Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Learn Mem ; 27(6): 222-235, 2020 06.
Article in English | MEDLINE | ID: mdl-32414940

ABSTRACT

Perineuronal nets (PNNs) are specialized extracellular matrix structures that surround subsets of neurons throughout the central nervous system (CNS). They are made up of chondroitin sulfate proteoglycans (CSPGs), hyaluronan, tenascin-R, and many other link proteins that together make up their rigid and lattice-like structure. Modulation of PNNs can alter synaptic plasticity and thereby affect learning, memory, and cognition. In the present study, we degraded PNNs in the medial prefrontal (mPFC) and posterior parietal (PPC) cortices of Long-Evans rats using the enzyme chondroitinase ABC (ChABC), which cleaves apart CSPGs. We then measured the consequences of PNN degradation on spatial working memory (WM) with a trial-unique, non-matching-to location (TUNL) automated touchscreen task. All rats were trained with a standard 6 sec delay and 20 sec inter-trial interval (ITI) and then tested under four different conditions: a 6 sec delay, a variable 2 or 6 sec delay, a 2 sec delay with a 1 sec ITI (interference condition), and a 20 sec delay. Rats that received mPFC ChABC treatment initially performed TUNL with higher accuracy, more selection trials completed, and fewer correction trials completed compared to controls in the 20 sec delay condition but did not perform differently from controls in any other condition. Rats that received PPC ChABC treatment did not perform significantly differently from controls in any condition. Posthumous immunohistochemistry confirmed an increase in CSPG degradation products (C4S stain) in the mPFC and PPC following ChABC infusions while WFA staining intensity and parvalbumin positive neuron number were decreased following mPFC, but not PPC, ChABC infusions. These findings suggest that PNNs in the mPFC play a subtle role in spatial WM, but PNNs in the PPC do not. Furthermore, it appears that PNNs in the mPFC are involved in adapting to a challenging novel delay, but that they do not play an essential role in spatial WM function.


Subject(s)
Chondroitin ABC Lyase/pharmacology , Chondroitin Sulfate Proteoglycans/drug effects , Extracellular Matrix/drug effects , Memory, Short-Term/drug effects , Parietal Lobe/drug effects , Prefrontal Cortex/drug effects , Psychomotor Performance/drug effects , Spatial Memory/drug effects , Animals , Behavior, Animal/drug effects , Male , Rats , Rats, Long-Evans , Time Factors
2.
Brain ; 143(1): 266-288, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31848580

ABSTRACT

Huntington's disease is associated with a reactive microglial response and consequent inflammation. To address the role of these cells in disease pathogenesis, we depleted microglia from R6/2 mice, a rapidly progressing model of Huntington's disease marked by behavioural impairment, mutant huntingtin (mHTT) accumulation, and early death, through colony-stimulating factor 1 receptor inhibition (CSF1Ri) with pexidartinib (PLX3397) for the duration of disease. Although we observed an interferon gene signature in addition to downregulated neuritogenic and synaptic gene pathways with disease, overt inflammation was not evident by microglial morphology or cytokine transcript levels in R6/2 mice. Nonetheless, CSF1Ri-induced microglial elimination reduced or prevented disease-related grip strength and object recognition deficits, mHTT accumulation, astrogliosis, and striatal volume loss, the latter of which was not associated with reductions in cell number but with the extracellular accumulation of chondroitin sulphate proteoglycans (CSPGs)-a primary component of glial scars. A concurrent loss of proteoglycan-containing perineuronal nets was also evident in R6/2 mice, and microglial elimination not only prevented this but also strikingly increased perineuronal nets in the brains of naïve littermates, suggesting a new role for microglia as homeostatic regulators of perineuronal net formation and integrity.


Subject(s)
Aminopyridines/pharmacology , Extracellular Matrix/drug effects , Huntingtin Protein/drug effects , Huntington Disease/metabolism , Microglia/drug effects , Neostriatum/drug effects , Pyrroles/pharmacology , Recognition, Psychology/drug effects , Animals , Astrocytes/drug effects , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Cytokines/drug effects , Cytokines/genetics , Disease Models, Animal , Down-Regulation , Extracellular Matrix/metabolism , Hand Strength , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/pathology , Huntington Disease/physiopathology , Inflammation , Mice , Mice, Transgenic , Neostriatum/pathology , Neurites/drug effects , RNA, Messenger/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Synapses/drug effects , Transcriptome
3.
J Control Release ; 297: 14-25, 2019 03 10.
Article in English | MEDLINE | ID: mdl-30690102

ABSTRACT

Central nervous system (CNS) injuries, such as stroke and spinal cord injuries, result in the formation of a proteoglycan-rich glial scar, which acts as a barrier to axonal regrowth and limits the regenerative capacity of the CNS. Chondroitinase ABC (ChABC) is a potent bacterial enzyme that degrades the chondroitin sulfate proteoglycan (CSPG) component of the glial scar and promotes tissue recovery; however, its use is significantly limited by its inherent instability at physiological temperatures. Here, we demonstrate that ChABC can be stabilized using site-directed mutagenesis and covalent modification with poly(ethylene glycol) chains (i.e. PEGylation). Rosetta protein structure modeling was used to screen >20,000 single point mutations, and four potentially stabilizing mutations were tested in vitro. One of the mutations, N1000G (asparagine ➔ glycine at residue 1000), significantly improved the long-term activity of the protein, doubling its functional half-life. PEGylation of this ChABC mutant inhibited unfolding and aggregation and resulted in prolonged bioactivity with a 10-fold increase in activity compared to the unmodified protein after two days. Local, affinity-controlled release of the modified protein (PEG-N1000G-ChABC) was achieved by expressing it as a fusion protein with Src homology 3 (SH3) and delivering the protein from a methylcellulose hydrogel modified with SH3 binding peptides. This affinity-based release strategy provided sustained PEG-N1000G-ChABC-SH3 release over several days in vitro. Direct implantation of the hydrogel delivery vehicle containing stabilized PEG-N1000G-ChABC-SH3 onto the rat brain cortex in a sub-acute model of stroke resulted in significantly reduced CSPG levels in the penumbra of 49% at 14 and 40% at 28 days post-injury compared to animals treated with the vehicle alone.


Subject(s)
Chondroitin ABC Lyase/chemistry , Chondroitin ABC Lyase/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Nanocapsules/chemistry , Nerve Regeneration/drug effects , Polyethylene Glycols/chemistry , Stroke/metabolism , Animals , Axons/metabolism , Brain/drug effects , Chondroitin ABC Lyase/genetics , Chondroitin Sulfate Proteoglycans/drug effects , Drug Liberation , Male , Mutagenesis/drug effects , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neuroglia/metabolism , Proteus vulgaris/enzymology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , src Homology Domains
4.
Glia ; 62(2): 259-71, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24311516

ABSTRACT

In utero ethanol exposure causes fetal alcohol spectrum disorders, associated with reduced brain plasticity; the mechanisms of these effects are not well understood, particularly with respect to glial involvement. Astrocytes release factors that modulate neurite outgrowth. We explored the hypothesis that ethanol inhibits neurite outgrowth by increasing the levels of inhibitory chondroitin sulfate proteoglycans (CSPGs) in astrocytes. Astrocyte treatment with ethanol inhibited the activity of arylsulfatase B (ARSB), the enzyme that removes sulfate groups from chondroitin-4-sulfate (C4S) and triggers the degradation of C4S, increased total sulfated glycosaminoglycans (GAGs), C4S, and neurocan core-protein content and inhibited neurite outgrowth in neurons cocultured with ethanol-treated astrocytes in vitro, effects reversed by treatment with recombinant ARSB. Ethanol also inhibited ARSB activity and increased sulfate GAG and neurocan levels in the developing hippocampus after in vivo ethanol exposure. ARSB silencing increased the levels of sulfated GAGs, C4S, and neurocan in astrocytes and inhibited neurite outgrowth in cocultured neurons, indicating that ARSB activity directly regulates C4S and affects neurocan expression. In summary, this study reports two major findings: ARSB modulates sulfated GAG and neurocan levels in astrocytes and astrocyte-mediated neurite outgrowth in cocultured neurons; and ethanol inhibits the activity of ARSB, increases sulfated GAG, C4S, and neurocan levels, and thereby inhibits astrocyte-mediated neurite outgrowth. An unscheduled increase in CSPGs in the developing brain may lead to altered brain connectivity and to premature decrease in neuronal plasticity and therefore represents a novel mechanism by which ethanol can exert its neurodevelopmental effects.


Subject(s)
Astrocytes/drug effects , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfates/metabolism , Ethanol/pharmacology , N-Acetylgalactosamine-4-Sulfatase/pharmacology , Neurites/drug effects , Animals , Astrocytes/metabolism , Brain/drug effects , Brain/growth & development , Brain/metabolism , Cells, Cultured , Glycosaminoglycans/metabolism , Neurites/metabolism , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
5.
J Natl Cancer Inst ; 102(19): 1496-512, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20852124

ABSTRACT

BACKGROUND: The cell surface proteoglycan, chondroitin sulfate proteoglycan 4 (CSPG4), is a potential target for monoclonal antibody (mAb)-based immunotherapy for many types of cancer. The lack of effective therapy for triple-negative breast cancer (TNBC) prompted us to examine whether CSPG4 is expressed in TNBC and can be targeted with CSPG4-specific mAb. METHODS: CSPG4 protein expression was assessed in 44 primary TNBC lesions, in TNBC cell lines HS578T, MDA-MB-231, MDA-MB-435, and SUM149, and in tumor cells in pleural effusions from 12 metastatic breast cancer patients. The effect of CSPG4-specific mAb 225.28 on growth, adhesion, and migration of TNBC cells was tested in vitro. The ability of mAb 225.28 to induce regression of tumor metastases (n = 7 mice) and to inhibit spontaneous metastasis and tumor recurrence (n = 12 mice per group) was tested in breast cancer models in mice. The mechanisms responsible for the antitumor effect of mAb 225.28 were also investigated in the cell lines and in the mouse models. All statistical tests were two-sided. RESULTS: CSPG4 protein was preferentially expressed in 32 of the 44 (72.7%) primary TNBC lesions tested, in TNBC cell lines, and in tumor cells in pleural effusions from 12 metastatic breast cancer patients. CSPG4-specific mAb 225.28 statistically significantly inhibited growth, adhesion, and migration of TNBC cells in vitro. mAb 225.28 induced 73.1% regression of tumor metastasis in a TNBC cell-derived experimental lung metastasis model (mAb 225.28 vs control, mean area of metastatic nodules = 44590.8 vs 165950.8 µm(2); difference of mean = 121360.0 µm(2), 95% confidence interval = 91010.7 to 151709.4 µm(2); P < .001). Additionally, mAb 225.28 statistically significantly reduced spontaneous lung metastases and tumor recurrences in an orthotopic xenograft mouse model. The mechanisms responsible for antitumor effect included increased apoptosis and reduced mitotic activity in tumor cells, decreased blood vessel density in the tumor microenvironment, and reduced activation of signaling pathways involved in cell survival, proliferation and metastasis. CONCLUSIONS: This study identified CSPG4 as a new target for TNBC. The antitumor activity of CSPG4-specific mAb was mediated by multiple mechanisms, including the inhibition of signaling pathways crucial for TNBC cell survival, proliferation, and metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/immunology , Immunotherapy/methods , Membrane Proteins/drug effects , Membrane Proteins/immunology , Aged , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/immunology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Confounding Factors, Epidemiologic , Disease Models, Animal , Female , Flow Cytometry , Humans , Immunoblotting , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Melanoma/drug therapy , Melanoma/immunology , Mice , Pleural Effusion/pathology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/immunology , Transplantation, Heterologous
6.
J Neuropathol Exp Neurol ; 69(3): 215-23, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20142769

ABSTRACT

Glial tumors exhibit a high morbidity and mortality because of their invasive nature. Matrix metalloproteinase 19 (MMP19) is a secreted protease that together with epilysin (MMP28) forms a structural subgroup of MMPs. We analyzed their expression by quantitative reverse transcription polymerase chain reaction, Western blot, and immunohistochemistry in tumor and normal control brain tissues and in glioblastoma (GB) cells and performed MMP19 silencing functional assays. Matrix metalloproteinase 28 was transcribed to the same extent in normal brain samples and gliomas but was undetectable in GB cell lines. In contrast, MMP19 was detected by immunohistochemistry in normal brain samples only in endothelial cells but was found at high levels in astrocytomas of different World Health Organization grades in situ and in GB cells in vitro. Matrix metalloproteinase 19 was upregulated in GB cells after exposure to proinflammatory cytokines. In Transwell invasion assays, MMP19-silenced cells migrated more slowly through laminin-, basal lamina-, and brevican-coated membranes than controls. Matrix metalloproteinase 19-silenced GB cells also migrated into brain tissue slices compared with control cells. Brevican, a brain-specific proteoglycan and major component of brain extracellular matrix, was degraded by recombinant human MMP19. Taken together, these results indicate that MMP19 is highly expressed in proliferating astrocytoma/glioma cells, and that its expression may facilitate their invasion through brain extracellular matrix components.


Subject(s)
Astrocytoma/enzymology , Astrocytoma/physiopathology , Brain Neoplasms/enzymology , Brain Neoplasms/physiopathology , Matrix Metalloproteinases, Secreted/metabolism , Neoplasm Invasiveness/physiopathology , Astrocytoma/pathology , Basement Membrane/metabolism , Basement Membrane/ultrastructure , Blotting, Western , Brain Neoplasms/pathology , Brevican , Cell Line, Tumor , Cell Movement/physiology , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Cytokines/metabolism , Cytokines/pharmacology , Endothelial Cells/enzymology , Extracellular Matrix , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunohistochemistry , Lectins, C-Type/drug effects , Lectins, C-Type/metabolism , Matrix Metalloproteinases, Secreted/genetics , Matrix Metalloproteinases, Secreted/pharmacology , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , RNA Interference , RNA, Messenger/analysis , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects , Up-Regulation/physiology
7.
J Neurosci ; 28(46): 11998-2009, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-19005065

ABSTRACT

Experimental therapeutics designed to enhance recovery from spinal cord injury (SCI) primarily focus on augmenting the growth of damaged axons by elevating their intrinsic growth potential and/or by nullifying the influence of inhibitory proteins present in the mature CNS. However, these strategies may also influence the wiring of intact pathways. The direct contribution of such effects to functional restoration after injury has been mooted, but as yet not been described. Here, we provide evidence to support the hypothesis that reorganization of intact spinal circuitry enhances function after SCI. Adult rats that underwent unilateral cervical spared-root lesion (rhizotomy of C5, C6, C8, and T1, sparing C7) exhibited profound sensory deficits for 4 weeks after injury. Delivery of a focal intraspinal injection of the chondroitin sulfate proteoglycan-degrading enzyme chondroitinase ABC (ChABC) was sufficient to restore sensory function after lesion. In vivo electrophysiological recordings confirm that behavioral recovery observed in ChABC-treated rats was consequent on reorganization of intact C7 primary afferent terminals and not regeneration of rhizotomized afferents back into the spinal cord within adjacent segments. These data confirm that intact spinal circuits have a profound influence on functional restoration after SCI. Furthermore, comprehensive understanding of these targets may lead to therapeutic interventions that can be spatially tailored to specific circuitry, thereby reducing unwanted maladaptive axon growth of distal pathways.


Subject(s)
Chondroitin ABC Lyase/pharmacology , Neuronal Plasticity/drug effects , Rhizotomy , Spinal Cord Injuries/drug therapy , Spinal Cord/drug effects , Spinal Nerve Roots/drug effects , Action Potentials/physiology , Afferent Pathways/drug effects , Afferent Pathways/enzymology , Afferent Pathways/injuries , Animals , Chondroitin ABC Lyase/metabolism , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Disease Models, Animal , Male , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neural Conduction/physiology , Neuronal Plasticity/physiology , Rats , Rats, Wistar , Recovery of Function/drug effects , Recovery of Function/physiology , Sensation Disorders/drug therapy , Sensation Disorders/etiology , Sensation Disorders/physiopathology , Sensory Receptor Cells/physiology , Spinal Cord/enzymology , Spinal Cord/physiopathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Spinal Nerve Roots/enzymology , Spinal Nerve Roots/injuries , Treatment Outcome
8.
Neuroscience ; 144(3): 865-77, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17141961

ABSTRACT

We investigated the effect of a single administration of recombinant human erythropoietin (rhEPO) on the preservation of the ventral white matter of rats at 4 weeks after contusive spinal cord injury (SCI), a time at which functional recovery is significantly improved in comparison to the controls [Gorio A, Necati Gokmen N, Erbayraktar S, Yilmaz O, Madaschi L, Cichetti C, Di Giulio AM, Enver Vardar E, Cerami A, Brines M (2002) Recombinant human erythropoietin counteracts secondary injury and markedly enhances neurological recovery from experimental spinal cord trauma. Proc Natl Acad Sci U S A 99:9450-9455; Gorio A, Madaschi L, Di Stefano B, Carelli S, Di Giulio AM, De Biasi S, Coleman T, Cerami A, Brines M (2005) Methylprednisolone neutralizes the beneficial effects of erythropoietin in experimental spinal cord injury. Proc Natl Acad Sci U S A 102:16379-16384]. Specifically, we examined, by morphological and cytochemical methods combined with light, confocal and electron microscopy, i) myelin preservation, ii) activation of adult oligodendrocyte progenitors (OPCs) identified for the expression of NG2 transmembrane proteoglycan, iii) changes in the amount of the chondroitin sulfate proteoglycans neurocan, versican and phosphacan and of their glycosaminoglycan component labeled with Wisteria floribunda lectin, and iv) ventral horn density of the serotonergic plexus as a marker of descending motor control axons. Injured rats received either saline or a single dose of rhEPO within 30 min after SCI. The results showed that the significant improvement of functional outcome observed in rhEPO-treated rats was associated with a better preservation of myelin in the ventral white matter. Moreover, the significant increase of both the number of NG2-positive OPCs and the labeling for Nogo-A, a marker of differentiated oligodendrocytes, suggested that rhEPO treatment could result in the generation of new myelinating oligodendrocytes. Sparing of fiber tracts in the ventral white matter was confirmed by the increased density of the serotonergic plexus around motor neurons. As for chondroitin sulfate proteoglycans, only phosphacan, increased in saline-treated rats, returned to normal levels in rhEPO group, probably reflecting a better maintenance of glial-axolemmal relationships along nerve fibers. In conclusion, this investigation expands previous studies supporting the pleiotropic neuroprotective effect of rhEPO on secondary degenerative response and its therapeutic potential for the treatment of SCI and confirms that the preservation of the ventral white matter, which contains descending motor pathways, may be critical for limiting functional deficit.


Subject(s)
Erythropoietin/pharmacology , Nerve Fibers, Myelinated/drug effects , Neuroprotective Agents/pharmacology , Spinal Cord Injuries/drug therapy , Spinal Cord/drug effects , Wallerian Degeneration/drug therapy , Animals , Antigens/drug effects , Antigens/metabolism , Axons/metabolism , Axons/ultrastructure , Cell Membrane/drug effects , Cell Membrane/metabolism , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Erythropoietin/therapeutic use , Male , Microscopy, Electron, Transmission , Myelin Proteins/drug effects , Myelin Proteins/metabolism , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neuroprotective Agents/therapeutic use , Nogo Proteins , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/ultrastructure , Proteoglycans/drug effects , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Serotonin/metabolism , Spinal Cord/metabolism , Spinal Cord/physiopathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/ultrastructure , Treatment Outcome , Wallerian Degeneration/physiopathology , Wallerian Degeneration/prevention & control
9.
Orthod Craniofac Res ; 9(3): 143-52, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16918679

ABSTRACT

OBJECTIVES: Previously, we demonstrated rapid changes in temporomandibular joint (TMJ) disk gene expression during monolayer expansion. This study's objective was to investigate the ability of pellet culture and growth factors to rescue TMJ disk gene expression changes. DESIGN: Temporomandibular joint disk cells were isolated from mature porcine tissue and passaged up to five times. At each passage, 300 000 cells were placed in a monolayer or pellet culture environment before being exposed to transforming growth factor-beta 3 (TGF-beta3) (5 ng/ml), TGF-beta1 (5 ng/ml), and insulin-like growth factor I (IGF-I) (10 ng/ml). OUTCOME MEASURE: After 24 h, gene expression was analyzed via reverse transcriptase-polymerase chain reaction (RT-PCR). RESULTS: Pelleting was detrimental to TMJ disk gene expression, marked by gene expression decreases in collagen type I (5.5-fold), aggrecan (1.4-fold), decorin (0.73-fold), and biglycan (0.73-fold) relative to monolayer cultures. IGF-I, TGF-beta1, and TGF-beta3 demonstrated limited ability to rescue TMJ disk gene expression in the pellet culture. In monolayer, TGF-beta3 and TGF-beta1 increased decorin and biglycan gene expression relative to passaged controls. Collagen type I expression, the TMJ disk's primary matrix constituent, was highest in TGF-beta3 cultures; however, differences were not statistically significant. CONCLUSION: These results indicate that pellet cultures are a poor choice for TMJ disk tissue engineering, and the effects of TGF-beta1, TGF-beta3, and IGF-I on TMJ disk gene expression are minimal relative to passaging and pelleting effects.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Temporomandibular Joint Disc/drug effects , Transforming Growth Factor beta/pharmacology , Aggrecans , Animals , Biglycan , Cell Culture Techniques , Cells, Cultured , Chondroitin Sulfate Proteoglycans/analysis , Chondroitin Sulfate Proteoglycans/drug effects , Collagen Type I/analysis , Collagen Type I/drug effects , Decorin , Extracellular Matrix Proteins/analysis , Extracellular Matrix Proteins/drug effects , Female , Gene Expression Regulation/drug effects , Lectins, C-Type/analysis , Lectins, C-Type/drug effects , Proteoglycans/analysis , Proteoglycans/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Swine , Temporomandibular Joint Disc/cytology , Tissue Engineering , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta1 , Transforming Growth Factor beta3
10.
Nephrol Dial Transplant ; 21(5): 1212-22, 2006 May.
Article in English | MEDLINE | ID: mdl-16421158

ABSTRACT

BACKGROUND: Heparin exerts beneficial effects in different experimental models of nephropathy, as observed by the preservation of the structural morphology of the kidney after heparin therapy. Here we investigate molecular and cellular events involved in the protective effects of heparin in the progression of renal disease after unilateral ureteral obstruction. METHODS: Thirty-six rats were divided into six groups: group C (control) was not subjected to any surgical manipulation; group S (sham) was subjected to surgical manipulation but without ureteral ligation; group UUO was subjected to ureteral obstruction and received no treatment; group UUO + S was subjected to ureteral obstruction and received saline subcutaneously (s.c.) once daily; group UUO + H was subjected to ureteral obstruction and received low molecular weight heparin (LMW-Hep; 4 mg/kg) s.c. once daily; and group C + H was not subjected to any surgical manipulation and received LMW-Hep (4 mg/kg) s.c. once daily. After 14 days, the content of collagen, fibronectin, total glycosaminoglycans (GAGS), chondroitin sulfate/dermatan sulfate proteoglycans (CS/DSPGs), transforming growth factor-beta (TGF-beta) and cellular infiltration were determined in the kidneys by immunohistochemical and biochemical techniques. RESULTS: Collagen, fibronectin, total GAGS, CS/DSPGs, TGF-beta and cellular infiltration increased significantly in group UUO. LMW-Hep treatment reduced collagen, fibronectin and TGF-beta, but induced an increase in the content of total GAGS, CS/DSPGs and macrophage infiltration in group UUO + H when compared with group UUO. CONCLUSIONS: LMW-Hep diminishes fibrosis in obstructed kidneys by downregulating the synthesis of collagen, fibronectin and TGF-beta. The mechanisms underlying the overproduction of CS/DSPGs and the increase in cellular infiltration upon LMW-Hep administration remain to be elucidated.


Subject(s)
Biomarkers/analysis , Heparin, Low-Molecular-Weight/pharmacology , Ureteral Obstruction/drug therapy , Animals , Biopsy, Needle , Cell Movement/drug effects , Chondroitin/metabolism , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Collagen/drug effects , Collagen/metabolism , Dermatan Sulfate/metabolism , Disease Models, Animal , Down-Regulation , Fibronectins/drug effects , Fibronectins/metabolism , Immunohistochemistry , Kidney/drug effects , Kidney/pathology , Macrophages/drug effects , Macrophages/physiology , Male , Probability , Random Allocation , Rats , Rats, Wistar , Reference Values , Sensitivity and Specificity , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism , Up-Regulation , Ureteral Obstruction/pathology
11.
Spine (Phila Pa 1976) ; 30(17): 1940-8, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16135983

ABSTRACT

STUDY DESIGN: This study examines changes in the production of extracellular matrix molecules as well as the induction of tissue degradation in in vitro formed nucleus pulposus (NP) tissues following incubation with tumor necrosis factor (TNF)alpha. OBJECTIVE: To characterize the response of NP cells to TNF-alpha, a proinflammatory cytokine present in herniated NP tissues. SUMMARY OF BACKGROUND DATA: TNF-alpha is a proinflammatory cytokine expressed by NP cells of degenerate intervertebral discs. It is implicated in the pain associated with disc herniation, although its role in intervertebral disc degeneration remains poorly understood. METHODS: In vitro formed NP tissues were treated with TNF-alpha (up to 50 ng/mL) over 48 hours. Tissues were assessed for histologic appearance, proteoglycan and collagen contents, as well as proteoglycan and collagen synthesis. Reverse transcriptase polymerase chain reaction was used to determine the effect of TNF-alpha on NP cell gene expression. Proteoglycan degradation was assessed by immunoblot analysis. RESULTS: At doses of 1-5 ng/mL, TNF-alpha induced multiple cellular responses, including: decreased expression of both aggrecan and type II collagen genes; decreases in the accumulation and overall synthesis of aggrecan and collagen; increased expression of MMP-1, MMP-3, MMP-13, ADAM-TS4, and ADAM-TS5; and induction of ADAM-TS dependent proteoglycan degradation. Within 48 hours, these cellular responses resulted in NP tissue with only 25% of its original proteoglycan content. CONCLUSIONS: Because low levels of TNF-alpha, comparable to those present physiologically, induced NP tissue degradation, this suggests that TNF-alpha may contribute to the degenerative changes that occur in disc disease.


Subject(s)
Extracellular Matrix/metabolism , Intervertebral Disc/metabolism , Tumor Necrosis Factor-alpha/physiology , ADAM Proteins/metabolism , Aggrecans , Animals , Cattle , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Collagen/metabolism , Dose-Response Relationship, Drug , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/drug effects , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression/drug effects , Humans , Intervertebral Disc/pathology , Lectins, C-Type/drug effects , Lectins, C-Type/metabolism , Matrix Metalloproteinases/metabolism , Proteoglycans/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Tissue Culture Techniques , Tissue Inhibitor of Metalloproteinases/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology
12.
J Neurosci ; 25(31): 7150-8, 2005 Aug 03.
Article in English | MEDLINE | ID: mdl-16079397

ABSTRACT

Chondroitin sulfate proteoglycans are major constituents of the extracellular matrix and form perineuronal nets. Information regarding the growth-inhibitory activity of these molecules after injury is rapidly expanding. However, less is known about their physiological role in the adult undamaged CNS. Here, we investigated the function of chondroitin sulfate proteoglycans in maintaining the proper structure of Purkinje axons in the cerebellum of adult rats. To this end, we examined the morphology and distribution of intracortical Purkinje neurites after intraparenchymal injection of chondroitinase ABC. Staining with the lectin Wisteria floribunda agglutinin or 2B6 antibodies showed that this treatment efficiently removed chondroitin sulfate proteoglycans from wide areas of the cerebellar cortex. In the same sites, there was a profuse outgrowth of terminal branches from the Purkinje infraganglionic plexus, which invaded the deeper regions of the granular layer. In contrast, myelinated axon segments were not affected and maintained their normal relationship with oligodendroglial sheaths. Purkinje axon sprouting was first evident at 4 d and increased further at 7 d after enzyme application. Within 42 d, the expression pattern of chondroitin sulfate proteoglycans gradually recovered, whereas axonal modifications progressively regressed. Our results show that, in the absence of injury or novel external stimuli, degradation of chondroitin sulfate proteoglycans is sufficient to induce Purkinje axon sprouting but not the formation of long-lasting synaptic contacts. Together with other growth-inhibitory molecules, such as myelin-associated proteins, chondroitin sulfate proteoglycans restrict structural plasticity of intact Purkinje axons to maintain normal wiring patterns in the adult cerebellar cortex.


Subject(s)
Axons/physiology , Chondroitin Sulfate Proteoglycans/metabolism , Nerve Regeneration/physiology , Purkinje Cells/physiology , Animals , Axons/ultrastructure , Cerebellum/cytology , Cerebellum/metabolism , Chondroitin ABC Lyase/pharmacology , Chondroitin Sulfate Proteoglycans/drug effects , Myelin Sheath/physiology , Purkinje Cells/ultrastructure , Rats , Rats, Wistar , Tissue Distribution
13.
Exp Neurol ; 182(1): 160-8, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12821386

ABSTRACT

We have previously demonstrated that enzymatic digestion of chondroitin sulfate proteoglycan (CSPG) at the scar promotes the axonal regrowth of Clarke's nucleus (CN) neurons into an implanted peripheral nerve graft after hemisection of the spinal cord. The present study examined whether degradation of CSPG using chondroitinase ABC promoted the regeneration of CN neurons through the scar into the rostral spinal cord in neonatal and adult rats. Following hemisection of the spinal cord at T11, either vehicle or chondroitinase ABC was applied onto the lesion site. The postoperative survival periods were 2 and 4 weeks. The regenerated CN neurons were retrogradely labeled by Fluoro-Gold injected at spinal cord level C7. In the sham group, there was no regeneration of injured CN neurons in both neonatal and adult rats. Treatment with 2.5 unit/ml chondroitinase ABC in neonates resulted in 11.8 and 8.3% of the injured CN neurons regenerated into the rostral spinal cord at 2 and 4 weeks, respectively. In adults, 9.4 and 12.3%, at 2 and 4 weeks, respectively, of the injured CN neurons regenerated their axons to the rostral spinal cord. The immunoreactivity for the carbohydrate epitope of CSPG was dramatically decreased around the lesion site after treatment with chondroitinase ABC compared to sham control in both neonatal and adult animals. Our results show that axonal regeneration in the spinal cord can be promoted by degradation of CSPG with chondroitinase ABC. This result further suggests that CSPG is inhibitory to the regeneration of neurons in the spinal cord after traumatic injury.


Subject(s)
Chondroitin ABC Lyase/pharmacology , Cicatrix/pathology , Nerve Regeneration/drug effects , Neurons/drug effects , Spinal Cord Injuries/drug therapy , Age Factors , Animals , Animals, Newborn , Axons/drug effects , Axons/physiology , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Chondroitin Sulfates/biosynthesis , Disease Models, Animal , Female , Immunohistochemistry , Neurons/pathology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology
14.
J Neurochem ; 83(3): 738-46, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12390535

ABSTRACT

Brevican is a neural-specific proteoglycan of the brain extracellular matrix, which is particularly abundant in the terminally differentiated CNS. It is expressed by neuronal and glial cells, and as a component of the perineuronal nets it decorates the surface of large neuronal somata and primary dendrites. One brevican isoform harbors a glycosylphosphatidylinositol anchor attachment site and, as shown by ethanolamine incorporation studies, is indeed glypiated in stably transfected HEK293 cells as well as in oligodendrocyte precursor Oli-neu cells. The major isoform is secreted into the extracellular space, although a significant amount appears to be tightly attached to the cell membrane, as it floats up in sucrose gradients. Flotation is sensitive to detergent treatment. Brevican is most prominent in the microsomal, light membrane and synaptosomal fractions of rat brain membrane preparations. The association with the particulate fraction is in part sensitive to chondroitinase ABC and phosphatidylinositol-specific phospholipase C treatment. Furthermore, brevican staining on the surface of hippocampal neurons in culture is diminished after hyaluronidase or chondroitinase ABC treatment. Taken together, this could provide a mechanism by which perineuronal nets are anchored on neuronal surfaces.


Subject(s)
Cell Membrane/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Blotting, Western , Brain Chemistry , Brevican , Cell Membrane/chemistry , Cells, Cultured , Centrifugation, Density Gradient , Chondroitin Sulfate Proteoglycans/chemistry , Chondroitin Sulfate Proteoglycans/drug effects , Ethanolamine/metabolism , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/pharmacology , Glycosylphosphatidylinositols/metabolism , Kidney/cytology , Kidney/metabolism , Lectins, C-Type , Microsomes/chemistry , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/drug effects , Neurons/chemistry , Neurons/ultrastructure , Oligodendroglia/cytology , Oligodendroglia/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Rats , Solubility/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Subcellular Fractions/chemistry , Synaptosomes/chemistry
16.
Glycoconj J ; 19(4-5): 275-85, 2002.
Article in English | MEDLINE | ID: mdl-12975606

ABSTRACT

Lumican and keratocan are members of the small leucine-rich proteoglycan (SLRP) family, and are the major keratan sulfate (KS) proteoglycans in corneal stroma. Both lumican and keratocan are essential for normal cornea morphogenesis during embryonic development and maintenance of corneal topography in adults. This is attributed to their bi-functional characteristic (protein moiety binding collagen fibrils to regulate collagen fibril diameters, and highly charged glycosaminoglycan (GAG) chains extending out to regulate interfibrillar spacings) that contributes to their regulatory role in extracellular matrix assembly. The absence of lumican leads to formation of cloudy corneas in homozygous knockout mice due to altered collagenous matrix characterized by larger fibril diameters and disorganized fibril spacing. In contrast, keratocan knockout mice exhibit thin but clear cornea with insignificant alteration of stromal collaegenous matrix. Mutations of keratocan cause cornea plana in human, which is often associated with glaucoma. These observations suggest that lumican and keratocan have different roles in regulating formation of stromal extracellular matrix. Experimental evidence indicates that lumican may have additional biological functions, such as modulation of cell migration and epithelium-mesenchyme transition in wound healing and tumorgenesis, besides regulating collagen fibrillogenesis.


Subject(s)
Chondroitin Sulfate Proteoglycans/physiology , Cornea/physiology , Keratan Sulfate/physiology , Proteoglycans/physiology , Amino Acid Sequence , Animals , Base Sequence , Chondroitin Sulfate Proteoglycans/chemistry , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/genetics , Corneal Opacity/genetics , Corneal Opacity/physiopathology , DNA/genetics , Humans , Keratan Sulfate/chemistry , Keratan Sulfate/genetics , Lumican , Mice , Mice, Knockout , Molecular Sequence Data , Molecular Structure , Phenotype , Promoter Regions, Genetic , Proteoglycans/chemistry , Proteoglycans/deficiency , Proteoglycans/genetics , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
17.
J Periodontol ; 72(3): 341-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11327061

ABSTRACT

BACKGROUND: Periodontal wound healing and regeneration require that new matrix be synthesized, creating an environment into which cells can migrate. One agent which has been described as promoting periodontal regeneration is an enamel matrix protein derivative (EMD). Since no specific growth factors have been identified in EMD preparations, it is postulated that EMD acts as a matrix enhancement factor. This study was designed to investigate the effect of EMD in vitro on matrix synthesis by cultured periodontal fibroblasts. METHODS: The matrix response of the cells was evaluated by determination of the total proteoglycan synthesis, glycosaminoglycan profile, and hyaluronan synthesis by the uptake of radiolabeled precursors. The response of the individual proteoglycans, versican, decorin, and biglycan were examined at the mRNA level by Northern blot analysis. Hyaluronan synthesis was probed by identifying the isotypes of hyaluronan synthase (HAS) expressed in periodontal fibroblasts as HAS-2 and HAS-3 and the effect of EMD on the levels of mRNA for each enzyme was monitored by reverse transcription polymerase chain reaction (RTPCR). Comparisons were made between gingival fibroblast (GF) cells and periodontal ligament (PDLF) cells. RESULTS: EMD was found to significantly affect the synthesis of the mRNAs for the matrix proteoglycans versican, biglycan, and decorin, producing a response similar to, but potentially greater than, mitogenic cytokines. EMD also stimulated hyaluronan synthesis in both GF and PDLF cells. Although mRNA for HAS-2 was elevated in GF after exposure to EMD, the PDLF did not show a similar response. Therefore, the point at which the stimulation of hyaluronan becomes effective may not be at the level of stimulation of the mRNA for hyaluronan synthase, but, rather, at a later point in the pathway of regulation of hyaluronan synthesis. In all cases, GF cells appeared to be more responsive to EMD than PDLF cells in vitro. CONCLUSIONS: EMD has the potential to significantly modulate matrix synthesis in a manner consistent with early regenerative events.


Subject(s)
Dental Enamel Proteins/pharmacology , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Gingiva/drug effects , Periodontal Ligament/drug effects , Analysis of Variance , Biglycan , Blotting, Northern , Cell Movement/drug effects , Cells, Cultured , Chondroitin Sulfate Proteoglycans/biosynthesis , Chondroitin Sulfate Proteoglycans/drug effects , Decorin , Extracellular Matrix/metabolism , Extracellular Matrix Proteins , Fibroblasts/metabolism , Gingiva/cytology , Gingiva/metabolism , Glycosaminoglycans/biosynthesis , Humans , Hyaluronic Acid/biosynthesis , Lectins/biosynthesis , Lectins/drug effects , Lectins, C-Type , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Proteoglycans/biosynthesis , Proteoglycans/drug effects , RNA, Messenger/analysis , Regeneration , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/antagonists & inhibitors , Versicans , Wound Healing
18.
Croat Med J ; 40(4): 528-32, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10554355

ABSTRACT

AIM: To analyze the influence of the premature termination codon on mRNA transport and stability METHODS: Chondrocyte mRNA was isolated from homozygous and heterozygous nanomelic 17-days old embryos and examined by RT-PCR analysis. To analyze aggrecan mRNA stability, mRNA synthesis was inhibited with DRB [5,6 dichloro-1-(-D-ribofuranosyl benzimidazole)], a specific inhibitor of RNA polymerase II. Visualization of the aggrecan alleles was performed by in situ hybridization. RESULTS: The level of mutant aggrecan mRNA within the nucleus was equal to that of the control, but no mutant mRNA was observed in the cytoplasm. RT-PCR revealed that the mutant transcript was only detectable in the nucleus, compared with house-keeping glyceraldehyde-3-phosphate dehydrogenase (GAPDH) gene or collagen type II. A restriction site induced by premature termination codon TAA allowed the distinction of normal and mutant transcripts in chondrocytes derived from embryos heterozygous for the nanomelic mutation. After the treatment with DRB, identical decay rates were demonstrated for both transcripts within the heterozygous nucleus. In situ hybridization showed no abnormal mRNA accumulation. CONCLUSION: This is the first evidence suggesting that the transcript of the mRNA with the premature termination codon within an exon does exit the nucleus.


Subject(s)
Cartilage Diseases/genetics , Cartilage/metabolism , Chondroitin Sulfate Proteoglycans/genetics , Codon, Terminator/genetics , Extracellular Matrix Proteins , Protein Biosynthesis/genetics , Proteoglycans/genetics , RNA, Messenger/metabolism , Aggrecans , Animals , Cartilage/embryology , Cartilage Diseases/drug therapy , Cartilage Diseases/metabolism , Cell Culture Techniques , Chick Embryo , Chondrocytes/cytology , Chondrocytes/metabolism , Chondroitin Sulfate Proteoglycans/drug effects , Dichlororibofuranosylbenzimidazole/pharmacology , Genotype , Lectins, C-Type , Mutation , Nucleic Acid Synthesis Inhibitors/pharmacology , Proteoglycans/drug effects , RNA Polymerase II/antagonists & inhibitors , RNA, Messenger/drug effects , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
Exp Neurol ; 160(1): 51-65, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10630190

ABSTRACT

Extrinsic factors appear to contribute to the lack of regeneration in the injured adult spinal cord. It is likely that these extrinsic factors include a group of putative growth inhibitory molecules known as chondroitin sulfate proteoglycans (CSPGs). The aims of this study were to determine: (1) the consequences of spinal cord contusion injury on CSPG expression, (2) if CSPGs can be degraded in vivo by exogenous enzyme application, and (3) the effects of intraspinal transplantation on the expression of CSPGs. Chondroitin 6-sulfate proteoglycan immunoreactivity (CSPG-IR) dramatically increased following spinal cord contusion injury both at and adjacent to the injury site compared to normal controls (no surgical procedure) and laminectomy-only controls by 4 days postinjury. The dramatic increase in CSPG-IR persisted around the lesion and in the dorsal one-half to two-thirds of the spinal cord for at least 40 days postinjury. Glial fibrillary acidic protein (GFAP)-IR patterns were similarly intensified and spatially restricted as CSPG-IR patterns. These results suggest that: (1) CSPGs may contribute to the lack of regeneration following spinal cord injury and (2) astrocytes may contribute to the production of CSPGs. In addition, our results show that CSPGs could be cleaved in vivo with exogenous chondroitinase ABC application. This demonstration of cleavage may the basis for a model to directly assess CSPGs' role in growth inhibition in vivo (studies in progress) and hold potential as a therapeutic approach to enhance growth. Interestingly, the robust, injury-induced CSPG-IR patterns were not altered by intraspinal grafts of fetal spinal cord. The CSPG expression profile in the host spinal cord was similar to time-matched contusion-only animals. This was also true of GFAP-IR patterns. Furthermore, the fetal spinal cord tissue, which was generally CSPG negative at the time of transplantation, developed robust CSPG expression by 30 days posttransplantation. This increase in CSPG expression in the graft was paired with a moderate increase in GFAP-IR. CSPG-IR patterns suggest that these molecules may contribute to the limited regeneration seen following intraspinal transplantation. In addition, it suggests that the growth permissiveness of the graft may change overtime as CSPG expression develops within the graft. These correlations in the injured and transplanted spinal cord support CSPGs' putative growth inhibitory effect in the adult spinal cord.


Subject(s)
Chondroitin Sulfate Proteoglycans/biosynthesis , Contusions/metabolism , Fetal Tissue Transplantation , Growth Inhibitors/biosynthesis , Nerve Regeneration , Spinal Cord Injuries/metabolism , Spinal Cord/transplantation , Animals , Astrocytes/physiology , Chondroitin ABC Lyase/pharmacology , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/physiology , Glial Fibrillary Acidic Protein/analysis , Graft Survival , Growth Inhibitors/physiology , Rats , Thoracic Vertebrae
20.
Anat Embryol (Berl) ; 195(1): 71-8, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9006717

ABSTRACT

During murine embryogenesis, uptake sites for the neurotransmitter serotonin (5-HT) are transiently expressed in craniofacial epithelial structures. Based on malformations produced in cultured mouse embryos exposed to uptake inhibitors or receptor ligands, we have proposed that 5-HT acts as a dose-dependent morphogenetic signal during critical periods of craniofacial development. Several 5-HT receptor subtypes are co-distributed with tenascin and the calcium binding protein S-100 beta in developing craniofacial mesenchyme. Since these molecules are thought to be important for craniofacial development, their regulation by 5-HT could mediate some of its morphogenetic actions. Mandibular mesenchyme cells, from E12 mouse embryos (plug day = E1), grown in micromass cultures were used as an in vitro model to investigate whether 5-HT regulates expression of these molecules. Immunocytochemistry revealed expression of S-100 beta, tenascin, cartilage proteoglycan core protein (a component of the cartilage matrix) and a variety of 5-HT receptors in these cultures. To block the actions of 5-HT (from serum in the culture medium), cultures were exposed to one of these selective 5-HT receptor antagonists and effects on expression were investigated using quantitative immunobinding and in situ hybridization assays. These antagonists differentially regulated expression of cartilage core protein, S-100 beta and tenascin. Antagonism of 5-HT3 receptors by Zofran or 5-HT1A receptors by NAN-190 reduced the amount of core protein, whereas antagonism of 5-HT2A-C receptors by mianserin had no significant effect. All three antagonists stimulated levels of tenascin mRNA and protein. Expression of S-100 beta mRNA and protein was inhibited by Zofran and stimulated by mianserin, whereas NAN-190 had no significant effect. The differential effects of antagonists suggest that in vivo, 5-HT could: (1) promote expression of cartilage core protein by activation of 5-HT3 or 5-HT1A receptors, (2) inhibit production of tenascin by activation of multiple receptors, (3) promote or inhibit synthesis of S-100 beta by activation of 5-HT3 or 5-HT2 receptors, respectively. These actions may be important components of the morphogenetic functions of 5-HT during craniofacial development.


Subject(s)
Extracellular Matrix Proteins , Gene Expression Regulation, Developmental/drug effects , Mandible/embryology , Mesoderm/drug effects , Serotonin Antagonists/pharmacology , Aggrecans , Animals , Calcium-Binding Proteins/drug effects , Calcium-Binding Proteins/metabolism , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Culture Techniques , Female , Lectins, C-Type , Mandible/drug effects , Mandible/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mianserin/pharmacology , Mice , Mice, Inbred ICR , Nerve Growth Factors , Ondansetron/pharmacology , Piperazines/pharmacology , Pregnancy , Proteoglycans/drug effects , Proteoglycans/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/drug effects , S100 Proteins/metabolism , Tenascin/drug effects , Tenascin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL