Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Complement Ther Med ; 52: 102482, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32951732

ABSTRACT

Sickle cell disease (SCD) is a hematologic disorder with complex pathophysiology that includes chronic hemolysis, vaso-occlusion and inflammation. Increased leukocyte-erythrocyte-endothelial interactions, due to upregulated expression of adhesion molecules and activated endothelium, are thought to play a primary role in initiation and progression of SCD vaso-occlusive crisis and end-organ damage. Several new pathophysiology-based therapeutic options for SCD are being developed, chiefly targeting the inflammatory pathways. Omega-3 fatty acids are polyunsaturated fatty acids that are known to have effects on diverse physiological processes. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are the principal biologically active omega-3 fatty acids. The therapeutic effects of DHA and EPA on chronic inflammatory disorders and cardiovascular diseases are well recognized. The therapeutic effects of omega-3 fatty acids are attributed to their anti-inflammatory and anti-thrombotic eicosanoids, and the novel class of EPA and DHA derived lipid mediators: resolvins, protectins and maresins. Blood cell membranes of patients with SCD have abnormal fatty acids composition characterized by high ratio of pro-inflammatory arachidonic acid (AA) to anti-inflammatory DHA and EPA (high omega-6/omega-3 ratio). In addition, experimental and clinical studies provide evidence that treatment with DHA does confer improvement in rheological properties of sickle RBC, inflammation and hemolysis. The clinical studies have shown improvements in VOC rate, markers of inflammation, adhesion, and hemolysis. In toto, the results of studies on the therapeutic effects of omega-3 fatty acids in SCD provide good body of evidence that omega-3 fatty acids could be a safe and effective treatment for SCD.


Subject(s)
Anemia, Sickle Cell/drug therapy , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Humans
2.
Article in English | MEDLINE | ID: mdl-32447175

ABSTRACT

Sickle cell disease (SCD) is one of the most common inherited blood disorder among African Americans affecting 70,000-100,000 individuals in the United States. It is characterized by abnormal hemoglobin (HbS) which develops into severe hemolytic anemia and vaso-occlusive crisis. Therefore, patients with SCD suffer from a chronic state of inflammation, which is responsible for multiple organ damage, ischemic attacks, and premature death. Another major hallmark of SCD patients is the abnormally low levels of omega-3 fatty acids, especially docosahexaenoic acid (DHA) in their red blood cell membranes. Treatment with DHA can reduce red blood cell adhesion and enhance cerebral blood flow, thus, our main goal is to investigate the effect of SC411, which is a novel, highly purified DHA ethyl ester formulation with a proprietary delivery platform in SCD. Utilizing a transgenic mouse model of SCD (HbSS-Townes) and recurrent hypoxic challenges (10%O2, 0.5% CO2 and balance N2 for 3 h) to mimic ischemic-like conditions, our data suggest that SC411 can elevate blood DHA and eicosapentaenoic acid (EPA) levels after 8 weeks of treatment. SC411 can also decrease arachidonic acid (AA) and sickling of red blood cells. In addition, SC411-treated SCD mice showed presented with cerebral blood flow, alleviated neuroinflammation, and revived working memory which ultimately enhanced overall survival. In summary, this study suggests that treatment with SC411 improves cellular and functional outcomes in SCD mice. This finding may provide novel therapeutic opportunities in the treatment against ischemic injury elicited by SCD.


Subject(s)
Anemia, Sickle Cell/drug therapy , Docosahexaenoic Acids/chemistry , Esters/administration & dosage , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/psychology , Animals , Arachidonic Acid/blood , Cerebrovascular Circulation , Disease Models, Animal , Docosahexaenoic Acids/blood , Esters/chemistry , Esters/pharmacology , Humans , Male , Memory, Short-Term/drug effects , Mice , Mice, Transgenic , Survival Analysis , Treatment Outcome
3.
J Drug Deliv ; 2019: 1957360, 2019.
Article in English | MEDLINE | ID: mdl-31360551

ABSTRACT

Despite recent advances, the drug development process continues to face significant challenges to efficiently improve the poor solubility of active pharmaceutical ingredients (API) in aqueous media or to improve the bioavailability of lipid-based formulations. The inherent high intra- and interindividual variability of absorption of oral lipophilic drug leads to inconsistent and unpredictable bioavailability and magnitude of the therapeutic effect. For this reason, the development of lipid-based drugs remains a challenging endeavour with a high risk of failure. Therefore, effective strategies to assure a predictable, consistent, and reproducible bioavailability and therapeutic effect for lipid-based medications are needed. Different solutions to address this problem have been broadly studied, including the approaches of particle size reduction, prodrugs, salt forms, cocrystals, solid amorphous forms, cyclodextrin clathrates, and lipid-based drug delivery systems such as self-emulsifying systems and liposomes. Here, we provide a brief description of the current strategies commonly employed to increase the bioavailability of lipophilic drugs and present Advanced Lipid Technologies® (ALT®), a combination of different surfactants that has been demonstrated to improve the absorption of omega-3 fatty acids under various physiological and pathological states.

4.
J Clin Lipidol ; 11(2): 394-405, 2017.
Article in English | MEDLINE | ID: mdl-28502496

ABSTRACT

BACKGROUND: The absorption of eicosapentaenoic acid (EPA; 20:5n-3) and docosahexaenoic acid (DHA; 22:6n-3) omega-3-acid ethyl esters (EEs) is influenced by food. There is a need for a formulation of EE that is less impacted by food effect. SC401 is a novel Advanced Lipid Technologies-based formulation of EPA-EE and DHA-EE. In the presence of an aqueous medium, Advanced Lipid Technologies forms stable micelles in situ independent of bile salt secretion. This effect is hypothesized to improve EPA-EE and DHA-EE bioavailability while it helps mitigate the food effect associated with their consumption. OBJECTIVE: The aim of the article was to assess the effect of food on the bioavailability of DHA and EPA after a single oral dose of 1530 mg omega-3 fatty acids EE (SC401) in 24 healthy subjects under fasted and low-fat (9% of total calories from fat) and high-fat (50% of total calories from fat) meal conditions. METHODS: This was a randomized, open-label, single-dose, 3-period, 3-way crossover study. Blood samples for pharmacokinetic analyses were taken at predose and at 0.5, 1, 2, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 8, 10, 12 and 24 hours postdose. To assess the safety of the intervention, active monitoring of adverse events, physical examinations, vital signs, clinical laboratory assessments (chemistry, hematology, and urinalysis), and 12-lead electrocardiograms were conducted. RESULTS: SC401 showed high bioavailability of both EPA and DHA in fasted, low-fat meal, and high-fat meal conditions. No differences were found in SC401 DHA AUC0-t (t = 24 hours) among the 3 conditions (91.69% high-fat/fasted, 97.12% low-fat/fasted, and 105.92% low-fat/high-fat; P > .05 in all cases). In contrast, SC401 EPA AUC0-t was affected by food intake (179.06% high-fat/fasted, P < .0001; 150.05% low-fat/fasted, P < .0001) and the amount of fat taken with SC401 (83.80% low-fat/high-fat; P = .0009). SC401 was safe and well tolerated. CONCLUSIONS: A single dose of SC401 resulted in high levels of EPA and DHA total lipids in plasma in fasting and fed conditions. SC401 overcame the food effect for DHA and partially ameliorated it for EPA. SC401 represents a convenient option for treatment of severe hypertriglyceridemia, especially for patients under a restricted intake of dietary fat.


Subject(s)
Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/pharmacokinetics , Drug Compounding , Eicosapentaenoic Acid/chemistry , Eicosapentaenoic Acid/pharmacokinetics , Esters/chemistry , Food , Adult , Biological Availability , Diet, High-Fat , Docosahexaenoic Acids/adverse effects , Eicosapentaenoic Acid/adverse effects , Fasting , Female , Humans , Male
5.
Clin Ther ; 39(3): 581-591, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28189364

ABSTRACT

PURPOSE: The US Food and Drug Administration has approved several highly purified ω-3 fatty acid prescription drugs for the treatment of severe hypertriglyceridemia. These differ in the amounts and forms of docosahexaenoic acid (DHA) and/or eicosapentaenoic acid (EPA). This study compared the bioavailability of SC401 (1530 mg EPA-ethyl esters [EEs] and DHA-EEs plus Advanced Lipid Technologies⁎ [ALT†], a proprietary lipid-delivery platform to improve absorption), with. Lovaza‡ (3600 mg ω-3, primarily EPA-EEs and DHA-EEs) under low-fat feeding conditions. METHODS: This was a Phase I, randomized, open-label, single-dose, 2-way crossover study in healthy participants housed from day -3 to day 2 in each treatment period. Blood samples for pharmacokinetic measurements were collected before and after dosing, and safety profile and tolerability were assessed. FINDINGS: In unadjusted analyses, SC401 had 5% lower Cmax and approximately the same AUC0-last of EPA + DHA total lipids compared with Lovaza. When adjusted for baseline, SC401 had ~6% higher Cmax and 18% higher AUC0-last for EPA + DHA total lipids, and dose- and baseline-adjusted analyses found that SC401 had ~149% higher Cmax and 178% higher AUC0-last than Lovaza for EPA + DHA total lipids. The Tmax was also substantially longer with Lovaza (~10 hours) than with SC401 (~6 hours). IMPLICATIONS: These results indicate that SC401, an ω-3 acid EE formulation containing ALT† achieved high bioavailability of EPA and DHA, at a lower dose (1530 mg) than Lovaza (3600 mg), under low-fat feeding conditions.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Fatty Acids, Omega-3/administration & dosage , Hypolipidemic Agents/administration & dosage , Adult , Biological Availability , Chemistry, Pharmaceutical , Cross-Over Studies , Docosahexaenoic Acids/pharmacokinetics , Drug Combinations , Eicosapentaenoic Acid/pharmacokinetics , Fatty Acids, Omega-3/therapeutic use , Female , Humans , Hypertriglyceridemia/drug therapy , Hypolipidemic Agents/therapeutic use , Male , Middle Aged , Young Adult
6.
Nat Biotechnol ; 30(5): 453-9, 2012 Mar 25.
Article in English | MEDLINE | ID: mdl-22446693

ABSTRACT

The ability to specifically upregulate genes in vivo holds great therapeutic promise. Here we show that inhibition or degradation of natural antisense transcripts (NATs) by single-stranded oligonucleotides or siRNAs can transiently and reversibly upregulate locus-specific gene expression. Brain-derived neurotrophic factor (BDNF) is normally repressed by a conserved noncoding antisense RNA transcript, BDNF-AS. Inhibition of this transcript upregulates BDNF mRNA by two- to sevenfold, alters chromatin marks at the BDNF locus, leads to increased protein levels and induces neuronal outgrowth and differentiation both in vitro and in vivo. We also show that inhibition of NATs leads to increases in glial-derived neurotrophic factor (GDNF) and ephrin receptor B2 (EPHB2) mRNA. Our data suggest that pharmacological approaches targeting NATs can confer locus-specific gene upregulation effects.


Subject(s)
Oligonucleotides, Antisense/antagonists & inhibitors , Up-Regulation , Animals , Cell Line , Chromatin/chemistry , Chromatin/metabolism , Exons , Gene Expression Profiling , Genomics , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , HEK293 Cells , Humans , Mice , Models, Genetic , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptor, EphB2/biosynthesis , Sequence Analysis, DNA , Transcription, Genetic
7.
Int J Alzheimers Dis ; 2011: 929042, 2011.
Article in English | MEDLINE | ID: mdl-21785702

ABSTRACT

Background. Alzheimer's disease (AD) is a devastating neurological disorder and the main cause of dementia in the elderly population worldwide. Adult neurogenesis appears to be upregulated very early in AD pathogenesis in response to some specific aggregates of beta-amyloid (Aß) peptides, exhausting the neuronal stem cell pools in the brain. Previously, we characterized a conserved nonprotein-coding antisense transcript for ß-secretase-1 (BACE1), a critical enzyme in AD pathophysiology. We showed that the BACE1-antisense transcript (BACE1-AS) is markedly upregulated in brain samples from AD patients and promotes the stability of the (sense) BACE1 transcript. In the current paper, we examine the relationship between BACE1, BACE1-AS, adult neurogenesis markers, and amyloid plaque formation in amyloid precursor protein (APP) transgenic mice (Tg-19959) of various ages. Results. Consistent with previous publications in other APP overexpressing mouse models, we found adult neurogenesis markers to be noticeably upregulated in Tg-19959 mice very early in the development of the disease. Knockdown of either one of BACE1 or BACE1-AS transcripts by continuous infusion of locked nucleic acid- (LNA-) modified siRNAs into the third ventricle over the period of two weeks caused concordant downregulation of both transcripts in Tg-19959 mice. Downregulation of BACE1 mRNA was followed by reduction of BACE1 protein and insoluble Aß. Modulation of BACE1 and BACE1-AS transcripts also altered oligomeric Aß aggregation pattern, which was in turn associated with an increase in neurogenesis markers at the RNA and protein level. Conclusion. We found alterations in the RNA and protein concentrations of several adult neurogenesis markers, as well as non-protein-coding BACE1-AS transcripts, in parallel with the course of ß-amyloid synthesis and aggregation in the brain of Tg15999 mice. In addition, by knocking down BACE1 or BACE1-AS (thereby reducing Aß production and plaque deposition), we were able to modulate expression of these neurogenesis markers. Our findings suggest a distortion of adult neurogenesis that is associated with Aß production very early in amyloid pathogenesis. We believe that these alterations, at the molecular level, could prove useful as novel therapeutic targets and/or as early biomarkers of AD.

8.
J Alzheimers Dis ; 20(2): 395-408, 2010.
Article in English | MEDLINE | ID: mdl-20164555

ABSTRACT

Alzheimer's disease (AD) is a devastating age-related neurodegenerative disorder characterized by progressive impairment of cognition and short-term memory loss. The deposition of amyloid-beta (Abeta) 1-42 into senile plaques is an established feature of AD neuropathology. Controversy still exists about the amyloid pathway as the initiating mechanism or a mere consequence of the events leading to AD. Nevertheless, Abeta toxicity has been probed in vitro and in vivo and increased production or decreased clearance of Abeta peptides are reported to play a major role in the development of AD. Treatment of neural stem cells with Abeta in vitro induces neuronal differentiation. Increased neurogenesis has been also described in AD patients as well as in amyloid-beta protein precursor (AbetaPP) transgenic mice. Adult neurogenesis is greatly enhanced in young AbetaPP transgenic mice, before other AD-liked pathologies, and reduced in older animals. This increased neurogenesis at young ages might be the first pathology related to AD, which is detectable long before other harmful manifestation of the disease. Therefore, understanding the mechanisms of Abeta-induced neurogenesis will reveal insights into the pathogenesis of AD and may prove useful as an early AD biomarker.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/physiopathology , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/drug effects , Neurogenesis/physiology , Peptide Fragments , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Humans , Neurons/physiology , Peptide Fragments/metabolism , Stem Cells/physiology
9.
Proc Natl Acad Sci U S A ; 106(9): 3507-12, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19196972

ABSTRACT

N-methyl-D-aspartate (NMDA) glutamate receptors are regulators of fast neurotransmission and synaptic plasticity in the brain. Disruption of NMDA-mediated glutamate signaling has been linked to behavioral deficits displayed in psychiatric disorders such as schizophrenia. Recently, noncoding RNA molecules such as microRNAs (miRNAs) have emerged as critical regulators of neuronal functions. Here we show that pharmacological (dizocilpine) or genetic (NR1 hypomorphism) disruption of NMDA receptor signaling reduces levels of a brain-specific miRNA, miR-219, in the prefrontal cortex (PFC) of mice. Consistent with a role for miR-219 in NMDA receptor signaling, we identify calcium/calmodulin-dependent protein kinase II gamma subunit (CaMKIIgamma), a component of the NMDA receptor signaling cascade, as a target of miR-219. In vivo inhibition of miR-219 by specific antimiR in the murine brain significantly modulated behavioral responses associated with disrupted NMDA receptor transmission. Furthermore, pretreatment with the antipsychotic drugs haloperidol and clozapine prevented dizocilpine-induced effects on miR-219. Taken together, these data support an integral role for miR-219 in the expression of behavioral aberrations associated with NMDA receptor hypofunction.


Subject(s)
Genetic Therapy , MicroRNAs/genetics , MicroRNAs/therapeutic use , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Behavior, Animal/drug effects , Biological Transport , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line, Tumor , Dizocilpine Maleate/pharmacology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Nervous System Diseases/physiopathology , Nervous System Diseases/therapy , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction
10.
FASEB J ; 22(11): 3823-35, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18703672

ABSTRACT

Hyperhomocysteinemia (HHcy)-abnormally elevated plasma levels of homocysteine (Hcy)-has been associated with the development of neurodegenerative dementia and mild cognitive impairment. This association suggests that HHcy might facilitate memory loss in the elderly. As memory loss can occur through a deteriorated neurogenic capacity, we have studied the effects of Hcy on neural progenitor cells (NPCs) both in vitro and in vivo. We show that Hcy exerts an antiproliferative effect on basic fibroblast growth factor (bFGF) -stimulated NPCs isolated from the postnatal subventricular zone (SVZ), accompanied by inactivation of the extracellular signal-regulated kinase (Erk1/2) and inhibition of Erk1/2-dependent expression of cyclin E. Using a mice model we show that, under normal folate conditions, HHcy exerts an inhibitory effect on adult brain neurogenesis. This inhibition occurs in the caudal areas of the dentate gyrus (DG) of the hippocampus, a neurogenic area mainly involved in learning and memory performance, and in the SVZ, recently implicated in olfactory learning performance. In both areas reduced number of proliferative neuroblasts were found. Since neuroblasts are primarily bFGF-responsive progenitors already committed to a neuronal phenotype, our results strongly suggest that excess Hcy inhibits neurogenesis in the DG and SVZ by inhibiting the bFGF-dependent activation of Erk1/2 in these cells.


Subject(s)
Adult Stem Cells/metabolism , Cyclin E/biosynthesis , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation/drug effects , Homocysteine/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/enzymology , Adult Stem Cells/pathology , Aged , Aged, 80 and over , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cognition Disorders/enzymology , Cognition Disorders/pathology , Dementia/enzymology , Dementia/pathology , Dentate Gyrus/enzymology , Dentate Gyrus/pathology , Disease Models, Animal , Fibroblast Growth Factor 2/pharmacology , Folic Acid/metabolism , Homocysteine/metabolism , Humans , Hyperhomocysteinemia/enzymology , Hyperhomocysteinemia/pathology , Mice , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Trigeminal Caudal Nucleus/enzymology , Trigeminal Caudal Nucleus/pathology
11.
J Alzheimers Dis ; 12(3): 229-40, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18057556

ABSTRACT

APP overexpressing mice have been widely used in the study of Alzheimer's disease (AD), focusing mainly at older ages, with higher accumulation of amyloid-beta peptide (Abeta). A decrease in hippocampal adult neurogenesis has been described in these models and proposed to be a consequence of Abeta accumulation. Only one study demonstrates increased neurogenesis in the hippocampus of APP-overexpressing J20 mice, and suggests it is a compensatory effect due to a subtle Abeta-induced damage. We have previously reported that a specific aggregation of Abeta has neurogenic potential on neural stem cells (NSC) in vitro. In order to clarify the contradicting data reported in vivo, we investigated NSC proliferation and neuronal differentiation in the hippocampi of J20 mice at a broader range of ages. Using immunohistochemistry, we show increased proliferation and neuronal differentiation in the hippocampi of 3 month-old J20 mice that reverted when animals became older. The increase in neurogenesis correlated with detectable levels of oligomeric Abeta, measured by ELISA and western blot. We suggest that oligomeric Abeta directly induces neurogenesis in vivo as has been demonstrated in vitro. Understanding the mechanisms underlying these changes could lead to treatments to control the neuronal differentiation of endogenous precursors through the progress of AD.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/immunology , Amyloid beta-Protein Precursor/metabolism , Animals , Chromatography, High Pressure Liquid , Disease Models, Animal , Hippocampus/pathology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Mice , Mice, Transgenic , Neural Cell Adhesion Molecule L1/immunology , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Sialic Acids/immunology
12.
J Neurosci Res ; 85(6): 1170-82, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17342781

ABSTRACT

Glutamate is an excitatory amino acid that serves important functions in mammalian brain development through alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA)/ kainate receptor stimulation. Neural stem cells with self-renewal and multilineage potential are a useful tool to study the signals involved in the regulation of brain development. We have investigated the role played by AMPA/kainate receptors during the differentiation of neural stem cells derived from fetal rat striatum. The application of 1 and 10 microM kainic acid increased significantly the phosphorylation of the cyclic AMP response element binding protein (CREB), raised bromodeoxyuridine incorporation in O4-positive oligodendrocyte precursors, and increased the number of O1-positive cells in the cultures. Increased CREB phosphorylation and proliferation were prevented by the AMPA receptor antagonist 4-4(4-aminophenyl)-1,2-dihydro-1-methyl-2-propylcarbamoyl-6,7-methylenedioxyphthalazine (SYM 2206) and by protein kinase A and protein kinase C inhibitors. Cultures treated with 100 microM kainic acid showed decreased proliferation, a lower proportion of O1-positive cells, and apoptosis of O4-positive cells. None of these effects were prevented by SYM 2206, suggesting that kainate receptors take part in these events. We conclude that AMPA receptor stimulation by kainic acid promotes the proliferation of oligodendrocyte precursors derived from neural stem cells through a mechanism that requires the activation of CREB by protein kinase A and C. In the neurons derived from these cells, either AMPA or kainate receptor stimulation produces neuritic growth and larger cell bodies.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Corpus Striatum/cytology , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , Neurons/physiology , Oligodendroglia/drug effects , Stem Cells/drug effects , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Bromodeoxyuridine/metabolism , Calcium/metabolism , Cells, Cultured , Corpus Striatum/embryology , Cyclic AMP Response Element-Binding Protein/metabolism , Dose-Response Relationship, Drug , Embryo, Mammalian , In Situ Nick-End Labeling/methods , Neurons/drug effects , Oligodendroglia/physiology , Phthalazines/pharmacology , Rats , Stem Cells/classification
13.
J Neurochem ; 93(4): 861-74, 2005 May.
Article in English | MEDLINE | ID: mdl-15857389

ABSTRACT

Neurofilament light gene mutations have been linked to a subset of patients with Charcot-Marie-Tooth disease, the most common inherited motor and sensory neuropathy. We have previously shown that Charcot-Marie-Tooth-linked mutant neurofilament light assembles abnormally in non-neuronal cells. In this study, we have characterized the effects of expression of mutant neurofilament light proteins on axonal transport in a neuronal cell culture model. We demonstrated that the Charcot-Marie-Tooth-linked neurofilament light mutations: (i) affect the axonal transport of mutant neurofilaments; (ii) have a dominant-negative effect on the transport of wild-type neurofilaments; (iii) affect the transport of mitochondria and the anterograde axonal transport marker human amyloid precursor protein; (iv) result in alterations of retrograde axonal transport and (v) cause fragmentation of the Golgi apparatus. Increased neuritic degeneration was observed in neuronal cells overexpressing neurofilament light mutants. Our results suggest that these generalized axonal transport defects could be responsible for the neuropathy in Charcot-Marie-Tooth disease.


Subject(s)
Axonal Transport/physiology , Charcot-Marie-Tooth Disease/genetics , Mutation , Neurofilament Proteins/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Newborn , Cells, Cultured , Charcot-Marie-Tooth Disease/physiopathology , Cloning, Molecular/methods , Disease Models, Animal , Fluorescent Antibody Technique/methods , Gene Expression Regulation/genetics , Golgi Apparatus/metabolism , Humans , Mitochondria/metabolism , Mutagenesis/physiology , Neurofilament Proteins/deficiency , Neurons/metabolism , Neurons/ultrastructure , Rats , Sympathetic Nervous System/cytology , Time Factors , Transfection/methods
14.
J Neuropathol Exp Neurol ; 63(7): 759-74, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15290901

ABSTRACT

The human neurofilament medium (hNFM) subunit is one of the 3 neurofilament (NF) polypeptides, which are the most abundant intermediate filament (IF) proteins in post-mitotic neurons. The formation of neurofilamentous aggregates is a pathological hallmark of many neurodegenerative diseases, including the Lewy bodies found in Parkinson disease (PD). A Gly336Ser (G336S) variant in the rod domain of hNFM has recently been described in a patient with early-onset autosomal-dominant PD. In this study, we have generated a mammalian expression vector encoding the variant hNFM cDNA and characterized its effects on the formation of heteropolymeric IFs in heterologous cell lines. We have also investigated the distribution of the (G336S) hNFM variant protein in neuronal CAD cells, as well as the effects of the variant on the distribution of other cellular organelles and proteins. Our results demonstrate that the G336S variant does not affect the formation of IF networks nor the distribution of the variant hNFM protein. Our data suggest that if the G336S variant is involved in the development of PD, it does not appear to be due to defects in the assembly and distribution of NFs.


Subject(s)
Brain/metabolism , Lewy Bodies/genetics , Neurofilament Proteins/genetics , Neurons/metabolism , Parkinson Disease/genetics , Amino Acid Substitution , Amyloid beta-Protein Precursor/metabolism , Brain/pathology , Brain/physiopathology , Cell Line, Tumor , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/metabolism , DNA Mutational Analysis , DNA, Complementary/analysis , DNA, Complementary/genetics , Genetic Predisposition to Disease , Humans , Lewy Bodies/metabolism , Lewy Bodies/pathology , Mitochondria/genetics , Mitochondria/metabolism , Mutation/genetics , Neurofilament Proteins/biosynthesis , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Polymorphism, Genetic/genetics , Protein Transport/genetics
15.
J Histochem Cytochem ; 52(9): 1177-89, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15314085

ABSTRACT

Neural stem cells (NSC) with self-renewal and multilineage potential are considered good candidates for cell replacement of damaged nervous tissue. In vitro experimental conditions can differentiate these cells into specific neuronal phenotypes. In the present study, we describe the combined effect of basic fibroblast growth factor (bFGF) and dibutyryladenosine 3',5'-cyclic monophosphate (dbcAMP) on the differentiation of fetal rat striatal NSC into tyrosine hydroxylase-positive cells. Tyrosine hydroxylase induction was accompanied by the activation of ERK1/ERK2 mitogen-activated protein kinase and was inhibited by the ERK1/ERK2 pathway blocker PD98059, suggesting that ERK activation may be important for this process. In addition, protein kinase C (PKC) was shown to be required for tyrosine hydroxylase protein expression. The inhibition of PKC by staurosporin, as well as its downregulation, decreased the ability of bFGF+dbcAMP to generate tyrosine hydroxylase-positive cells. Moreover, the PKC activator phorbol 12-myristate 13-acetate (PMA) together with bFGF and dbcAMP led to a significant increase in phospho-ERK1/ERK2 levels, and the percentage of beta-tubulin III-positive cells that expressed tyrosine hydroxylase increased by 3.5-fold. PMA also promoted the phosphorylation of the cyclic AMP response element binding protein that might contribute to the increase in tyrosine hydroxylase-positive cells observed in bFGF+dbcAMP+PMA-treated cultures. From these results, we conclude that the manipulation in vitro of NSC from rat fetal striatum with bFGF, cyclic AMP analogs, and PKC activators promotes the generation of tyrosine hydroxylase-positive neurons.


Subject(s)
Cyclic AMP/pharmacology , DNA-Binding Proteins , Fibroblast Growth Factor 2/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Neurons/enzymology , Protein Kinase C/metabolism , Stem Cells/enzymology , Tyrosine 3-Monooxygenase/metabolism , Activating Transcription Factor 1 , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Cyclic AMP/analogs & derivatives , Enzyme Induction/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Neurons/cytology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Stem Cells/drug effects , Transcription Factors/metabolism
16.
J Neurosci ; 24(23): 5439-44, 2004 Jun 09.
Article in English | MEDLINE | ID: mdl-15190117

ABSTRACT

The adult mammalian brain contains neural stem cells (NSCs) with self-renewal and multilineage potential in the hippocampus and subventricular zone. However, neurogenesis from these areas does not compensate for neuronal loss in age-related neurodegenerative disorders such as Alzheimer's disease (AD). To test whether an impairment of neurogenesis could contribute to the pathogenesis of AD, we examined the effects of amyloid-beta peptide (Abeta) on the survival and neuronal differentiation of cultured NSCs from striatum and hippocampus. We show that Abeta peptide does not impair the neurogenic rate in NSC progeny, but that it increases the total number of neurons in vitro in a dose-dependent manner. The neurogenic effect of Abeta peptide is not dependent on soluble factors released from the NSC progeny. Neurogenesis is induced by Abeta42 and not Abeta40 or Abeta 25-35, and the activity appears to be a property of Abeta oligomers and not fibrils. These results suggest that Abeta may have positive as well as deleterious actions, and that a knowledge of the mechanisms involved in the former could be valuable in exploiting the regenerative and plastic potential of the brain in preventing and treating Alzheimer's disease.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Neurons/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/adverse effects , Animals , Animals, Newborn , Cell Culture Techniques , Cell Differentiation , Cell Survival , Corpus Striatum/cytology , Corpus Striatum/metabolism , Embryo, Mammalian , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Inbred Strains , Nerve Regeneration , Neuronal Plasticity , Neurons/pathology , Plaque, Amyloid/metabolism , Rats , Rats, Sprague-Dawley , Stem Cells
17.
J Histochem Cytochem ; 51(1): 89-103, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12502758

ABSTRACT

Neural stem cells proliferate in liquid culture as cell clusters (neurospheres). This study was undertaken to characterize the epidermal growth factor (EGF)-expanded free-floating neurospheres derived from rat fetal striatum. We examined the ultrastructural and antigenic characteristics of these spheres. They consisted of two cell types, electron-dense and electron-lucent cells. Lucent cells were immunopositive to actin, vimentin, and nestin, whereas dense cells were immunopositive to actin, weakly positive to vimentin, and nestin-negative. Neurospheres contained healthy, apoptotic, and necrotic cells. Healthy cells were attached to each other by adherens junctions. They showed many pseudopodia and occasionally a single cilium. Sphere cells showed phagocytic capability because healthy cells phagocytosed the cell debris derived from dead cells in a particular process that involves the engulfment of dying cells by cell processes from healthy cells. Sphere cells showed a cytoplasmic and a nuclear pool of fibroblast growth factor (FGF) receptors. They expressed E- and N-cadherin, alpha- and beta-catenin, EGF receptor, and a specific subset of FGF receptors. Because sphere cells expressed this factor in the absence of exogenous FGF-2, we propose that they are able to synthesize FGF-2.


Subject(s)
Corpus Striatum/cytology , Epidermal Growth Factor/metabolism , Stem Cells/ultrastructure , Animals , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cells, Cultured , Corpus Striatum/ultrastructure , Cytoskeletal Proteins/metabolism , ErbB Receptors/metabolism , Fetus , Rats , Rats, Sprague-Dawley , Stem Cells/metabolism , Transforming Growth Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...