Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cells ; 10(7)2021 06 23.
Article in English | MEDLINE | ID: mdl-34201858

ABSTRACT

Theepithelial-mesenchymal transition (EMT) is an essential event during cell development, in which epithelial cells acquire mesenchymal fibroblast-like features including reduced intercellular adhesion and increased motility. EMT also plays a key role in wound healing processes, which are mediated by inflammatory cells and fibroblasts. These cells secrete specific factors that interact with molecules of the extracellular matrix (ECM) such as collagens, laminins, elastin and tenascins. Wound healing follows four distinct and successive phases characterized by haemostasis, inflammation, cell proliferation and finally tissue remodeling. EMT is classified into three diverse subtypes: type-1 EMT, type-2 EMT and type-3 EMT. Type-1 EMT is involved in embryogenesis and organ development. Type-2 EMT is associated with wound healing, tissue regeneration and organ fibrosis. During organ fibrosis, type-2 EMT occurs as a reparative-associated process in response to ongoing inflammation and eventually leads to organ destruction. Type-3 EMT is implicated in cancer progression, which is linked to the occurrence of genetic and epigenetic alterations, in detail the ones promoting clonal outgrowth and the formation of localized tumors. The current review aimed at exploring the role of EMT process with particular focus on type-2 EMT in wound healing, fibrosis and tissue regeneration, as well as some recent progresses in the EMT and tissue regeneration field, including the modulation of EMT by biomaterials.


Subject(s)
Epithelial-Mesenchymal Transition , Organ Specificity , Wound Healing , Fibroblasts/pathology , Fibrosis , Humans , Regeneration
2.
Front Physiol ; 12: 676512, 2021.
Article in English | MEDLINE | ID: mdl-34093237

ABSTRACT

After oral mucosal injury, the healing response following specific steps that lead to wound closure and to tissue repair. Multiple cell populations are involved in this process; in particular, fibroblasts play a key role in the production of extracellular matrix (ECM). During wound healing the remodeling of ECM is a key stage to restore the tissue functionality through multifunctional fibroblast populations that are placed in the connective tissues of gingiva and periodontal ligament. Notably, a fibroblast sub-type (myofibroblast) is centrally involved in collagen synthesis and fibrillar remodeling. The present work evidenced the role of Transforming Growth Factor-beta1 (TGF-ß1) to mediate human gingival fibroblasts (hGFs) differentiation into myofibroblasts derived from gingival fibroblasts (myo-hGFs). The morphological and functional features were analyzed through Confocal Laser Scanning Microscopy (CLSM), flow cytometry, and western blotting analyses. The specific markers, such as alpha-Smooth Muscle Actin (α-SMA), Vimentin, E-cadherin, ß-catenin, and Smad 2/3, were modulated in myo-hGFs after the induction with TGF-ß1, at different time points (24, 48, and 72 h). After 72 h of treatment TGF-ß1 operates as an inducer of hGFs into myo-hGFs differentiation. We propose that TGF-ß1 may promote in vitro the fibroblasts-to-myofibroblasts transition via the morphological and molecular modifications, as the induction of α-SMA, Vimentin, E-cadherin, ß-catenin, and Smad 2/3.

3.
Int J Mol Sci ; 22(10)2021 May 15.
Article in English | MEDLINE | ID: mdl-34063438

ABSTRACT

In the last few decades, tissue engineering has become one of the most studied medical fields. Even if bone shows self-remodeling properties, in some cases, due to injuries or anomalies, bone regeneration can be required. In particular, oral bone regeneration is needed in the dentistry field, where the functional restoration of tissues near the tooth represents a limit for many dental implants. In this context, the application of biomaterials and mesenchymal stem cells (MSCs) appears promising for bone regeneration. This review focused on in vivo studies that evaluated bone regeneration using biomaterials with MSCs. Different biocompatible biomaterials were enriched with MSCs from different sources. These constructs showed an enhanced bone regenerative power in in vivo models. However, we discussed also a future perspective in tissue engineering using the MSC secretome, namely the conditioned medium and extracellular vesicles. This new approach has already shown promising results for bone tissue regeneration in experimental models.


Subject(s)
Biocompatible Materials/therapeutic use , Bone Regeneration/physiology , Mesenchymal Stem Cells , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Humans , Regenerative Medicine/methods , Tissue Scaffolds
4.
Antioxidants (Basel) ; 10(5)2021 May 18.
Article in English | MEDLINE | ID: mdl-34069836

ABSTRACT

Human gingival mesenchymal stem cells (hGMSCs) and endothelial committed hGMSCs (e-hGMSCs) have considerable potential to serve as an in vitro model to replicate the inflammation sustained by Porphyromonas gingivalis in periodontal and cardiovascular diseases. The present study aimed to investigate the effect of ascorbic acid (AA) on the inflammatory reverting action of lipopolysaccharide (LPS-G) on the cell metabolic activity, inflammation pathway and reactive oxygen species (ROS) generation in hGMSCs and e-hGMSCs. Cells were treated with LPS-G (5 µg mL-1) or AA (50 µg mL-1) and analyzed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay, immunofluorescence and Western blot methods. The rate of cell metabolic activity was decreased significantly in LPS-G-treated groups, while groups co-treated with LPS-G and AA showed a logarithmic cell metabolic activity rate similar to untreated cells. AA treatment attenuated the inflammatory effect of LPS-G by reducing the expression of TLR4/MyD88/NFκB/NLRP3/Caspase-1/IL-1ß, as demonstrated by Western blot analysis and immunofluorescence acquisition. LPS-G-induced cells displayed an increase in ROS production, while AA co-treated cells showed a protective effect. In summary, our work suggests that AA attenuated LPS-G-mediated inflammation and ROS generation in hGMSCs and e-hGMSCs via suppressing the NFκB/Caspase-1/IL-1ß pathway. These findings indicate that AA may be considered as a potential factor involved in the modulation of the inflammatory pathway triggered by LPS-G in an vitro cellular model.

5.
Oxid Med Cell Longev ; 2021: 6679708, 2021.
Article in English | MEDLINE | ID: mdl-33542783

ABSTRACT

Periodontitis is usually sustained from microorganism of oral cavity, like Porphyromonas gingivalis (P. gingivalis). Periodontal disease is an infectious disease that afflicts a large number of people. Researches are investigating on the mesenchymal stem cells (MSCs) response to inflammatory events in combination with antioxidant substances. In particular, ascorbic acid (AA) increased cell proliferation, upregulated the cells pluripotency marker expression, provide a protection from inflammation, and induced the regeneration of periodontal ligament tissue. The purpose of the present research was to investigate the effects of AA in primary culture of human periodontal ligament stem cells (hPDLSCs) exposed to P. gingivalis lipopolysaccharide (LPS-G). The effect of AA on hPDLSCs exposed to LPS-G was determined through the cell proliferation assay. The molecules involved in the inflammatory pathway and epigenetic regulation have been identified using immunofluorescence and Western blot analyses. miR-210 level was quantified by qRT-PCR, and the ROS generation was finally studied. Cells co-treated with LPS-G and AA showed a restoration in terms of cell proliferation. The expression of NFκB, MyD88, and p300 was upregulated in LPS-G exposed cells, while the expression was attenuated in the co-treatment with AA. DNMT1 expression is attenuated in the cells exposed to the inflammatory stimulus. The level of miR-210 was reduced in stimulated cells, while the expression was evident in the hPDLSCs co-treated with LPS-G and AA. In conclusion, the AA could enhance a protective effect in in vitro periodontitis model, downregulating the inflammatory pathway and ROS generation and modulating the miR-210 level.


Subject(s)
Ascorbic Acid/pharmacology , Epigenesis, Genetic/drug effects , Periodontitis/genetics , Stem Cells/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Humans , Lipopolysaccharides/isolation & purification , Lipopolysaccharides/pharmacology , Periodontal Ligament/drug effects , Periodontal Ligament/pathology , Periodontal Ligament/physiology , Periodontitis/chemically induced , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis/chemistry , Stem Cells/pathology , Stem Cells/physiology
6.
Int J Mol Sci ; 21(18)2020 Sep 17.
Article in English | MEDLINE | ID: mdl-32957696

ABSTRACT

At present, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection has quickly become a health emergency because no specifics vaccines or drugs, at this moment, are available. Recent studies have shown that the transplantation of mesenchymal stem cells (MSCs) into Coronavirus Disease 2019 (COVID-19) patients could represent a promising strategy for the development of new therapeutic methods. We speculate and suggest that the secretome of human Oral Tissue Stem Cells (hOTSCs), for their immunomodulatory and anti-inflammatory specific properties, could exert beneficial effects on the COVID-19 patients through an innovative aerosolisation technique. This non-invasive technique can offer multiple advantages in prophylaxis, as well as the prevention and treatment of severe epidemic respiratory syndrome with minimum risk and optimal therapeutic effects. This has the potential to create a novel pathway towards immunomodulatory therapy for the treatment of COVID-19 positive patients.


Subject(s)
Coronavirus Infections/drug therapy , Immunologic Factors/therapeutic use , Mesenchymal Stem Cells/metabolism , Mouth Mucosa/cytology , Pneumonia, Viral/drug therapy , Proteome/therapeutic use , COVID-19 , Humans , Immunologic Factors/metabolism , Pandemics , Proteome/metabolism , Secretory Pathway
7.
Eur J Histochem ; 63(3)2019 Sep 25.
Article in English | MEDLINE | ID: mdl-31696691

ABSTRACT

Recently, the development and the application of 3D scaffold able to promote stem cell differentiation represented an essential field of interest in regenerative medicine. In particular, functionalized scaffolds improve bone tissue formation and promote bone defects repair. This research aims to evaluate the role of ascorbic acid (AS) supplementation in an in vitro model, in which a novel 3D-scaffold, bovine pericardium collagen membrane called BioRipar (BioR) was functionalized with human Gingival Mesenchymal Stem Cells (hGMSCs). As extensively reported in the literature, AS is an essential antioxidant molecule involved in the extracellular matrix secretion and in the osteogenic induction. Specifically, hGMSCs were seeded on BioR and treated with 60 and 90 µg/mL of AS in order to assess their growth behavior, the expression of bone specific markers involved in osteogenesis (runt-related transcription factor 2, RUNX2; collagen1A1, COL1A1; osteopontin, OPN; bone morphogenetic protein2/4, BMP2/4), and de novo deposition of calcium. The expression of COL1A1, RUNX2, BMP2/4 and OPN was evaluated by RT-PCR, Western blotting and immunocytochemistry, and proved to be upregulated. Our results demonstrate that after three weeks of treatment AS at 60 and 90 µg/mL operates as an osteogenic inductor in hGMSCs. These data indicate that the AS supplementation produces an enhancement of osteogenic phenotype commitment in an in vitro environment. For this reason, AS could represent a valid support for basic and translational research in tissue engineering and regenerative medicine.


Subject(s)
Ascorbic Acid/metabolism , Collagen Type I/metabolism , Mesenchymal Stem Cells/metabolism , Pericardium/metabolism , Tissue Scaffolds/chemistry , Animals , Biomarkers/metabolism , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Cattle , Cell Differentiation/physiology , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/metabolism , Gingiva/metabolism , Humans , Osteogenesis/physiology , Osteopontin/metabolism , Pericardium/cytology , Regenerative Medicine/methods , Tissue Engineering/methods
8.
Int J Mol Sci ; 20(20)2019 Oct 09.
Article in English | MEDLINE | ID: mdl-31600975

ABSTRACT

Tissue engineering and/or regenerative medicine are fields of life science exploiting both engineering and biological fundamentals to originate new tissues and organs and to induce the regeneration of damaged or diseased tissues and organs. In particular, de novo bone tissue regeneration requires a mechanically competent osteo-conductive/inductive 3D biomaterial scaffold that guarantees the cell adhesion, proliferation, angiogenesis and differentiation into osteogenic lineage. Cellular components represent a key factor in tissue engineering and bone growth strategies take advantage from employment of mesenchymal stem cells (MSCs), an ideal cell source for tissue repair. Recently, the application of extracellular vesicles (EVs), isolated from stem cells, as cell-free therapy has emerged as a promising therapeutic strategy. This review aims at summarizing the recent and representative research on the bone tissue engineering field using a 3D scaffold enriched with human oral stem cells and their derivatives, EVs, as a promising therapeutic potential in the reconstructing of bone tissue defects.


Subject(s)
Biocompatible Materials , Bone Regeneration , Extracellular Vesicles/metabolism , Stem Cells/metabolism , Animals , Biomarkers , Collagen/metabolism , Humans , Phenotype , Regenerative Medicine , Tissue Engineering
9.
Nanomaterials (Basel) ; 9(7)2019 Jun 27.
Article in English | MEDLINE | ID: mdl-31252684

ABSTRACT

Dental pulp stem cells (DPSCs) represent a population of stem cells which could be useful in oral and maxillofacial reconstruction. They are part of the periendothelial niche, where their crosstalk with endothelial cells is crucial in the cellular response to biomaterials used for dental restorations. DPSCs and the endothelial cell line EA.hy926 were co-cultured in the presence of Chitlac-coated thermosets in culture conditions inducing, in turn, osteogenic or angiogenic differentiation. Cell proliferation was evaluated by 3-[4,5-dimethyl-thiazol-2-yl-]-2,5-diphenyl tetrazolium bromide (MTT) assay. DPSC differentiation was assessed by measuring Alkaline Phosphtase (ALP) activity and Alizarin Red S staining, while the formation of new vessels was monitored by optical microscopy. The IL-6 and PGE2 production was evaluated as well. When cultured together, the proliferation is increased, as is the DPSC osteogenic differentiation and EA.hy926 vessel formation. The presence of thermosets appears either not to disturb the system balance or even to improve the osteogenic and angiogenic differentiation. Chitlac-coated thermosets confirm their biocompatibility in the present co-culture model, being capable of improving the differentiation of both cell types. Furthermore, the assessed co-culture appears to be a useful tool to investigate cell response toward newly synthesized or commercially available biomaterials, as well as to evaluate their engraftment potential in restorative dentistry.

10.
Bioorg Med Chem Lett ; 27(7): 1608-1610, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28242276

ABSTRACT

Targeting the transcriptional activity of nuclear hormone receptors has proven an effective strategy to treat certain human diseases, and they have become a major focus point to develop novel therapies for the treatment of cancer, inflammation, autoimmune diseases, metabolic disorders, and others. One family of nuclear receptors that has attracted most interest in recent years is the retinoic acid receptor-related orphan receptors (RORs), in particular RORγ. RORγ is a critical regulator of the immune system and RORγ antagonists have shown activity in animal models of inflammatory autoimmune diseases. Here we present the synthesis and biological evaluation of dihydroimidazole tethered imidazolinethiones. We have identified several dual RORγ/α and pan-ROR antagonists with significant activity in cellular assays that could serve as starting points for future optimization efforts to generate potent and selective RORγ modulators.


Subject(s)
Imidazolines/pharmacology , Orphan Nuclear Receptors/antagonists & inhibitors , Thiones/pharmacology , Animals , CHO Cells , Cricetulus , Imidazolines/chemical synthesis , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 2/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Thiones/chemical synthesis
11.
ACS Chem Biol ; 12(2): 444-455, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28026162

ABSTRACT

Upregulation of antiapoptotic Bcl-2 proteins in certain tumors confers cancer cell resistance to chemotherapy or radiations. Members of the antiapoptotic Bcl-2 proteins, including Bcl-2, Mcl-1, Bcl-xL, Bcl-w, and Bfl-1, inhibit apoptosis by selectively binding to conserved α-helical regions, named BH3 domains, of pro-apoptotic proteins such as Bim, tBid, Bad, or NOXA. Five antiapoptotic proteins have been identified that interact with various selectivity with BH3 containing pro-apoptotic counterparts. Cancer cells present various and variable levels of these proteins, making the design of effective apoptosis based therapeutics challenging. Recently, BH3 profiling was introduced as a method to classify cancer cells based on their ability to resist apoptosis following exposure to selected BH3 peptides. However, these studies were based on binding affinities measured with model BH3 peptides and Bcl-2-proteins taken from mouse sequences. While the majority of these interactions are conserved between mice and humans, we found surprisingly that human NOXA binds to human Bfl-1 potently and covalently via conserved Cys residues, with over 2 orders of magnitude increased affinity over hMcl-1. Our data suggest that some assumptions of the original BH3 profiling need to be revisited and that perhaps further targeting efforts should be redirected toward Bfl-1, for which no suitable specific inhibitors or pharmacological tools have been reported. In this regard, we also describe the initial design and characterizations of novel covalent BH3-based agents that potently target Bfl-1. These molecules could provide a novel platform on which to design effective Bfl-1 targeting therapeutics.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Minor Histocompatibility Antigens/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Sequence , Apoptosis , Cell Line, Tumor , Humans
12.
Adv Exp Med Biol ; 885: 77-82, 2016.
Article in English | MEDLINE | ID: mdl-26747071

ABSTRACT

The carotid body is a highly specialized chemoreceptive organ of neural crest origin whose role is to detect changes in arterial oxygen content. The sensory units are the chemoreceptor cells, which are neuronal-like cells, surrounded by sustentacular or glial-like cells. It is suggested that the carotid body contains self-renewing multipotent stem cells, which are putatively represented by glial-like sustentacular cells. The mechanisms of renewal of neuronal-like cells are unclear. Recently, we have demonstrated the expression of galanin, a peptide promoting neurogenesis, in chemoreceptor cells in the human CB. Thus, in the present study we seek to determine whether galanin expression in chemoreceptor cells could be matched with that of nestin, a peptide that is a marker of multipotent neural stem cells, or rather with the glial fibrillary acidic protein (GFAP), a marker for glial cells. The latter would underscore the pluasibly essential role of sustentacular cells in the self-renewal capability of chemorecetors. We found that galanin expression is matched with nestin in chemoreceptor cells of the human carotid body, but not with that of GFAP. Thus, galanin expression in chemoreceptor cells could provide a signal for neurogenesis and chemoreceptor cell differentiation in the carotid body.


Subject(s)
Carotid Body/chemistry , Galanin/analysis , Nestin/analysis , Adult , Aged , Glial Fibrillary Acidic Protein/analysis , Humans , Immunohistochemistry , Middle Aged
13.
Front Physiol ; 5: 427, 2014.
Article in English | MEDLINE | ID: mdl-25400591

ABSTRACT

The carotid body is a highly specialized chemoreceptive structure for the detection of and reaction to hypoxia, through induction of an increase in hypoxia inducible factor. As tissue hypoxia increases with aging and can have dramatic effects in respiratory depression induced by drug addiction, we investigated the carotid body in young and old healthy subjects in comparison with drug-addicted subjects, including the expression of the neurotransmitter galanin. Galanin expression was recently reported for neuronal-like cells of the human carotid body, and it is implicated in several functions in neurons. In particular, this includes the regulation of differentiation of neural stem cells, and participation in the development and plasticity of the nervous system. Using immunohistochemistry detection, we demonstrate that galanin expression in the human carotid body in healthy older subjects and drug-addicted subjects is significantly reduced in comparison with healthy young subjects. This demonstrates not only the effects of normal aging and senescence, but also in the drug-addicted subjects, this appears to be due to a disorganization of the chemo-sensory region. With both aging and drug addiction, this results in a physiological reduction in neuronal-like cells, coupled with interlobular and intralobular increases in connective tissue fibers. Consequently, in both aging and drug addiction, this reduction of neuronal-like cells and the regeneration suggest that the carotid body is losing its sensory capabilities, with the transmission of chemoreceptive signals dramatically and vitally reduced. The level of galanin expression would thus provide a signal for neurogenesis in young subjects, and for neurodegeneration in older and drug-addicted subjects.

SELECTION OF CITATIONS
SEARCH DETAIL