Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Pharmazie ; 73(1): 16-18, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29441945

ABSTRACT

Aqueous pharmaceutical solutions provide prosperous living conditions for microbiological agents. In order to eliminate these microbes, we use preservatives which can harm human cells as well. Their cytotoxicity is concentration-dependent and the aim of our study was to find how other pharmaceutical excipients modify the cytotoxic attributes of preservatives. We tested the following compounds: methylparaben, benzalkonium chloride, polysorbate 20, Labrasol® and hydroxyethyl cellulose. The MTT tests indicated that surfactants increase the cytotoxicity while polymers may decrease it in some cases.


Subject(s)
Excipients/toxicity , Polymers/toxicity , Preservatives, Pharmaceutical/toxicity , Caco-2 Cells , Excipients/administration & dosage , Excipients/chemistry , Humans , Pharmaceutical Solutions , Polymers/administration & dosage , Polymers/chemistry , Preservatives, Pharmaceutical/administration & dosage , Preservatives, Pharmaceutical/chemistry , Toxicity Tests
2.
Oral Dis ; 24(1-2): 261-276, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29480614

ABSTRACT

This narrative review on the use of biophotonics therapies for management of oral diseases is written as a tribute to Prof. Crispian Scully. His seminal contributions to the field are highlighted by the detailed, comprehensive description of clinical presentations of oral diseases. This has enabled a more thorough, fundamental understanding of many of these pathologies by research from his group as well as inspired mechanistic investigations in many groups globally. In the same vein, a major emphasis of this narrative review is to focus on the evidence from human case reports rather than in vitro or in vivo animal studies that showcases the growing and broad impact of biophotonics therapies. The similarities and differences between two distinct forms of low-dose biophotonics treatments namely photodynamic therapy and photobiomodulation therapy are discussed. As evident in this review, a majority of these reports provide promising evidence for their clinical efficacy. However, a lack of adequate technical details, precise biological rationale, and limited outcome measures limits the current utility of these treatments. Future investigations should attempt to address these shortcomings and develop better designed, rigorous, controlled studies to fully harness the tremendous potential of low-dose biophotonics therapies.


Subject(s)
Bacterial Infections/drug therapy , Low-Level Light Therapy , Mouth Diseases/drug therapy , Mouth Diseases/radiotherapy , Photochemotherapy , Photons/therapeutic use , Bacterial Infections/prevention & control , Biofilms , Endodontics , Humans , Laser Therapy , Learning , Optics and Photonics , Phenotype
4.
J Periodontal Res ; 52(3): 360-367, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27396269

ABSTRACT

BACKGROUND: There is increasing popularity of high-power lasers for surgical debridement and antimicrobial therapy in the management of peri-implantitis and periodontal therapy. Removal of the noxious foci would naturally promote tissue healing directly. However, there are also anecdotal reports of better healing around routine high-power laser procedures. The precise mechanisms mediating these effects remain to be fully elucidated. This work examines these low-dose laser bystander effects on oral human epithelial and fibroblasts, particularly focusing on the role of human ß-defensin 2 (HBD-2 or DEFB4A), a potent factor capable of antimicrobial effects and promoting wound healing. MATERIAL AND METHODS: Laser treatments were performed using a near-infrared laser (810 nm diode) at low doses. Normal human oral keratinocytes and fibroblast cells were used and HBD-2 mRNA and protein expression was assessed with real time polymerase chain reaction, western blotting and immunostaining. Role of transforming growth factor (TGF)-ß1 signaling in this process was dissected using pathway-specific small molecule inhibitors. RESULTS: We observed laser treatments robustly induced HBD-2 expression in an oral fibroblast cell line compared to a keratinocyte cell line. Low-dose laser treatments results in activation of the TGF-ß1 pathway that mediated HBD-2 expression. The two arms of TGF-ß1 signaling, Smad and non-Smad are involved in laser-mediated HBD-2 expression. CONCLUSIONS: Laser-activated TGF-ß1 signaling and induced expression of HBD-2, both of which are individually capable of promoting healing in tissues adjacent to high-power surgical laser applications. Moreover, the use of low-dose laser therapy itself can provide additional therapeutic benefits for effective clinical management of periodontal or peri-implant disease.


Subject(s)
Low-Level Light Therapy , Peri-Implantitis/radiotherapy , Periodontitis/radiotherapy , Transforming Growth Factor beta1/metabolism , beta-Defensins/metabolism , Blotting, Western , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Keratinocytes/metabolism , Keratinocytes/radiation effects , Low-Level Light Therapy/methods , Real-Time Polymerase Chain Reaction
5.
J Dent Res ; 95(9): 977-84, 2016 08.
Article in English | MEDLINE | ID: mdl-27161014

ABSTRACT

The fundamental pathophysiologic response for the survival of all organisms is the process of wound healing. Inadequate or lack of healing constitutes the etiopathologic basis of many oral and systemic diseases. Among the numerous efforts to promote wound healing, biophotonics therapies have shown much promise. Advances in photonic technologies and a better understanding of light-tissue interactions, from parallel biophotonics fields such as in vivo optical imaging and optogenetics, are spearheading their popularity in biology and medicine. Use of high-dose lasers and light devices in dermatology, ophthalmology, oncology, and dentistry are now popular for specific clinical applications, such as surgery, skin rejuvenation, ocular and soft tissue recontouring, and antitumor and antimicrobial photodynamic therapy. However, a less well-known clinical application is the therapeutic use of low-dose biophotonics termed photobiomodulation (PBM) therapy, which is aimed at alleviating pain and inflammation, modulating immune responses, and promoting wound healing and tissue regeneration. Despite significant volumes of scientific literature from clinical and laboratory studies noting the phenomenological evidence for this innovative therapy, limited mechanistic insights have prevented rigorous and reproducible PBM clinical protocols. This article briefly reviews current evidence and focuses on gaps in knowledge to identify potential paths forward for clinical translation with PBM therapy with an emphasis on craniofacial wound healing. PBM offers a novel opportunity to examine fundamental nonvisual photobiological processes as well as develop innovative clinical therapies, thereby presenting an opportunity for a paradigm shift from conventional restorative/prosthetic approaches to regenerative modalities in clinical dentistry.


Subject(s)
Face/surgery , Low-Level Light Therapy , Skull/surgery , Wound Healing/radiation effects , Dentistry/methods , Facial Injuries/radiotherapy , Humans , Low-Level Light Therapy/methods , Skull/injuries
6.
J Dent Res ; 95(2): 188-95, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26503912

ABSTRACT

Tumor necrosis factor-α (TNF-α) is a proalgesic cytokine that is commonly expressed following tissue injury. TNF-α expression not only promotes inflammation but can also lead to pain hypersensitivity in nociceptors. With the established link between TNF-α and inflammatory pain, we identified its increased expression in the teeth of patients affected with caries and pulpitis. We generated a transgenic mouse model (TNF-α(glo)) that could be used to conditionally overexpress TNF-α. These mice were bred with a dentin matrix protein 1 (DMP1)-Cre line for overexpression of TNF-α in both the tooth pulp and bone to study oral pain that would result from subsequent development of pulpitis and bone loss. The resulting DMP1/TNF-α(glo) mice show inflammation in the tooth pulp that resembles pulpitis while also displaying periodontal bone loss. Inflammatory infiltrates and enlarged blood vessels were observed in the tooth pulp. Pulpitis and osteitis affected the nociceptive neurons innervating the orofacial region by causing increased expression of inflammatory cytokines within the trigeminal ganglia. With this new mouse model morphologically mimicking pulpitis and osteitis, we tested it for signs of oral pain with an oral function assay (dolognawmeter). This assay/device records the time required by a mouse to complete a discrete gnawing task. The duration of gnawing required by the DMP1/TNF-α(glo) mice to complete the task was greater than that for the controls; extended gnaw time in a dolognawmeter indicates reduced orofacial function. With the DMP1/TNF-α(glo) mice, we have shown that TNF-α expression alone can produce inflammation similar to pulpitis and osteitis and that this mouse model can be used to study dental inflammatory pain.


Subject(s)
Alveolar Process/metabolism , Nociceptors/metabolism , Osteitis/etiology , Pulpitis/etiology , Tooth/metabolism , Tumor Necrosis Factor-alpha/metabolism , Alveolar Bone Loss/etiology , Alveolar Bone Loss/metabolism , Animals , Dental Caries/metabolism , Dental Pulp/blood supply , Dilatation, Pathologic/pathology , Disease Models, Animal , Extracellular Matrix Proteins/physiology , Humans , Inflammation , Inflammation Mediators/metabolism , Mastication/physiology , Mice , Mice, Transgenic , Microvessels/pathology , Osteitis/metabolism , Pulpitis/metabolism , Time Factors , Toothache/metabolism , Transfection , Trigeminal Ganglion/metabolism , Tumor Necrosis Factor-alpha/genetics
7.
Oral Dis ; 21(1): e51-61, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24893951

ABSTRACT

Drug-induced gingival overgrowth (DIGO) is a disfiguring side effect of anti-convulsants, calcineurin inhibitors, and calcium channel blocking agents. A unifying hypothesis has been constructed which begins with cation flux inhibition induced by all three of these drug categories. Decreased cation influx of folic acid active transport within gingival fibroblasts leads to decreased cellular folate uptake, which in turn leads to changes in matrix metalloproteinases metabolism and the failure to activate collagenase. Decreased availability of activated collagenase results in decreased degradation of accumulated connective tissue which presents as DIGO. Studies supporting this hypothesis are discussed.


Subject(s)
Gingival Overgrowth/chemically induced , Animals , Anticonvulsants/adverse effects , Calcineurin Inhibitors/adverse effects , Calcium Channel Blockers/adverse effects , Gingiva/drug effects , Humans , Models, Biological
8.
J Dent Res ; 93(12): 1250-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25143513

ABSTRACT

Tissue loss due to oral diseases requires the healing and regeneration of tissues of multiple lineages. While stem cells are native to oral tissues, a current major limitation to regeneration is the ability to direct their lineage-specific differentiation. This work utilizes polymeric scaffold systems with spatiotemporally controlled morphogen cues to develop precise morphogen fields to direct mesenchymal stem cell differentiation. First, a simple three-layer scaffold design was developed that presented two spatially segregated, lineage-specific cues (Dentinogenic TGF-ß1 and Osteogenic BMP4). However, this system resulted in diffuse morphogen fields, as assessed by the in vitro imaging of cell-signaling pathways triggered by the morphogens. Mathematical modeling was then exploited, in combination with incorporation of specific inhibitors (neutralizing antibodies or a small molecule kinase inhibitor) into each morphogen in an opposing spatial pattern as the respective morphogen, to design a five-layer scaffold that was predicted to yield distinct, spatially segregated zones of morphogen signaling. To validate this system, undifferentiated MSCs were uniformly seeded in these scaffold systems, and distinct mineralized tissue differentiation were noted within these morphogen zones. Finally, to demonstrate temporal control over morphogen signaling, latent TGF-ß1 was incorporated into one region of a concentric scaffold design, and laser treatment was used to activate the morphogen on-demand and to induce dentin differentiation solely within that specific spatial zone. This study demonstrates a significant advance in scaffold design to generate precise morphogen fields that can be used to develop in situ models to explore tissue differentiation and may ultimately be useful in engineering multi-lineage tissues in clinical dentistry.


Subject(s)
Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/drug effects , Tissue Engineering/methods , Animals , Bone Morphogenetic Protein 4/pharmacology , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Lineage/drug effects , Dentinogenesis/drug effects , Diffusion , Humans , Lactic Acid/chemistry , Lasers, Semiconductor , Mice , Models, Biological , Osteogenesis/drug effects , Paracrine Communication/drug effects , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Prosthesis Design , Tissue Engineering/instrumentation , Tissue Scaffolds/chemistry , Tissue Scaffolds/classification , Transforming Growth Factor beta1/pharmacology
9.
Oral Dis ; 17(3): 241-51, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20860763

ABSTRACT

Oral Diseases (2011) 17, 241-251 The rapid advancement in basic biology knowledge, especially in the stem cell field, has created new opportunities to develop biomaterials capable of orchestrating the behavior of transplanted and host cells. Based on our current understanding of cellular differentiation, a conceptual framework for the use of materials to program cells in situ is presented, namely a domino vs a switchboard model, to highlight the use of single vs multiple cues in a controlled manner to modulate biological processes. Further, specific design principles of material systems to present soluble and insoluble cues that are capable of recruiting, programming and deploying host cells for various applications are presented. The evolution of biomaterials from simple inert substances used to fill defects, to the recent development of sophisticated material systems capable of programming cells in situ is providing a platform to translate our understanding of basic biological mechanisms to clinical care.


Subject(s)
Biocompatible Materials/pharmacology , Stem Cells/drug effects , Tissue Engineering/methods , Cell Adhesion/drug effects , Cell Adhesion Molecules/pharmacology , Cell Differentiation/drug effects , Cell Physiological Phenomena/drug effects , Guided Tissue Regeneration/methods , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Nucleic Acids/pharmacology
10.
Oncogene ; 27(17): 2507-12, 2008 Apr 10.
Article in English | MEDLINE | ID: mdl-17952112

ABSTRACT

The ability of transforming growth factor-beta (TGF-beta) to modulate various effects on distinct cell lineages has been a central feature of its multi-faceted nature. The purpose of this study was to access the effects of deletion of a key TGF-beta signal transducer, Smad3, on MAPK activation and v-Ras(Ha)-transformation of primary mouse embryonic fibroblasts (MEFs). We observe reduced TGF-beta1 and v-ras(Ha) mediated activation of the JNK and ERK MAPK pathway upon ablation of Smad3. Further, Smad3-deficient MEFs demonstrate resistance to v-ras(Ha)-induced transformation while the absence of Smad3 results in increased inhibition of farnesyl transferase activity. Taken together, these observations demonstrate that the absence of Smad3 protects fibroblasts from oncogenic transformation by (i) augmenting farnesyl transferase inhibition and (ii) suppressing the Ras-JNK MAPK pathway. These results provide new insights into the molecular mechanisms involved in v-Ras(Ha) oncogene-induced mesenchymal phenotypic transformation.


Subject(s)
Alkyl and Aryl Transferases/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Oncogene Protein p21(ras)/metabolism , Smad3 Protein/metabolism , Transgenes/genetics , Animals , Cell Line, Transformed , Cell Transformation, Neoplastic , Cells, Cultured , Chlorocebus aethiops , Enzyme Activation/drug effects , Gene Expression Regulation , MAP Kinase Signaling System/drug effects , Mice , Oncogene Protein p21(ras)/genetics , Phenotype , Smad3 Protein/deficiency , Smad3 Protein/genetics , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...