Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Adv Healthc Mater ; 11(2): e2101854, 2022 01.
Article in English | MEDLINE | ID: mdl-34748685

ABSTRACT

Uncontrolled amyloid-beta (Aß) fibrillation leads to the deposition of neurotoxic amyloid plaques and is associated with Alzheimer's disease. Inhibiting Aß monomer fibrillation and dissociation of the formed fibers is regarded as a promising therapeutic strategy. Here, amphiphilic polyphenylene dendrons (APDs) are demonstrated to interrupt Aß assembly and reduce Aß-cell interactions. Containing alternating negatively charged sulfonic acid and hydrophobic n-propyl peripheral groups, APDs bind to the secondary structure of the Aß aggregates, inhibiting fibrillation and disassemble the already formed Aß fibrils. APDs reveal vesicular cellular uptake in endosomes as well as cell compatibility for endothelial and neuronal cells, and significantly reduce Aß-induced neuron cytotoxicity in vitro. Moreover, they are transported into the brain and successfully cross the blood-brain barrier after systemic application in mice, indicating their high potential to inhibit Aß fibrillation in vivo, which can be beneficial for developing therapeutic strategy for Alzheimer's disease.


Subject(s)
Alzheimer Disease , Dendrimers , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Animals , Dendrimers/pharmacology , Mice , Neurons/metabolism , Polymers
2.
J Comp Neurol ; 528(1): 3-13, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31226222

ABSTRACT

Endocannabinoids and their receptors are highly abundant in the developing cerebral cortex and play major roles in early developmental processes, for example, neuronal proliferation, migration, and axonal guidance as well as postnatal plasticity. To investigate the role of the cannabinoid type 1 receptor (CB1) in the formation of sensory maps in the cerebral cortex, the topographic representation of the whiskers in the primary somatosensory cortex (barrel field) of adult mice with different cell type specific genetic deletion of CB1 was studied. A constitutive absence of CB1 (CB1-KO) significantly decreased the total area of the somatosensory cortical map, affecting barrel, and septal areas. Cell specific CB1 deletion in dorsal telencephalic glutamatergic neurons only (Glu-CB1-KO) or in both glutamatergic and forebrain GABAergic neurons (Glu/GABA-CB1-KO) resulted in an increased septa area in the barrel field map. No significant modifications in area parameters could be observed in GABA-CB1-KO mice. These data demonstrate that CB1 signaling especially in cortical glutamatergic neurons is essential for the development of topographic maps in the cerebral cortex.


Subject(s)
Brain Mapping/methods , Receptor, Cannabinoid, CB1/deficiency , Signal Transduction/physiology , Somatosensory Cortex/cytology , Somatosensory Cortex/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Receptor, Cannabinoid, CB1/analysis
3.
Small ; 15(42): e1902992, 2019 10.
Article in English | MEDLINE | ID: mdl-31465151

ABSTRACT

Nanotheranostics, combining diagnostics and therapy, has the potential to revolutionize treatment of neurological disorders. But one of the major obstacles for treating central nervous system diseases is the blood-brain barrier (BBB) preventing systemic delivery of drugs and optical probes into the brain. To overcome these limitations, nanodiamonds (NDs) are investigated in this study as they are a powerful sensing and imaging platform for various biological applications and possess outstanding stable far-red fluorescence, do not photobleach, and are highly biocompatible. Herein, fluorescent NDs encapsulated by a customized human serum albumin-based biopolymer (polyethylene glycol) coating (dcHSA-PEG) are taken up by target brain cells. In vitro BBB models reveal transcytosis and an additional direct cell-cell transport via tunneling nanotubes. Systemic application of dcHSA-NDs confirms their ability to cross the BBB in a mouse model. Tracking of dcHSA-NDs is possible at the single cell level and reveals their uptake into neurons and astrocytes in vivo. This study shows for the first time systemic NDs brain delivery and suggests transport mechanisms across the BBB and direct cell-cell transport. Fluorescent NDs are envisioned as traceable transporters for in vivo brain imaging, sensing, and drug delivery.


Subject(s)
Brain/metabolism , Nanodiamonds/chemistry , Animals , Astrocytes/metabolism , Biological Transport , Blood-Brain Barrier/metabolism , Cell Movement , Cell Survival , Endocytosis , Endothelial Cells/metabolism , Fluorescence , Mice , Nanodiamonds/ultrastructure , Neurons/metabolism , Polyethylene Glycols/chemistry , Serum Albumin, Human/chemistry
4.
J Neurotrauma ; 36(14): 2272-2278, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30755137

ABSTRACT

Plasminogen activator inhibitor-2 (PAI-2/SerpinB2) inhibits extracellular urokinase plasminogen activator (uPA). Under physiological conditions, PAI-2 is expressed at low levels but is rapidly induced by inflammatory triggers. It is a negative regulator of fibrinolysis and serves to stabilize clots. In the present study, PAI-2 expression is upregulated 25-fold in pericontusional brain tissue at 6 h after traumatic brain injury (TBI), with a maximum increase of 87-fold at 12 h. To investigate a potentially detrimental influence of PAI-2 on secondary post-traumatic processes, male PAI-2-deficient (PAI-2-KO) and wild-type mice (WT) were subjected to TBI by controlled cortical impact injury. Brain lesion volume and cerebral inflammation were not different. Total brain volume was significantly smaller in PAI-2-KO, indicating reduced brain swelling. The brain water content at 24 h post-insult was significantly smaller in PAI-2-KO mice. Markers of vasogenic brain edema showed no difference in blood-brain barrier integrity and expression of blood-brain barrier proteins (claudin-5, zonula occludens-1). In contrast to plasminogen activator inhibitor-1 (PAI-1), PAI-2 plays a limited role for brain lesion formation and does not influence blood-brain barrier integrity. PAI-2 contributes to brain edema formation and could therefore be a promising new target to treat post-traumatic brain edema.


Subject(s)
Brain Edema/metabolism , Brain Edema/pathology , Brain Injuries, Traumatic/pathology , Plasminogen Activator Inhibitor 2/metabolism , Animals , Brain Edema/etiology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Adv Mater ; 31(2): e1805044, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30411838

ABSTRACT

The synthesis of hybrid hydrogels by pH-controlled structural transition with exceptional rheological properties as cellular matrix is reported. "Depsi" peptide sequences are grafted onto a polypeptide backbone that undergo a pH-induced intramolecular O-N-acyl migration at physiological conditions affording peptide nanofibers (PNFs) as supramolecular gelators. The polypeptide-PNF hydrogels are mechanically remarkably robust. They reveal exciting thixotropic behavior with immediate in situ recovery after exposure to various high strains over long periods and self-repair of defects by instantaneous reassembly. High cytocompatibility, convenient functionalization by coassembly, and controlled enzymatic degradation but stability in 2D and 3D cell culture as demonstrated by the encapsulation of primary human umbilical vein endothelial cells and neuronal cells open many attractive opportunities for 3D tissue engineering and other biomedical applications.

6.
Adv Sci (Weinh) ; 5(5): 1700897, 2018 May.
Article in English | MEDLINE | ID: mdl-29876217

ABSTRACT

Neurological disorders are undoubtedly among the most alarming diseases humans might face. In treatment of neurological disorders, the blood-brain barrier (BBB) is a challenging obstacle preventing drug penetration into the brain. Advances in dendrimer chemistry for central nervous system (CNS) treatments are presented here. A poly(amido)amine (PAMAM) dendrimer bioconjugate with a streptavidin adapter for the attachment of dendrons or any biotinylated drug is constructed. In vitro studies on porcine or murine models and in vivo mouse studies are performed and reveal the permeation of dendronized streptavidin (DSA) into the CNS. The bioconjugate is taken up mainly by the caveolae pathway and transported across the BBB via transcytosis escaping from lysosomes. After transcytosis DSA are delivered to astrocytes and neurons. Furthermore, DSA offer high biocompatibility in vitro and in vivo. In summary, a new strategy for implementing therapeutic PAMAM function as well as drug delivery in neuropathology is presented here.

7.
Adv Healthc Mater ; 4(3): 377-84, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25182694

ABSTRACT

The design and synthesis of a polyphenylene dendrimer (PPD 3) with discrete binding sites for lipophilic guest molecules and characteristic surface patterns is presented. Its semi-rigidity in combination with a precise positioning of hydrophilic and hydrophobic groups at the periphery yields a refined architecture with lipophilic binding pockets that accommodate defined numbers of biologically relevant guest molecules such as fatty acids or the drug doxorubicin. The size, architecture, and surface textures allow to even penetrate brain endothelial cells that are a major component of the extremely tight blood-brain barrier. In addition, low to no toxicity is observed in in vivo studies using zebrafish embryos. The unique PPD scaffold allows the precise placement of functional groups in a given environment and offers a universal platform for designing drug transporters that closely mimic many features of proteins.


Subject(s)
Dendrimers/administration & dosage , Dendrimers/chemistry , Doxorubicin/administration & dosage , Polymers/administration & dosage , Animals , Brain/cytology , Cell Line/drug effects , Chemistry Techniques, Synthetic , Dendrimers/pharmacokinetics , Doxorubicin/chemistry , Drug Carriers , Drug Design , Embryo, Nonmammalian/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Polymers/chemistry , Polymers/pharmacokinetics , Tissue Distribution , Zebrafish/embryology
8.
Methods Mol Biol ; 1135: 403-13, 2014.
Article in English | MEDLINE | ID: mdl-24510882

ABSTRACT

The cerebral microvasculature possesses certain cellular features that constitute the blood-brain barrier (BBB) (Abbott et al., Neurobiol Dis 37:13-25, 2010). This dynamic barrier separates the brain parenchyma from peripheral blood flow and is of tremendous clinical importance: for example, BBB breakdown as in stroke is associated with the development of brain edema (Rosenberg and Yang, Neurosurg Focus 22:E4, 2007), inflammation (Kuhlmann et al., Neurosci Lett 449:168-172, 2009; Coisne and Engelhardt, Antioxid Redox Signal 15:1285-1303, 2011), and increased mortality. In vivo, the BBB consists of brain endothelial cells (BEC) that are embedded within a precisely regulated environment containing astrocytes, pericytes, smooth muscle cells, and glial cells. These cells experience modulation by various pathways of intercellular communication and by pathophysiological processes, e.g., through neurovascular coupling (Attwell et al., Nature 468:232-243, 2010), cortical spreading depression (Gursoy-Ozdemir et al., J Clin Invest 113:1447-1455, 2004), or formation of oxidative stress (Yemisci et al., Nat Med 15:1031-1037, 2009). Hence, this interdependent assembly of cells is referred to as the neurovascular unit (NVU) (Zlokovic, Nat Med 16:1370-1371, 2010; Zlokovic, Neuron 57:178-201, 2008). Experimental approaches to investigate the BBB in vitro are highly desirable to study the cerebral endothelium in health and disease. However, due to the complex interactions taking place within the NVU in vivo, it is difficult to mimic this interplay in vitro.Here, we describe a murine blood-brain barrier coculture model consisting of cortical organotypic slice cultures and brain endothelial cells that includes most of the cellular components of the NVU including neurons, astrocytes, and brain endothelial cells. This model allows the experimental analysis of several crucial BBB parameters such as transendothelial electrical resistance or tight junction protein localization by immunohistochemistry and live cell imaging of reactive oxygen species.


Subject(s)
Blood-Brain Barrier/cytology , Brain/blood supply , Endothelial Cells/physiology , Animals , Astrocytes/physiology , Cell Line , Coculture Techniques , Endothelium, Vascular/cytology , Mice , Mice, Inbred C57BL , Microvessels/cytology , Neurons/physiology , Reactive Oxygen Species/metabolism , Tissue Culture Techniques , Tissue Fixation
9.
PLoS One ; 8(12): e82823, 2013.
Article in English | MEDLINE | ID: mdl-24324834

ABSTRACT

Re-canalization of cerebral vessels in ischemic stroke is pivotal to rescue dysfunctional brain areas that are exposed to moderate hypoxia within the penumbra from irreversible cell death. Goal of the present study was to evaluate the effect of moderate hypoxia followed by reoxygenation (MHR) on the evolution of reactive oxygen species (ROS) and blood-brain barrier (BBB) integrity in brain endothelial cells (BEC). BBB integrity was assessed in BEC in vitro and in microvessels of the guinea pig whole brain in situ preparation. Probes were exposed to MHR (2 hours 67-70 mmHg O2, 3 hours reoxygenation, BEC) or towards occlusion of the arteria cerebri media (MCAO) with or without subsequent reperfusion in the whole brain preparation. In vitro BBB integrity was evaluated using trans-endothelial electrical resistance (TEER) and transwell permeability assays. ROS in BEC were evaluated using 2',7'-dichlorodihydrofluorescein diacetate (DCF), MitoSox and immunostaining for nitrotyrosine. Tight-junction protein (TJ) integrity in BEC, stainings for nitrotyrosine and FITC-albumin extravasation in the guinea pig brain preparation were assessed by confocal microscopy. Diphenyleneiodonium (DPI) was used to investigate NADPH oxidase dependent ROS evolution and its effect on BBB parameters in BEC. MHR impaired TJ proteins zonula occludens 1 (ZO-1) and claudin 5 (Cl5), decreased TEER, and significantly increased cytosolic ROS in BEC. These events were blocked by the NADPH oxidase inhibitor DPI. MCAO with or without subsequent reoxygenation resulted in extravasation of FITC-albumin and ROS generation in the penumbra region of the guinea pig brain preparation and confirmed BBB damage. BEC integrity may be impaired through ROS in MHR on the level of TJ and the BBB is also functionally impaired in moderate hypoxic conditions followed by reperfusion in a complex guinea pig brain preparation. These findings suggest that the BBB is susceptible towards MHR and that ROS play a key role in this process.


Subject(s)
Blood-Brain Barrier/metabolism , Hypoxia/metabolism , Oxidative Stress , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Animals , Blood-Brain Barrier/pathology , Cell Membrane/metabolism , Claudin-5/metabolism , Endothelial Cells/metabolism , Guinea Pigs , Microvessels/metabolism , Microvessels/pathology , Mitochondria/metabolism , Permeability , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Zonula Occludens-1 Protein/metabolism
10.
Cell Mol Life Sci ; 70(2): 309-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22940918

ABSTRACT

The in vivo roles of meprin metalloproteases in pathophysiological conditions remain elusive. Substrates define protease roles. Therefore, to identify natural substrates for human meprin α and ß we employed TAILS (terminal amine isotopic labeling of substrates), a proteomics approach that enriches for N-terminal peptides of proteins and cleavage fragments. Of the 151 new extracellular substrates we identified, it was notable that ADAM10 (a disintegrin and metalloprotease domain-containing protein 10)-the constitutive α-secretase-is activated by meprin ß through cleavage of the propeptide. To validate this cleavage event, we expressed recombinant proADAM10 and after preincubation with meprin ß, this resulted in significantly elevated ADAM10 activity. Cellular expression in murine primary fibroblasts confirmed activation. Other novel substrates including extracellular matrix proteins, growth factors and inhibitors were validated by western analyses and enzyme activity assays with Edman sequencing confirming the exact cleavage sites identified by TAILS. Cleavages in vivo were confirmed by comparing wild-type and meprin(-/-) mice. Our finding of cystatin C, elafin and fetuin-A as substrates and natural inhibitors for meprins reveal new mechanisms in the regulation of protease activity important for understanding pathophysiological processes.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Extracellular Matrix Proteins/metabolism , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , ADAM10 Protein , Amino Acid Sequence , Animals , Caco-2 Cells , Cell Line , Cystatin C/metabolism , Cytokines/metabolism , Elafin/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , alpha-2-HS-Glycoprotein/metabolism
11.
PLoS One ; 7(12): e50752, 2012.
Article in English | MEDLINE | ID: mdl-23251381

ABSTRACT

Disruption of the blood-brain barrier (BBB) results in cerebral edema formation, which is a major cause for high mortality after traumatic brain injury (TBI). As anesthetic care is mandatory in patients suffering from severe TBI it may be important to elucidate the effect of different anesthetics on cerebral edema formation. Tight junction proteins (TJ) such as zonula occludens-1 (ZO-1) and claudin-5 (cl5) play a central role for BBB stability. First, the influence of the volatile anesthetics sevoflurane and isoflurane on in-vitro BBB integrity was investigated by quantification of the electrical resistance (TEER) in murine brain endothelial monolayers and neurovascular co-cultures of the BBB. Secondly brain edema and TJ expression of ZO-1 and cl5 were measured in-vivo after exposure towards volatile anesthetics in native mice and after controlled cortical impact (CCI). In in-vitro endothelial monocultures, both anesthetics significantly reduced TEER within 24 hours after exposure. In BBB co-cultures mimicking the neurovascular unit (NVU) volatile anesthetics had no impact on TEER. In healthy mice, anesthesia did not influence brain water content and TJ expression, while 24 hours after CCI brain water content increased significantly stronger with isoflurane compared to sevoflurane. In line with the brain edema data, ZO-1 expression was significantly higher in sevoflurane compared to isoflurane exposed CCI animals. Immunohistochemical analyses revealed disruption of ZO-1 at the cerebrovascular level, while cl5 was less affected in the pericontusional area. The study demonstrates that anesthetics influence brain edema formation after experimental TBI. This effect may be attributed to modulation of BBB permeability by differential TJ protein expression. Therefore, selection of anesthetics may influence the barrier function and introduce a strong bias in experimental research on pathophysiology of BBB dysfunction. Future research is required to investigate adverse or beneficial effects of volatile anesthetics on patients at risk for cerebral edema.


Subject(s)
Anesthetics, Inhalation/pharmacology , Blood-Brain Barrier/drug effects , Brain Injuries/metabolism , Isoflurane/pharmacology , Methyl Ethers/pharmacology , Tight Junctions/drug effects , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Brain Edema/metabolism , Brain Edema/physiopathology , Brain Injuries/physiopathology , Cell Line , Claudin-5/metabolism , Coculture Techniques , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Male , Mice , Sevoflurane , Tight Junctions/metabolism , Zonula Occludens-1 Protein/metabolism
12.
Oncol Rep ; 27(5): 1573-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22294154

ABSTRACT

Novel strategies of cancer therapy combine irradiation and anti-angiogenic active compounds. However, little is known concerning the undesired cellular and molecular effects caused by this novel treatment concept. We used a mouse squamous cell carcinoma (SCC) xenotransplantation model to evaluate the potential undesired effects which compromise the success of this therapeutic combination. SCCs were subcutanously implanted in nude mice. Animals were treated with a fractionated irradiation scheme (5x4 Gy) alone or in combination with daily injections of anti-vascular endothelial growth factor (VEGF) antibodies. Controls remained untreated. Before and after treatment, resonance imaging (MRI), ultrasound and near-infrared spectrometry were used to evaluate tumor vessel integrity. Finally, tumors were explanted and VEGF, basic fibroblast growth factor (bFGF), vessel density, proliferation and apoptotic activity were analyzed by immunohistochemistry. Irradiation caused VEGF release and we found evidence for VEGF-mediated vessel protection. In the tumors derived from the combined treatment, blood volume was decreased, and apoptotic indices were increased. Remarkably, bFGF levels and proliferative indices were also increased. Combined irradiation/anti-VEGF treatment resulted in the desired VEGF depletion and increased tumor cell apoptosis. Nonetheless, bFGF and proliferation also increased, possibly suggesting a compensatory response. The application of additional targeted drugs may help develop more effective SCC treatments.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Fibroblast Growth Factor 2/metabolism , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Hemodynamics , Humans , Mice , Mice, Nude , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
13.
Biochemistry ; 49(39): 8599-607, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20806899

ABSTRACT

Meprin α and ß, zinc metalloproteinases, play significant roles in inflammation, including inflammatory bowel disease (IBD), possibly by activating cytokines, like interleukin 1ß, interleukin 18, or tumor growth factor α. Although a number of potential activators for meprins are known, no endogenous inhibitors have been identified. In this work, we analyzed the inhibitory potential of human plasma and identified bovine fetuin-A as an endogenous meprin inhibitor with a K(i) (inhibition constant) of 4.2 × 10(-5) M for meprin α and a K(i) of 1.1 × 10(-6) M meprin ß. This correlated with data obtained for a fetuin-A homologue from carp (nephrosin inhibitor) that revealed a potent meprin α and ß inhibition (residual activities of 27 and 22%, respectively) at a carp fetuin concentration of 1.5 × 10(-6) M. Human fetuin-A is a negative acute phase protein involved in inflammatory diseases, thus being a potential physiological regulator of meprin activity. We report kinetic studies of fetuin-A with the proteolytic enzymes astacin, LAST, LAST_MAM, trypsin, and chymotrypsin, indeed demonstrating that fetuin-A is a broad-range protease inhibitor. Fetuin-A inhibition of meprin α activity was 40 times weaker than that of meprin ß activity. Therefore, we tested cystatin C, a protein structurally closely related to fetuin-A. Indeed, cystatin C was an inhibitor for human meprin α (K(i) = 8.5 × 10(-6) M) but, interestingly, not for meprin ß. Thus, the identification of fetuin-A and cystatin C as endogenous proteolytic regulators of meprin activity broadens our understanding of the proteolytic network in plasma.


Subject(s)
Blood Proteins/metabolism , Cystatin C/metabolism , Metalloendopeptidases/antagonists & inhibitors , Plasma/metabolism , Amino Acid Sequence , Animals , Blood Proteins/isolation & purification , Carps , Cattle , Humans , Metalloendopeptidases/metabolism , Molecular Sequence Data , Sequence Alignment , alpha-2-HS-Glycoprotein
14.
Oral Oncol ; 46(9): 678-83, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20729134

ABSTRACT

Promoter hypermethylation of tumor suppressor genes is a common feature of primary cancer cells. However, at date the somatic epigenetic events that occur in head and neck squamous cell carcinoma (HNSCC) tumorigenesis are not yet been well defined. In the present study we analysed the methylation status of the gene hypermethylated in cancer-1 (hic1), a gene located on chromosome 17p13.3, a region frequently lost in HNSCC. We analysed 22 HNSCC samples and three cell lines using methylation specific PCR (MSP). We found hic1 methylated in 21 out of 22 samples and in all three cell lines. Treatment of the cell lines with the demethylating agent 5-Azacytidin (5-Aza) resulted in the demethylation of the hic1 promoter and reactivation of hic1 expression as determined by MSP, qPCR and Western blot. Functional analyses revealed decreased proliferative activity and colony forming ability of treated cells. In summary, we found in HNSCC hic1 regulated by promoter methylation. 5-Aza application resulted in the reexpression of hic1 and was followed by decreased aggressiveness of the cancer cells. Our data indicate that hic1 might be a player in HNSCC development and suggest further evaluation of 5-Aza for HNSCC treatment.


Subject(s)
Carcinoma, Squamous Cell/pathology , DNA Methylation/drug effects , Head and Neck Neoplasms/pathology , Kruppel-Like Transcription Factors/metabolism , Adult , Aged , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , DNA Methylation/physiology , Female , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Male , Middle Aged , Neoplasm Invasiveness , Promoter Regions, Genetic
15.
PLoS One ; 5(1): e8835, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-20098618

ABSTRACT

BACKGROUND: Meprin metalloproteases are thought to be involved in basic physiological functions such as cell proliferation and tissue differentiation. However, the specific functions of these enzymes are still ambiguous, although a variety of growth factors and structural proteins have been identified as meprin substrates. The discovery of meprins alpha(1), alpha(2) and beta in teleost fish provided the basis for uncovering their physiological functions by gene silencing in vivo. METHODOLOGY/PRINCIPAL FINDINGS: A Morpholino knockdown in zebrafish embryos targeting meprin alpha(1) and beta mRNA caused defects in general tissue differentiation. But meprin alpha(2) morphants were affected more specifically and showed severe failures in the formation of the vascular system provoking the hypothesis of a pro-angiogenic effect. The blood circulation was largely diminished resulting in erythrocyte accumulation. These phenotypes mimic a previously described VEGF-A morphant, revealing a possible role of meprin alpha in VEGF-A activation. Indeed, human recombinant meprin alpha processed the vascular endothelial growth factor-A (VEGF-A) specifically, revealing the same cleavage products detectable for VEGF from zebrafish whole lysate. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that meprin metalloproteases are important for cell differentiation and proliferation already during embryogenesis, predominantly by the activation of growth factors. Thus, we conclude that meprins play a significant role in VEGF-A processing, subsequently regulating angiogenesis. Therefore, meprin alpha might be a new therapeutic target in cardiovascular diseases or in tumor growth inhibition.


Subject(s)
Angiogenesis Inducing Agents , Gene Knockdown Techniques , Metalloendopeptidases/physiology , Morpholines/pharmacology , Zebrafish/embryology , Animals , Vascular Endothelial Growth Factor A/agonists
SELECTION OF CITATIONS
SEARCH DETAIL