses, a reasonable conclusion may be that the inflammation process activated by LPS-G can affect endothelial cells and could represent in vivo a possible pathological and predictor state of CVD.


Subject(s)
Cardiovascular Diseases/genetics , Inflammation/genetics , Periodontal Diseases/genetics , Stem Cells/cytology , Animals , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/complications , Cardiovascular Diseases/pathology , Cell Differentiation/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Heart Valves/growth & development , Heart Valves/pathology , Humans , Inflammation/chemically induced , Inflammation/complications , Inflammation/pathology , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/genetics , Myeloid Differentiation Factor 88/genetics , NF-kappa B/genetics , Periodontal Diseases/chemically induced , Periodontal Diseases/complications , Periodontal Diseases/pathology , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Reactive Oxygen Species/metabolism , Stem Cells/pathology , Swine , Toll-Like Receptor 4/genetics
12.
Acta Neuropathol ; 135(5): 779-798, 2018 05.
Article in English | MEDLINE | ID: mdl-29305721

ABSTRACT

Tumours can be viewed as aberrant tissues or organs sustained by tumorigenic stem-like cells that engage into dysregulated histo/organogenetic processes. Paragangliomas, prototypical organoid tumours constituted by dysmorphic variants of the vascular and neural tissues found in normal paraganglia, provide a model to test this hypothesis. To understand the origin of paragangliomas, we built a biobank comprising 77 cases, 18 primary cultures, 4 derived cell lines, 80 patient-derived xenografts and 11 cell-derived xenografts. We comparatively investigated these unique complementary materials using morphofunctional, ultrastructural and flow cytometric assays accompanied by microRNA studies. We found that paragangliomas contain stem-like cells with hybrid mesenchymal/vasculoneural phenotype, stabilized and expanded in the derived cultures. The viability and growth of such cultures depended on the downregulation of the miR-200 and miR-34 families, which allowed high PDGFRA and ZEB1 protein expression levels. Both tumour tissue- and cell culture-derived xenografts recapitulated the vasculoneural paraganglioma structure and arose from mesenchymal-like cells through a fixed developmental sequence. First, vasculoangiogenesis organized the microenvironment, building a perivascular niche which in turn supported neurogenesis. Neuroepithelial differentiation was associated with severe mitochondrial dysfunction, not present in cultured paraganglioma cells, but acquired in vivo during xenograft formation. Vasculogenesis was the Achilles' heel of xenograft development. In fact, imatinib, that targets endothelial-mural signalling, blocked paraganglioma xenograft formation (11 xenografts from 12 cell transplants in the control group versus 2 out of 10 in the treated group, P = 0.0015). Overall our key results were unaffected by the SDHx gene carrier status of the patient, characterized for 70 out of 77 cases. In conclusion, we explain the biphasic vasculoneural structure of paragangliomas and identify an early and pharmacologically actionable phase of paraganglioma organization.


Subject(s)
Antineoplastic Agents/therapeutic use , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/physiopathology , Imatinib Mesylate/therapeutic use , Paraganglioma/drug therapy , Paraganglioma/physiopathology , Animals , Antineoplastic Agents/pharmacology , Cell Line , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Imatinib Mesylate/pharmacology , Mice, Inbred NOD , Mice, SCID , MicroRNAs/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Organogenesis/drug effects , Organogenesis/physiology , Paraganglioma/genetics , Paraganglioma/pathology , Primary Cell Culture , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology , Xenograft Model Antitumor Assays
14.
Int J Mol Sci ; 19(4)2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29673202

ABSTRACT

Improving the efficacy of gene therapy vectors is still an important goal toward the development of safe and efficient gene therapy treatments. S/MAR (scaffold/matrix attached region)-based vectors are maintained extra-chromosomally in numerous cell types, which is similar to viral-based vectors. Additionally, when established as an episome, they show a very high mitotic stability. In the present study we tested the idea that addition of an S/MAR element to a CFTR (cystic fibrosis transmembrane conductance regulator) expression vector, may allow the establishment of a CFTR episome in bronchial epithelial cells. Starting from the observation that the S/MAR vector pEPI-EGFP (enhanced green fluorescence protein) is maintained as an episome in human bronchial epithelial cells, we assembled the CFTR vector pBQ-S/MAR. This vector, transfected in bronchial epithelial cells with mutated CFTR, supported long term wt CFTR expression and activity, which in turn positively impacted on the assembly of tight junctions in polarized epithelial cells. Additionally, the recovery of intact pBQ-S/MAR, but not the parental vector lacking the S/MAR element, from transfected cells after extensive proliferation, strongly suggested that pBQ-S/MAR was established as an episome. These results add a new element, the S/MAR, that can be considered to improve the persistence and safety of gene therapy vectors for cystic fibrosis pulmonary disease.


Subject(s)
Bronchi/cytology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Genetic Vectors/genetics , Plasmids/genetics , Respiratory Mucosa/cytology , Bronchi/metabolism , Cell Line , Cystic Fibrosis/genetics , Cystic Fibrosis/therapy , Epithelial Cells/cytology , Epithelial Cells/metabolism , Genetic Therapy/methods , Humans , Respiratory Mucosa/metabolism , Transfection/methods
15.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3243-3253, 2017 12.
Article in English | MEDLINE | ID: mdl-28847515

ABSTRACT

Although cystic fibrosis (CF) patients exhibit signs of endothelial perturbation, the functions of the cystic fibrosis conductance regulator (CFTR) in vascular endothelial cells (EC) are poorly defined. We sought to uncover biological activities of endothelial CFTR, relevant for vascular homeostasis and inflammation. We examined cells from human umbilical cords (HUVEC) and pulmonary artery isolated from non-cystic fibrosis (PAEC) and CF human lungs (CF-PAEC), under static conditions or physiological shear. CFTR activity, clearly detected in HUVEC and PAEC, was markedly reduced in CF-PAEC. CFTR blockade increased endothelial permeability to macromolecules and reduced trans­endothelial electrical resistance (TEER). Consistent with this, CF-PAEC displayed lower TEER compared to PAEC. Under shear, CFTR blockade reduced VE-cadherin and p120 catenin membrane expression and triggered the formation of paxillin- and vinculin-enriched membrane blebs that evolved in shrinking of the cell body and disruption of cell-cell contacts. These changes were accompanied by enhanced release of microvesicles, which displayed reduced capability to stimulate proliferation in recipient EC. CFTR blockade also suppressed insulin-induced NO generation by EC, likely by inhibiting eNOS and AKT phosphorylation, whereas it enhanced IL-8 release. Remarkably, phosphodiesterase inhibitors in combination with a ß2 adrenergic receptor agonist corrected functional and morphological changes triggered by CFTR dysfunction in EC. Our results uncover regulatory functions of CFTR in EC, suggesting a physiological role of CFTR in the maintenance EC homeostasis and its involvement in pathogenetic aspects of CF. Moreover, our findings open avenues for novel pharmacology to control endothelial dysfunction and its consequences in CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/pathology , Endothelial Cells/pathology , Antigens, CD/metabolism , Cadherins/metabolism , Cell Proliferation/physiology , Cyclic AMP/metabolism , Cystic Fibrosis/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Homeostasis/physiology , Human Umbilical Vein Endothelial Cells , Humans , Insulin/pharmacology , Interleukin-8/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitrogen Oxides/metabolism , Phosphorylation , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , beta-Arrestin 2/metabolism
16.
J Mater Sci Mater Med ; 27(5): 88, 2016 May.
Article in English | MEDLINE | ID: mdl-26970770

ABSTRACT

Composite materials are increasingly used as dental restoration. In the field of biomaterials, infections remain the main reason of dental devices failure. Silver, in the form of nanoparticles (AgNPs), ions and salt, well known for its antimicrobial properties, is used in several medical applications in order to avoid bacterial infection. To reduce both bacterial adhesion to dental devices and cytotoxicity against eukaryotic cells, we coated BisGMA/TEGDMA methacrylic thermosets with a new material, Chitlac-nAg, formed by stabilized AgNPs with a polyelectrolyte solution containing Chitlac. Here we analyzed the proliferative and adhesive ability of human gingival fibroblasts (HGFs) on BisGMA/TEGDMA thermosets uncoated and coated with AgNPs in a coculture model system with Streptococcus mitis. After 48 h, HGFs well adhered onto both surfaces, while S. mitis cytotoxic response was higher in the presence of AgNPs coated thermosets. After 24 h thermosets coated with Chitlac as well as those coated with Chitlac-nAg exerted a minimal cytotoxic effect on HGFs, while after 48 h LDH release raised up to 20 %. Moreover the presence of S. mitis reduced this release mainly when HGFs adhered to Chitlac-nAg coated thermosets. The reduced secretion of collagen type I was significant in the presence of both surfaces with the co-culture system even more when saliva is added. Integrin ß1 localized closely to cell membranes onto Chitlac-nAg thermosets and PKCα translocated into nuclei. These data confirm that Chitlac-nAg have a promising utilization in the field of restorative dentistry exerting their antimicrobial activity due to AgNPs without cytotoxicity for eukaryotic cells.


Subject(s)
Bacterial Adhesion/physiology , Cell Adhesion/physiology , Fibroblasts/microbiology , Nanocomposites/chemistry , Streptococcus mitis/physiology , Cell Survival , Coculture Techniques , Culture Media , Fibroblasts/physiology , Humans , Materials Testing , Microscopy, Confocal , Microscopy, Electron, Scanning , Saliva , Surface Properties
17.
Cell Physiol Biochem ; 36(1): 259-73, 2015.
Article in English | MEDLINE | ID: mdl-25967965

ABSTRACT

BACKGROUND/AIMS: Mesenchymal stem cells from human amniotic fluid (huAFMSCs) can differentiate into multiple lineages and are not tumorigenic after transplantation, making them good candidates for therapeutic purposes. The aim was to determine the effects of calcitonin on these huAFMSCs during osteogenic differentiation, in terms of the physiological role of calcitonin in bone homeostasis. METHODS: For huAFMSCs cultured under different conditions, we assayed: expression of the calcitonin receptor, using immunolabelling techniques; proliferation and osteogenesis, using colorimetric and enzymatic assays; intracellular Ca(2+) and cAMP levels, using videomicroscopy and spectrophotometry. RESULTS: The calcitonin receptor was expressed in proliferating and osteo-differentiated huAFMSCs. Calcitonin triggered intracellular Ca(2+) increases and cAMP production. Its presence in cell medium also induced dose-dependent inhibitory effects on proliferation and increased osteogenic differentiation of huAFMSCs, as also indicated by enhancement of specific markers and alkaline phosphatase activity. CONCLUSIONS: These data show that huAFMSCs represent a potential osteogenic model to study in-vitro cell responses to calcitonin (and other members of the calcitonin family). This leads the way to the opening of new lines of research that will add new insight both in cell therapies and in the pharmacological use of these molecules.


Subject(s)
Amniotic Fluid/cytology , Calcitonin/pharmacology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Amniotic Fluid/drug effects , Amniotic Fluid/metabolism , Biomarkers/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Female , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Pregnancy , Receptors, Calcitonin/metabolism
18.
BMC Microbiol ; 15: 109, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25994406

ABSTRACT

BACKGROUND: Staphylococcus pseudintermedius is an opportunistic pathogen recognized as the leading cause of skin, ear, and post-operative bacterial infections in dogs and cats. Zoonotic infections have also recently been reported causing endocarditis, infection of surgical wounds, rhinosinusitis, and catheter-related bacteremia. The aim of the present study is to evaluate, for the first time, the pathogenic potential of S. pseudintermedius isolated from a human infection. To this end, strain DSM 25713, which was recently isolated from a wound of a leukemic patient who underwent a bone marrow transplantation, was investigated for biofilm formation and antibiotic-resistance under conditions relevant for wound infection. RESULTS: The effect of pH (5.5, 7.1, and 8.7) and the presence of serum (diluted at 1:2, 1:10, and 1:100) on biofilm formation was assessed through a crystal violet assay. The presence of serum significantly reduced the ability to form biofilm, regardless of the pH value tested. In vitro activity of eight antibiotics against biofilm formation and mature 48 h-old biofilms was comparatively assessed by crystal violet assay and viable cell count, respectively. Antibiotics at sub-inhibitory concentrations reduced biofilm formation in a dose-dependent manner, although cefoxitin was the most active, causing a significant reduction already at 1/8xMIC. Rifampicin showed the highest activity against preformed biofilms (MBEC90: 2xMIC). None of the antibiotics completely eradicated the preformed biofilms, regardless of tested concentrations. Confocal and electron microscopy analyses of mature biofilm revealed a complex "mushroom-like" architecture consisting of microcolonies embedded in a fibrillar extracellular matrix. CONCLUSIONS: For the first time, our results show that human wound-associated S. pseudintermedius is able to form inherently antibiotic-resistant biofilms, suggestive of its pathogenic potential, and consistent with recent reports of zoonotic infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Staphylococcus/drug effects , Staphylococcus/pathogenicity , Surgical Wound Infection/microbiology , Cefoxitin/pharmacology , Drug Resistance, Multiple, Bacterial , Humans , Hydrogen-Ion Concentration , Rifampin/pharmacology , Staphylococcal Infections/microbiology , Staphylococcus/isolation & purification , Staphylococcus/physiology
19.
Biofactors ; 50(3): 509-522, 2024.
Article in English | MEDLINE | ID: mdl-38131134

ABSTRACT

Mesenchymal stem cells (MSCs) treatment has been widely explored as a therapy for myocardial infarction, peripheral ischemic vascular diseases, dilated cardiomyopathy, and pulmonary hypertension. Latest in vitro studies suggest that MSCs can differentiate into contractile cardiomyocytes. One of the best-characterized MSCs products are MSCs-derived extracellular vesicles (EVs). EVs are crucial paracrine effectors of MSCs. Based on previous works, paracrine effects of MSCs play a primary role in the regenerative ability. Hence, in the current paper, we focused our attention on an alternative approach, exploiting products derived from human dental pulp stem cells (hDPSCs) rather than MSCs themselves, which may denote a cost-effective and safer approach. The focus has been on EVs and the bioactive molecules they contain to evaluate their ability to influence the differentiation process toward cardiomyogenic lineage. The expression of GATA4, ACTC1, CX43, and Nkx2.5 was evaluated using Immunofluorescence, real time-PCR, and Western blotting analyses. Furthermore, the expression profiling analysis of the microRNA hsa-miR-200c-3p, targeting the GATA4 gene, was studied. The hsa-miR-200c-3p was found significantly down-regulated in both c-hDPSCs + EVs-hDPSCs and c-hDPSCs + EVs-HL-1 compared to untreated c-hDPSCs underlying a possible epigenetic mechanism behind the prevalent up-regulation of its targeted GATA4 gene. The aim of the present work was to develop an in vitro model of hDPSCs able to differentiate into cardiomyocytes in order to investigate the role of EVs derived from hDPSCs and derived from HL-1 cardiomyocyte cell line in modulating the differentiation process toward cardiomyogenic lineage.


Subject(s)
Cell Differentiation , Dental Pulp , Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Myocytes, Cardiac , Regeneration , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , MicroRNAs/genetics , MicroRNAs/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Dental Pulp/cytology , Dental Pulp/metabolism , Regeneration/physiology , Regeneration/genetics , Homeobox Protein Nkx-2.5/metabolism , Homeobox Protein Nkx-2.5/genetics , GATA4 Transcription Factor/metabolism , GATA4 Transcription Factor/genetics , Connexin 43/metabolism , Connexin 43/genetics , Cells, Cultured
20.
Sci Rep ; 14(1): 10882, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38740792

ABSTRACT

The aim of this study was to evaluate the antimicrobial efficacy of an air gas soft jet CAP for its potential use in removing oral biofilms, given that plasma-based technologies have emerged as promising methods in periodontology. Two types of biofilms were developed, one by Streptococcus mutans UA 159 bacterial strain and the other by a complex mixture of saliva microorganisms isolated from a patient with periodontitis. This latter biofilm was characterized via Next Generation Sequencing to determine the main bacterial phyla. The CAP source was applied at a distance of 6 mm for different time points. A statistically significant reduction of both CFU count and XTT was already detected after 60 s of CAP treatment. CLSM analysis supported CAP effectiveness in killing the microorganisms inside the biofilm and in reducing the thickness of the biofilm matrix. Cytotoxicity tests demonstrated the possible use of CAP without important side effects towards human gingival fibroblasts cell line. The current study showed that CAP treatment was able to significantly reduce preformed biofilms developed by both S. mutans and microorganisms isolated by a saliva sample. Further studies should be conducted on biofilms developed by additional saliva donors to support the potential of this innovative strategy to counteract oral pathogens responsible for periodontal diseases.


Subject(s)
Biofilms , Plasma Gases , Saliva , Streptococcus mutans , Biofilms/drug effects , Biofilms/growth & development , Humans , Plasma Gases/pharmacology , Streptococcus mutans/drug effects , Streptococcus mutans/physiology , Saliva/microbiology , Fibroblasts/microbiology , Fibroblasts/drug effects , Periodontitis/microbiology , Periodontitis/therapy , Cell Line , Mouth/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL