Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 9(1): 20325, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31889168

ABSTRACT

Due to increasing antibiotic resistance, the application of antimicrobial photodynamic therapy (aPDT) is gaining increasing popularity in dentistry. The aim of this study was to investigate the antimicrobial effects of aPDT using visible light (VIS) and water-filtered infrared-A (wIRA) in combination with a Hypericum perforatum extract on in situ oral biofilms. The chemical composition of H. perforatum extract was analyzed using ultra-high-performance liquid chromatography coupled with high resolution mass spectrometry (UPLC-HRMS). To obtain initial and mature oral biofilms in situ, intraoral devices with fixed bovine enamel slabs (BES) were carried by six healthy volunteers for two hours and three days, respectively. The ex situ exposure of biofilms to VIS + wIRA (200 mWcm-2) and H. perforatum (32 mg ml-1, non-rinsed or rinsed prior to aPDT after 2-min preincubation) lasted for five minutes. Biofilm treatment with 0.2% chlorhexidine gluconate solution (CHX) served as a positive control, while untreated biofilms served as a negative control. The colony-forming units (CFU) of the aPDT-treated biofilms were quantified, and the surviving microorganisms were identified using MALDI-TOF biochemical tests as well as 16 S rDNA-sequencing. We could show that the H. perforatum extract had significant photoactivation potential at a concentration of 32 mg ml-1. When aPDT was carried out in the presence of H. perforatum, all biofilms (100%) were completely eradicated (p = 0.0001). When H. perforatum was rinsed off prior to aPDT, more than 92% of the initial viable bacterial count and 13% of the mature oral biofilm were killed. Overall, the microbial composition in initial and mature biofilms was substantially altered after aPDT, inducing a shift in the synthesis of the microbial community. In conclusion, H. perforatum-mediated aPDT using VIS + wIRA interferes with oral biofilms, resulting in their elimination or the substantial alteration of microbial diversity and richness. The present results support the evaluation of H. perforatum-mediated aPDT for the adjunctive treatment of biofilm-associated oral diseases.


Subject(s)
Anti-Infective Agents/pharmacology , Biofilms/drug effects , Biofilms/radiation effects , Hypericum/chemistry , Infrared Rays , Light , Plant Extracts/pharmacology , Anti-Infective Agents/chemistry , Bacteria/drug effects , Bacteria/radiation effects , Bacterial Adhesion , Chromatography, High Pressure Liquid , Humans , Microbial Viability/drug effects , Microbial Viability/radiation effects , Mouth Mucosa/microbiology , Phytochemicals/chemistry , Phytochemicals/pharmacology , Plant Extracts/chemistry , Spectrometry, Mass, Electrospray Ionization
2.
J Neurosci ; 29(45): 14151-9, 2009 Nov 11.
Article in English | MEDLINE | ID: mdl-19906964

ABSTRACT

Restriction of adjacent same-type axons/dendrites to separate single columns for specific neuronal connections is commonly observed in vertebrates and invertebrates, and is necessary for proper processing of sensory information. Columnar restriction is conceptually similar to tiling, a phenomenon referring to the avoidance of neurites from adjacent same-type neurons. The molecular mechanism underlying the establishment of columnar restriction or axonal/dendritic tiling remains largely undefined. Here, we identify Turtle (Tutl), a member of the conserved Tutl/Dasm1/IgSF9 subfamily of the Ig superfamily, as a key player in regulating the tiling pattern of R7 photoreceptor terminals in Drosophila. Tutl functions to prevent fusion between two adjacent R7 terminals, and acts in parallel to the Activin pathway. Tutl mediates homophilic cell-cell interactions. We propose that extrinsic terminal-terminal recognition mediated by Tutl, acts in concert with intrinsic Activin-dependent control of terminal growth, to restrict the connection made by each R7 axon to a single column.


Subject(s)
Axons/physiology , Drosophila Proteins/metabolism , Immunoglobulins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Photoreceptor Cells, Invertebrate/physiology , Activins/metabolism , Animals , Animals, Genetically Modified , Cell Communication/physiology , Cell Line , Drosophila , Drosophila Proteins/genetics , Immunoglobulins/genetics , Male , Medulla Oblongata/growth & development , Medulla Oblongata/pathology , Medulla Oblongata/physiology , Membrane Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Optic Lobe, Nonmammalian/growth & development , Optic Lobe, Nonmammalian/pathology , Optic Lobe, Nonmammalian/physiology , Signal Transduction
3.
Mol Cell Biol ; 27(13): 4825-43, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17452454

ABSTRACT

The cell cycle regulatory retinoblastoma (Rb) protein is a key regulator of neural precursor proliferation; however, its role has been expanded to include a novel cell-autonomous role in mediating neuronal migration. We sought to determine the Rb-interacting factors that mediate both the cell cycle and migration defects. E2F1 and E2F3 are likely Rb-interacting candidates that we have shown to be deregulated in the absence of Rb. Using mice with compound null mutations of Rb and E2F1 or E2F3, we asked to what extent either E2F1 or E2F3 interacts with Rb in neurogenesis. Here, we report that E2F1 and E2F3 are both functionally relevant targets in neural precursor proliferation, cell cycle exit, and laminar patterning. Each also partially mediates the Rb requirement for neuronal survival. Neuronal migration, however, is specifically mediated through E2F3, beyond its role in cell cycle regulation. This study not only outlines overlapping and distinct functions for E2Fs in neurogenesis but also is the first to establish a physiologically relevant role for the Rb/E2F pathway beyond cell cycle regulation in vivo.


Subject(s)
Cell Cycle , Cell Movement , E2F3 Transcription Factor/metabolism , Neurons/cytology , Retinoblastoma Protein/metabolism , Animals , Cell Proliferation , Cell Survival , E2F1 Transcription Factor/metabolism , Female , Gene Expression Regulation , Interneurons/cytology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Protein Binding , Stem Cells/cytology , Stem Cells/metabolism , Telencephalon/embryology , Telencephalon/metabolism
4.
EMBO J ; 24(24): 4381-91, 2005 Dec 21.
Article in English | MEDLINE | ID: mdl-16308563

ABSTRACT

Precise cell cycle regulation is critical for nervous system development. To assess the role of the cell cycle regulator, retinoblastoma (Rb) protein, in forebrain development, we studied mice with telencephalon-specific Rb deletions. We examined the role of Rb in neuronal specification and migration of diverse neuronal populations. Although layer specification occurred at the appropriate time in Rb mutants, migration of early-born cortical neurons was perturbed. Consistent with defects in radial migration, neuronal cell death in Rb mutants specifically affected Cajal-Retzius neurons. In the ventral telencephalon, although calbindin- and Lhx6-expressing cortical neurons were generated at embryonic day 12.5, their tangential migration into the neocortex was dramatically and specifically reduced in the mutant marginal zone. Cell transplantation assays revealed that defects in tangential migration arose owing to a cell-autonomous loss of Rb in migrating interneurons and not because of a defective cortical environment. These results revealed a cell-autonomous role for Rb in regulating the tangential migration of cortical interneurons. Taken together, we reveal a novel requirement for the cell cycle protein, Rb, in the regulation of neuronal migration.


Subject(s)
Gene Expression Regulation, Developmental , Neurons/metabolism , Retinoblastoma Protein/metabolism , Animals , Apoptosis , Body Patterning , Calbindins , Cell Cycle , Cell Differentiation , Cell Lineage , Cell Movement , Cell Survival , Coculture Techniques , Embryo, Mammalian/metabolism , Genotype , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , LIM-Homeodomain Proteins , Mice , Models, Anatomic , Mutation , Nerve Tissue Proteins/metabolism , Retinoblastoma/metabolism , S100 Calcium Binding Protein G/metabolism , Stem Cells/metabolism , Telencephalon/metabolism , Time Factors , Transcription Factors
5.
J Neurosci ; 25(6): 1324-34, 2005 Feb 09.
Article in English | MEDLINE | ID: mdl-15703386

ABSTRACT

Mitochondria release proteins that propagate both caspase-dependent and caspase-independent cell death pathways. AIF (apoptosis-inducing factor) is an important caspase-independent death regulator in multiple neuronal injury pathways. Presently, there is considerable controversy as to whether AIF is neuroprotective or proapoptotic in neuronal injury, such as oxidative stress or excitotoxicity. To evaluate the role of AIF in BAX-dependent (DNA damage induced) and BAX-independent (excitotoxic) neuronal death, we used Harlequin (Hq) mice, which are hypomorphic for AIF. Neurons carrying double mutations for Hq/Apaf1-/- (apoptosis proteases-activating factor) are impaired in both caspase-dependent and AIF-mediated mitochondrial cell death pathways. These mutant cells exhibit extended neuroprotection against DNA damage, as well as glutamate-induced excitotoxicity. Specifically, AIF is involved in NMDA- and kainic acid- but not AMPA-induced excitotoxicity. In vivo excitotoxic studies using kainic acid-induced seizure showed that Hq mice had significantly less hippocampal damage than wild-type littermates. Our results demonstrate an important role for AIF in both BAX-dependent and BAX-independent mechanisms of neuronal injury.


Subject(s)
Apoptosis/physiology , Flavoproteins/physiology , Membrane Proteins/physiology , Neurons/cytology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Apoptosis Inducing Factor , Apoptotic Protease-Activating Factor 1 , Benzodiazepines/pharmacology , Benzothiadiazines/pharmacology , Camptothecin/pharmacology , Caspase Inhibitors , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cerebellum/cytology , Cerebral Cortex/cytology , Convulsants/toxicity , Dizocilpine Maleate/pharmacology , Drug Resistance , Flavoproteins/genetics , Glutamic Acid/pharmacology , Glycine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Kainic Acid/pharmacology , Kainic Acid/toxicity , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Mutant Strains , N-Methylaspartate/pharmacology , Neurons/drug effects , Neurons/metabolism , Neurotoxins/pharmacology , Proteins/genetics , Recombinant Fusion Proteins/physiology , Seizures/chemically induced , Seizures/metabolism , Seizures/pathology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , bcl-2-Associated X Protein
6.
J Cell Biol ; 166(6): 853-63, 2004 Sep 13.
Article in English | MEDLINE | ID: mdl-15353549

ABSTRACT

Here we show a novel function for Retinoblastoma family member, p107 in controlling stem cell expansion in the mammalian brain. Adult p107-null mice had elevated numbers of proliferating progenitor cells in their lateral ventricles. In vitro neurosphere assays revealed striking increases in the number of neurosphere forming cells from p107(-/-) brains that exhibited enhanced capacity for self-renewal. An expanded stem cell population in p107-deficient mice was shown in vivo by (a) increased numbers of slowly cycling cells in the lateral ventricles; and (b) accelerated rates of neural precursor repopulation after progenitor ablation. Notch1 was up-regulated in p107(-/-) neurospheres in vitro and brains in vivo. Chromatin immunoprecipitation and p107 overexpression suggest that p107 may modulate the Notch1 pathway. These results demonstrate a novel function for p107 that is distinct from Rb, which is to negatively regulate the number of neural stem cells in the developing and adult brain.


Subject(s)
Brain/cytology , Gene Expression Regulation, Developmental , Neurons/metabolism , Retinoblastoma Protein/genetics , Stem Cells/metabolism , Adenoviridae/genetics , Animals , Apoptosis , Blotting, Western , Bromodeoxyuridine/metabolism , Cell Division , Cells, Cultured , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Knockout , Olfactory Bulb/cytology , RNA, Messenger/metabolism , Receptors, Notch
7.
Trends Neurosci ; 26(6): 283-5, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12798593

ABSTRACT

Cortical development is a complex process in which extrinsic and intrinsic factors modulate the sequential generation of neurons and glia. Following successive rounds of division, precursors become determined along a neuronal or glial lineage prior to cell cycle exit and differentiation. Although the influence of growth factors in cell fate specification is not new, until recently little was known about the signaling pathways by which they regulate neuronal differentiation. Menard and colleagues have examined this issue and have demonstrated a role for the MEK-C/EBP (mitogen-activated-protein-kinase kinase and CCAAT/enhancer-binding protein) pathway in the promotion of growth factor-mediated neurogenesis.


Subject(s)
Cerebral Cortex/growth & development , Growth Substances/physiology , Neuroglia/metabolism , Neurons/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/physiology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Mitogen-Activated Protein Kinase Kinases/metabolism , Multipotent Stem Cells/physiology , Signal Transduction/physiology
8.
EMBO J ; 21(13): 3337-46, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12093735

ABSTRACT

Correct cell cycle regulation and terminal mitosis are critical for nervous system development. The retinoblastoma (Rb) protein is a key regulator of these processes, as Rb-/- embryos die by E15.5, exhibiting gross hematopoietic and neurological defects. The extensive apoptosis in Rb-/- embryos has been attributed to aberrant S phase entry resulting in conflicting growth control signals in differentiating cells. To assess the role of Rb in cortical development in the absence of other embryonic defects, we examined mice with telencephalon-specific Rb deletions. Animals carrying a floxed Rb allele were interbred with mice in which cre was knocked into the Foxg1 locus. Unlike germline knockouts, mice specifically deleted for Rb in the developing telencephalon survived until birth. In these mutants, Rb-/- progenitor cells divided ectopically, but were able to survive and differentiate. Mutant brains exhibited enhanced cellularity due to increased proliferation of neuroblasts. These studies demonstrate that: (i) cell cycle deregulation during differentiation does not necessitate apoptosis; (ii) Rb-deficient mutants exhibit enhanced neuroblast proliferation; and (iii) terminal mitosis may not be required to initiate differentiation.


Subject(s)
Nerve Tissue Proteins/physiology , Retinoblastoma Protein/physiology , Telencephalon/embryology , Alleles , Animals , Apoptosis , Biomarkers , Cell Cycle , Cell Differentiation , Cell Division , Cerebral Cortex/abnormalities , Cerebral Cortex/embryology , Crosses, Genetic , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Fetal Death/genetics , Forkhead Transcription Factors , Genes, Lethal , Genes, Retinoblastoma , Mice , Mice, Knockout , Mutagenesis, Insertional , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/pathology , Organ Specificity , Retinoblastoma Protein/deficiency , Telencephalon/abnormalities , Tubulin/biosynthesis , Tubulin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...