Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Neurotherapeutics ; : e00388, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38972779

ABSTRACT

Protein misfolding and mislocalization are common to both familial and sporadic forms of amyotrophic lateral sclerosis (ALS). Maintaining proteostasis through induction of heat shock proteins (HSP) to increase chaperoning capacity is a rational therapeutic strategy in the treatment of ALS. However, the threshold for upregulating stress-inducible HSPs remains high in neurons, presenting a therapeutic obstacle. This study used mouse models expressing the ALS variants FUSR521G or SOD1G93A to follow up on previous work in cultured motor neurons showing varied effects of the HSP co-inducer, arimoclomol, and class I histone deacetylase (HDAC) inhibitors on HSP expression depending on the ALS variant being expressed. As in cultured neurons, neither expression of the transgene nor drug treatments induced expression of HSPs in cortex, spinal cord or muscle of FUSR521G mice, indicating suppression of the heat shock response. Nonetheless, arimoclomol, and RGFP963, restored performance on cognitive tests and improved cortical dendritic spine densities. In SOD1G93A mice, multiple HSPs were upregulated in hindlimb skeletal muscle, but not in lumbar spinal cord with the exception of HSPB1 associated with astrocytosis. Drug treatments improved contractile force but reduced the increase in HSPs in muscle rather than facilitating their expression. The data point to mechanisms other than amplification of the heat shock response underlying recovery of cognitive function in ALS-FUS mice by arimoclomol and class I HDAC inhibition and suggest potential benefits in counteracting cognitive impairment in ALS, frontotemporal dementia and related disorders.

2.
Cell Stress Chaperones ; 29(3): 359-380, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38570009

ABSTRACT

Protein misfolding and mislocalization are common themes in neurodegenerative disorders, including motor neuron disease, and amyotrophic lateral sclerosis (ALS). Maintaining proteostasis is a crosscutting therapeutic target, including the upregulation of heat shock proteins (HSP) to increase chaperoning capacity. Motor neurons have a high threshold for upregulating stress-inducible HSPA1A, but constitutively express high levels of HSPA8. This study compared the expression of these HSPs in cultured motor neurons expressing three variants linked to familial ALS: TAR DNA binding protein 43 kDa (TDP-43)G348C, fused in sarcoma (FUS)R521G, or superoxide dismutase I (SOD1)G93A. All variants were poor inducers of Hspa1a, and reduced levels of Hspa8 mRNA and protein, indicating multiple compromises in chaperoning capacity. To promote HSP expression, cultures were treated with the putative HSP coinducer, arimoclomol, and class I histone deacetylase inhibitors, to promote active chromatin for transcription, and with the combination. Treatments had variable, often different effects on the expression of Hspa1a and Hspa8, depending on the ALS variant expressed, mRNA distribution (somata and dendrites), and biomarker of toxicity measured (histone acetylation, maintaining nuclear TDP-43 and the neuronal Brm/Brg-associated factor chromatin remodeling complex component Brg1, mitochondrial transport, FUS aggregation). Overall, histone deacetylase inhibition alone was effective on more measures than arimoclomol. As in the FUS model, arimoclomol failed to induce HSPA1A or preserve Hspa8 mRNA in the TDP-43 model, despite preserving nuclear TDP-43 and Brg1, indicating neuroprotective properties other than HSP induction. The data speak to the complexity of drug mechanisms against multiple biomarkers of ALS pathogenesis, as well as to the importance of HSPA8 for neuronal proteostasis in both somata and dendrites.


Subject(s)
Amyotrophic Lateral Sclerosis , Biomarkers , DNA-Binding Proteins , Histone Deacetylase Inhibitors , Motor Neurons , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Biomarkers/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Humans , Motor Neurons/metabolism , Motor Neurons/drug effects , Motor Neurons/pathology , Animals , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/metabolism , HSC70 Heat-Shock Proteins/genetics , Hydroxylamines/pharmacology , Cells, Cultured , RNA-Binding Protein FUS/metabolism , RNA-Binding Protein FUS/genetics , Superoxide Dismutase-1/metabolism , Superoxide Dismutase-1/genetics
3.
Front Oncol ; 10: 555945, 2020.
Article in English | MEDLINE | ID: mdl-33072581

ABSTRACT

Glioblastoma multiforme is an aggressive malignancy, resistant to standard treatment modalities and associated with poor prognosis. We analyzed the role of the IGF system in intracerebral glioma growth using human and rat glioma cells. The glioma cells C6 and U87MG were transduced with a genetically engineered retrovirus expressing type 1 insulin-like growth factor (IGF-IR) antisense RNA, either before or after intra-cerebral implantation of the cells into Sprague Dawley rats or nude mice, respectively and tumor growth and animal survival were monitored. Rat glioma cells transduced prior to orthotopic, intra-cerebral implantation had a significantly increased apoptotic rate in vivo and a significantly reduced tumor volume as seen 24 days post implantation (p < 0.0015). This resulted in increased survival, as greater than 70% of the rats were still alive 182 days after tumor implantation (p < 0.01), as compared to 80% mortality by day 24 in the control group. Histomorphology and histochemical studies performed on brain tissue that was obtained from rats that survived for 182 days revealed numerous single cells that were widely disseminated throughout the brain. These cells expressed the ß-galactosidase marker protein, but were Ki67negative, suggesting that they acquired a dormant phenotype. Direct targeting of the C6 cells with retroviral particles in vivo was effective and reduced tumor volumes by 22% relative to controls. A significant effect on tumor growth was also seen with human glioma U87MG cells that were virally transduced and implanted intra-cerebrally in nude mice. We observed in these mice a significant reduction in tumor volumes and 70% of the animals were still alive 6 months after tumor implantation, as compared to 100% mortality in the control group by day 63. Our results show that IGF-IR targeting can inhibit the intracerebral growth of glioma cells. They also suggest that IGF-IR expression levels may determine a delicate balance between glioma cell growth, death and the acquisition of a dormant state in the brain.

4.
Cell Stress Chaperones ; 25(1): 173-191, 2020 01.
Article in English | MEDLINE | ID: mdl-31900865

ABSTRACT

Upregulation of heat shock proteins (HSPs) is an approach to treatment of neurodegenerative disorders with impaired proteostasis. Many neurons, including motor neurons affected in amyotrophic lateral sclerosis (ALS), are relatively resistant to stress-induced upregulation of HSPs. This study demonstrated that histone deacetylase (HDAC) inhibitors enable the heat shock response in cultured spinal motor neurons, in a stress-dependent manner, and can improve the efficacy of HSP-inducing drugs in murine spinal cord cultures subjected to thermal or proteotoxic stress. The effect of particular HDAC inhibitors differed with the stress paradigm. The HDAC6 (class IIb) inhibitor, tubastatin A, acted as a co-inducer of Hsp70 (HSPA1A) expression with heat shock, but not with proteotoxic stress induced by expression of mutant SOD1 linked to familial ALS. Certain HDAC class I inhibitors (the pan inhibitor, SAHA, or the HDAC1/3 inhibitor, RGFP109) were HSP co-inducers comparable to the hydroxyamine arimoclomol in response to proteotoxic stress, but not thermal stress. Regardless, stress-induced Hsp70 expression could be enhanced by combining an HDAC inhibitor with either arimoclomol or with an HSP90 inhibitor that constitutively induced HSPs. HDAC inhibition failed to induce Hsp70 in motor neurons expressing ALS-linked mutant FUS, in which the heat shock response was suppressed; yet SAHA, RGFP109, and arimoclomol did reduce loss of nuclear FUS, a disease hallmark, and HDAC inhibition rescued the DNA repair response in iPSC-derived motor neurons carrying the FUSP525Lmutation, pointing to multiple mechanisms of neuroprotection by both HDAC inhibiting drugs and arimoclomol.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Heat-Shock Proteins/drug effects , Hydroxylamines/pharmacology , Motor Neurons/drug effects , Spinal Cord/drug effects , Amyotrophic Lateral Sclerosis/genetics , Animals , Cells, Cultured , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response/drug effects , Histone Deacetylase Inhibitors/pharmacology , Mice , Motor Neurons/metabolism , Spinal Cord/metabolism , Transcriptional Activation/drug effects , Up-Regulation/drug effects
5.
Clin Neurol Neurosurg ; 173: 20-30, 2018 10.
Article in English | MEDLINE | ID: mdl-30055402

ABSTRACT

OBJECTIVE: Glial tumor growth may accelerate during gestation, but epidemiological studies consistently demonstrated that parousity reduces life long risk of glial tumors. Pregnancy may also accelerate growth of medulloblastoma and meningioma, but parousity does not confer protection against these tumors. We were the first to show that medroxyprogesterone acetate (MPA) reduces rat C6 glioma growth in vitro. Now we aimed to determine the effects of MPA on human brain cancers (particularly glioblastoma) in vitro and C6 glioma in vivo. PATIENTS AND METHODS: We evaluated the effects of MPA on: i) monolayer growth of human U87 and U251 glioblastoma, ii) 3D-spheroid growth and invasion of C6 rat glioma and human U251 glioma, iii) interactions with PI3-Kinase inhibitors and coxsackie-adenovirus receptor (CAR) in modifying 3D-spheroid invasion of glioma. RESULTS: MPA at low doses (3.25-13 µM) insignificantly stimulated and at high doses (above 52 µM) strongly suppressed the growth of human U87 and U251 cells in vitro. MPA also binds to glucocorticoid receptors similar to dexamethasone (Dex) and unexpectedly, PI3-Kinase inhibitors at low doses suppressed anti-invasive efficacies of MPA and Dex. MPA exerted higher invasion-inhibitory effects on CAR-expressing human glioma cells. Lastly, MPA suppressed growth of C6 glioma implanted into rat brain. CONCLUSION: Progesterone analogues deserve to be studied in future experimental models of high grade glial brain tumors.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Glioma/drug therapy , Medroxyprogesterone/pharmacology , Animals , Brain/drug effects , Brain/pathology , Brain Neoplasms/pathology , Cerebellar Neoplasms/drug therapy , Dexamethasone/pharmacology , Disease Models, Animal , Glioblastoma/pathology , Glioma/pathology , Humans , Meningioma/drug therapy , Rats , Tumor Cells, Cultured/drug effects
6.
BMC Res Notes ; 7: 183, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24674422

ABSTRACT

BACKGROUND: Alterations in cell migration are a hallmark of cancer cell invasion and metastasis. In vitro assays commonly used to study cell migration, including the scratch wound healing assay, Boyden chamber assay, and newly developed advanced systems with microfluidics, each have several disadvantages. FINDINGS: Here we describe an easy and cost-effective in vitro assay for cell migration employing cloning rings to create gaps in the cell monolayer ("ring cell migration assay"). The assay was used to quantitate innate differences in cell motility and the effect of various extracellular matrix proteins on migration of five cancer cell lines: U87 and U251N glioma cells, MDA-MB-231 and MCF-7 breast cancer cells, and HeLa cervical cancer cells. Interestingly, collagen was a general promoter of cell migration for all five cancer cell lines, without affecting cell proliferation. CONCLUSIONS: Taken together, the ring cell migration assay is an easy, convenient and cost-effective assay to study cell migration in vitro.


Subject(s)
Cell Migration Assays , Cell Movement/drug effects , Extracellular Matrix Proteins/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Extracellular Matrix Proteins/chemistry , Humans
7.
PLoS One ; 8(8): e73296, 2013.
Article in English | MEDLINE | ID: mdl-24015300

ABSTRACT

The Coxsackievirus and Adenovirus Receptor (CAR) is a cell adhesion molecule originally characterized as a virus receptor but subsequently shown to be involved in physiological processes such as neuronal and heart development, epithelial tight junction integrity, and tumour suppression. Proteolysis of cell adhesion molecules and a wide variety of other cell surface proteins serves as a mechanism for protein turnover and, in some cases, cell signaling. Metalloproteases such as A Disintegrin and Metalloprotease (ADAM) family members cleave cell surface receptors to release their substrates' ectodomains, while the presenilin/ɣ-secretase complex mediates regulated intramembrane proteolysis (RIP), releasing intracellular domain fragments from the plasma membrane. In the case of some substrates such as Notch and amyloid precursor protein (APP), the released intracellular domains enter the nucleus to modulate gene expression. We report that CAR ectodomain is constitutively shed from glioma cells and developing neurons, and is also shed when cells are treated with the phorbol ester phorbol 12-myristate 13-acetate (PMA) and the calcium ionophore ionomycin. We identified ADAM10 as a sheddase of CAR using assays involving shRNA knockdown and rescue, overexpression of wild-type ADAM10 and inhibition of ADAM10 activity by addition of its prodomain. In vitro peptide cleavage, mass spectrometry and mutagenesis revealed the amino acids M224 to L227 of CAR as the site of ADAM10-mediated ectodomain cleavage. CAR also undergoes RIP by the presenilin/γ-secretase complex, and the intracellular domain of CAR enters the nucleus. Ectodomain shedding is a prerequisite for RIP of CAR. Thus, CAR belongs to the increasing list of cell surface molecules that undergo ectodomain shedding and that are substrates for ɣ-secretase-mediated RIP.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Cell Membrane/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Presenilins/metabolism , Proteolysis , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM10 Protein , Amyloid Precursor Protein Secretases/genetics , Animals , Carcinogens/pharmacology , Cell Line, Tumor , Cell Membrane/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutagenesis, Site-Directed , Neurons/cytology , Neurons/metabolism , Presenilins/genetics , Protein Structure, Tertiary , Tetradecanoylphorbol Acetate/pharmacology
8.
J Gene Med ; 14(12): 746-60, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23071006

ABSTRACT

BACKGROUND: Gutless adenovirus (helper-dependent adenoviral vector; HDAd) and lentiviral vectors (LV) are attractive vectors for the gene therapy of muscle diseases. Because the organization of their DNA (episomal versus integrated) differs, we investigated whether the strength and specificity of ΔUSEx3, a novel muscle-specific promoter previously tested with plasmid, were maintained in the context of these vectors. METHODS: Two HDAds expressing ß-galactosidase regulated by ΔUSEx3 or CAG [cytomegalovirus (CMV) enhancer/ß-actin promoter], and three LV expressing green fluorescent protein regulated by ΔUSEx3, CMV or a modified skeletal α-actin promoter (SPcΔ5-12), were constructed. Gene expression was compared in cell culture and after intravenous (HDAd only) and intramuscular injection of mice. RESULTS: Irrespective of the vector used, ΔUSEx3 remained poorly active in nonmuscle cells and tissues. In myotubes, ΔUSEx3 was as strong as CMV and SPcΔ5-12, although it was ten-fold weaker than CAG, a proven powerful promoter in muscle. In cell culture, ΔUSEx3 activity in the context of LV was more stable than CMV, indicating it is less prone to silencing. In the context of HDAd, the behavior of ΔUSEx3 in skeletal muscle mirrored that of cell culture (10% of the CAG activity and half the number of transduced fibers). Surprisingly, in muscles treated with LV, ΔUSEx3 activity was five-fold lower than SPcΔ5-12. CONCLUSIONS: The data obtained in the present study confirm that ΔUSEx3 is a strong and robust muscle-specific promoter in the context of HDAd (cell culture and in vivo) and LV (cell culture). However, it was less efficient in vivo in the context of LV.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Lentivirus/genetics , Muscle, Skeletal/metabolism , Promoter Regions, Genetic , Troponin I/genetics , Animals , Cell Line , Cytomegalovirus/genetics , Enhancer Elements, Genetic , Gene Expression , Gene Order , Humans , Mice , Organ Specificity/genetics , Troponin I/metabolism
9.
Endocrinology ; 152(12): 4581-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21952243

ABSTRACT

The protein tyrosine phosphatase (PTPase) Src-homology 2-domain-containing phosphatase (SHP)-1 was recently reported to be a novel regulator of insulin's metabolic action. In order to examine the role of this PTPase in skeletal muscle, we used adenovirus (AdV)-mediated gene transfer to express an interfering mutant of SHP-1 [dominant negative (DN)SHP-1; mutation C453S] in L6 myocytes. Expression of DNSHP-1 increased insulin-induced Akt serine-threonine kinase phosphorylation and augmented glucose uptake and glycogen synthesis. Pharmacological inhibition of glucose transporter type 4 (GLUT4) activity using indinavir and GLUT4 translocation assays revealed an important role for this transporter in the increased insulin-induced glucose uptake in DNSHP-1-expressing myocytes. Both GLUT4 mRNA and protein expression were also found to be increased by DNSHP-1 expression. Furthermore, AdV-mediated delivery of DNSHP-1 in skeletal muscle of transgenic mice overexpressing Coxsackie and AdV receptor also enhanced GLUT4 protein expression. Together, these findings confirm that SHP-1 regulates muscle insulin action in a cell-autonomous manner and further suggest that the PTPase negatively modulates insulin action through down-regulation of both insulin signaling to Akt and GLUT4 translocation, as well as GLUT4 expression.


Subject(s)
Glucose/metabolism , Muscle Fibers, Skeletal/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Receptor, Insulin/metabolism , Animals , Glucose Transporter Type 4/metabolism , Glycogen/biosynthesis , Mice , Mice, Transgenic , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
10.
J Muscle Res Cell Motil ; 31(3): 181-93, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20706865

ABSTRACT

Duchenne muscular dystrophy is a genetic muscle disease characterized by the absence of sub-sarcolemmal dystrophin that results in muscle fibre necrosis, progressive muscle wasting and is fatal. Numerous experimental studies with dystrophin-deficient mdx mice, an animal model for the disease, have demonstrated that extrasynaptic upregulation of utrophin, an analogue of dystrophin, can prevent muscle fibre deterioration and reduce or negate the dystrophic phenotype. A different approach for ectopic expression of utrophin relies on augmentation of CT-GalNAc transferase in muscle fibre. We investigated whether CT-GalNAc transferase overexpression in adult mice influence appearance of utrophin in the extrasynaptic sarcolemma. After electrotransfer of plasmid DNA carrying an expression cassette of CT-GalNAc transferase into tibialis anterior muscle of wild type and dystrophic mice, muscle sections were examined by immunofluorescence. CT-GalNAc transgene expression augmented sarcolemmal carbohydrate glycosylation and was accompanied by extrasynaptic utrophin. A 6-week time course study showed that the highest efficiency of utrophin overexpression in a plasmid harboured muscle fibres was 32.2% in CD-1 and 52% in mdx mice, 2 and 4 weeks after CT-GalNAc gene transfer, respectively. The study provides evidence that postnatal CT-GalNAc transferase overexpression stimulates utrophin upregulation that is inherently beneficial for muscle structure and strength restoration. Thus CT-GalNAc may provide an important therapeutic molecule for treatment of dystrophin deficiency in Duchenne muscular dystrophy.


Subject(s)
Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , N-Acetylgalactosaminyltransferases/biosynthesis , Neuromuscular Junction/metabolism , Utrophin/metabolism , Animals , Cell Line , Disease Models, Animal , Gene Expression , Glycosylation , Humans , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/genetics , N-Acetylgalactosaminyltransferases/genetics , Neuromuscular Junction/genetics , Sarcolemma/genetics , Sarcolemma/metabolism , Utrophin/genetics
11.
Behav Brain Funct ; 6: 35, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20594357

ABSTRACT

BACKGROUND: The Morris water maze task is a hippocampus-dependent learning and memory test that typically takes between 3 days to 2 weeks of training. This task is used to assess spatial learning and induces the expression of genes known to be crucial to learning and memory in the hippocampus. A major caveat in the protocol is the prolonged duration of training, and difficulty of assessing the time during training in which animals have learned the task. We introduce here a condensed version of the task that like traditional water maze tasks, creates lasting hippocampus-dependent spatial cognitive maps and elicits gene expression following learning. METHODS: This paradigm was designed for rats to quickly acquire a hippocampus-dependent spatial cognitive map and retain this memory for at least 24 hours. To accomplish this, we interspersed visible and hidden training trials, delivering them in a massed fashion so training takes a maximum of 15 minutes. Learning was assessed based on latencies to the platform during each training trial, as well as time spent in the goal quadrant during probe testing 30 minutes and 24 hours after training. Normal rats were compared to two impaired cohorts (rats with fimbria-fornix lesions and rats administered NMDA receptor antagonist (CPP)). To quantitate hippocampal expression of known learning genes, real-time polymerase chain reaction (RT-PCR) was performed on hippocampal cDNA. RESULTS: We show that massed training using alternating visible and hidden training trials generates robust short-term working and long-term reference memories in rats. Like the traditional Morris water maze paradigm, this task requires proper hippocampal function, as rats with fimbria-fornix lesions and rats administered CPP fail to learn the spatial component of the task. Furthermore, training in this paradigm elicits hippocampal expression of genes upregulated following learning in a variety of spatial tasks: homer1a, cfos and zif268. CONCLUSIONS: We introduce here a condensed version of the Morris water maze, which is like a traditional water maze paradigm, in that it is hippocampus-dependent, and elicits hippocampal expression of learning genes. However, this task is administered in 15 minutes and induces spatial memory for at least 24 hours.


Subject(s)
Genes, Immediate-Early , Hippocampus/physiology , Maze Learning/physiology , Memory/physiology , Neuropsychological Tests , Space Perception/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Genes, Immediate-Early/drug effects , Genes, fos , Hippocampus/drug effects , Homer Scaffolding Proteins , Male , Maze Learning/drug effects , Memory/drug effects , Piperazines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Space Perception/drug effects , Time Factors
12.
Cancer Biol Ther ; 9(11): 843-52, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20372085

ABSTRACT

Dysregulation of various signaling pathways that govern cerebellar development with respect to cell proliferation, growth arrest, apoptosis and differentiation has been postulated to contribute to medulloblastoma tumourigenesis. This review will highlight the unique nature of cerebellar development in terms of its derivation from two germinal matrices and significant postnatal expansion of the granule cell precursor (GCP) compartment resulting in granule cell development and migration to form the mature cerebellar cortex. The molecular signals that are critical for timely cell cycle exit and differentiation may become dysregulated leading to unrestrained cell proliferation and enhanced cell survival; indeed, changes in these molecular markers have been observed in medulloblastoma biopsy specimens. Furthermore, transgenic models that faithfully replicate these changes develop medulloblastoma that, by in large, recapitulates the clinico-histopathological features of these tumours. Cellular and developmental biological approaches have contributed greatly to the current debate on the relevance of the cancer stem cell hypothesis in understanding medulloblastoma initiation and propagation. Penultimately, research findings are being translated into experimental therapeutics that target the aberrant signal transduction machinery in medulloblastoma cells and that will hopefully lead to an improved risk-benefit profile.


Subject(s)
Antineoplastic Agents/therapeutic use , Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Histone Deacetylases/therapeutic use , Humans , Medulloblastoma/metabolism , Medulloblastoma/pathology , Models, Biological
13.
J Gene Med ; 12(3): 266-75, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20082422

ABSTRACT

BACKGROUND: Efficient adenovirus (AdV)-mediated gene transfer is possible only in immature muscle or regenerating muscle, suggesting that a developmentally regulated event plays a major role in limiting AdV uptake in mature skeletal muscle. Previously, we showed that the expression of the primary coxsackie and adenovirus receptor (CAR) is severely down-regulated during muscle maturation and that, in muscle-specific CAR transgenic mice, there is significant enhancement of AdV-mediated gene transfer to mature skeletal muscle. METHODS: To evaluate whether increasing CAR expression can also augment gene transfer to dystrophic muscle that has many regenerating fibers, we crossed CAR transgenics with dystrophin-deficient mice (mdx/CAR). We also tested a two-step protocol in which CAR levels were increased in the target muscle, prior to administration of AdV, through the use of recombinant adeno-associated virus (AAV2) expressing CAR. Lastly, we assessed the effect of histone deacetylase inhibitors on CAR and AdV transduction efficiency in myoblasts and mdx muscle. RESULTS: Although somewhat higher rates of transduction can be achieved in adult mdx mice than in normal mice as a result of ongoing muscle regeneration in these animals, CAR expression in the mdx background (mdx/CAR transgenics) still markedly improved the susceptibility of mature muscle to AdV-mediated gene transfer of dystrophin. Prior administration of AAV2-CAR to normal muscle led to significantly increased transduction by subsequent injection of AdV. The histone deacetylase inhibitor valproate increased CAR transcript and protein levels in myoblasts and mdx muscle, and also increased AdV-mediated gene transfer. CONCLUSIONS: We have developed a method of increasing CAR levels in both normal and regenerating muscle.


Subject(s)
Muscle Fibers, Skeletal/metabolism , Muscular Dystrophies/genetics , Receptors, Virus/genetics , Transduction, Genetic/methods , Adenoviridae , Animals , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Dystrophin/genetics , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle Fibers, Skeletal/drug effects , Regeneration , Transcription, Genetic/drug effects , Valproic Acid/pharmacology
14.
Int J Oncol ; 35(5): 1045-51, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19787258

ABSTRACT

Medulloblastoma is the most frequent type of childhood brain tumour. The insulin-like growth factor I receptor (IGF-IR) plays a significant neuroprotective role in medulloblastoma survival through regulation of the downstream effectors of the phosphoinositide-3-kinase-protein kinase-B (PI3K-PKB/c-Akt) pathway. One such target is Forkhead box O1 (FOXO1; FKHR), which is part of the FOXO family of Forkhead transcription factors. Phosphorylation by Akt results in cytoplasmic sequestration of FOXO1 thus inhibiting the expression of genes controlling cell death, cell proliferation, differentiation, cellular metabolism and oxidative stress. Here we show that serum starvation of medulloblastoma cells is accompanied by nuclear translocation of FOXO1. IGF-I stimulation of serum-starved cells resulted in rapid phosphorylation of Akt and FOXO1, and was associated with a significant increase in cell viability. In contrast, expression of a constitutively active form of FOXO1 that cannot be phosphorylated led to a significant reduction in medulloblastoma cell viability, even in the presence of growth factors provided by fetal bovine serum (FBS). These data suggest that the transcription factor FOXO1 may be a critical effector of medulloblastoma growth suppression.


Subject(s)
Cerebellar Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , Medulloblastoma/metabolism , Signal Transduction/physiology , Blotting, Western , Cell Line , Cell Survival/physiology , Enzyme Activation/physiology , Fluorescent Antibody Technique , Forkhead Box Protein O1 , Humans , Insulin-Like Growth Factor I/metabolism , Protein Transport/physiology , Receptor, IGF Type 1/metabolism
15.
Cancer Res ; 69(13): 5433-40, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19549905

ABSTRACT

Gliomas are the most common adult primary brain tumors, and the most malignant form, glioblastoma multiforme, is invariably fatal. The phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway is altered in most glioblastoma multiforme. PTEN, an important negative regulator of the PI3K-Akt pathway, is also commonly mutated in glioma, leading to constitutive activation of Akt. One ultimate consequence is phosphorylation and inactivation of FOXO forkhead transcription factors that regulate genes involved in apoptosis, cell cycle arrest, nutrient availability, DNA repair, stress, and angiogenesis. We tested the ability of a mutant FOXO1 factor that is not subject to Akt phosphorylation to overcome dysregulated PI3K-Akt signaling in two PTEN-null glioma cell lines, U87 and U251. Adenovirus-mediated gene transfer of the mutant FOXO1 successfully restored cell cycle arrest and induced cell death in vitro and prolonged survival in vivo in xenograft models of human glioma (33% survival at 1 year of animals bearing U251 tumors). However, U87 were much more resistant than U251 to mutant FOXO1-induced death, showing evidence of increased nuclear export and Akt-independent phosphorylation of FOXO1 at S249. A cyclin-dependent kinase 2 inhibitor decreased phosphorylation of S249 and rendered U87 cells significantly more susceptible to mutant FOXO1-induced death. Our results indicate that targeting FOXO1, which is at the convergence point of several growth factor receptor tyrosine kinase pathways, can effectively induce glioma cell death and inhibit tumor growth. They also highlight the importance of Akt-independent phosphorylation events in the nuclear export of FOXO1.


Subject(s)
Brain Neoplasms/pathology , Forkhead Transcription Factors/drug effects , Glioma/pathology , Adenoviridae/genetics , Animals , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cyclin D1/physiology , Cyclin D2 , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclins/physiology , DNA Repair , Disease Models, Animal , Forkhead Box Protein O1 , Forkhead Transcription Factors/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neovascularization, Pathologic/prevention & control , Phosphorylation
16.
J Biol Chem ; 283(50): 34720-7, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18945675

ABSTRACT

Duchenne muscular dystrophy is caused by a genetic defect in the dystrophin gene. The absence of dystrophin results in muscle fiber necrosis and regeneration, leading to progressive muscle fiber loss. Utrophin is a close analogue of dystrophin. A substantial, ectopic expression of utrophin in the extrasynaptic sarcolemma of dystrophin-deficient muscle fibers can prevent deleterious effects of dystrophin deficiency. An alternative approach for the extrasynaptic up-regulation of utrophin involves the augmentation of utrophin transcription via the endogenous utrophin A promoter using custom-designed transcriptional activator proteins with zinc finger (ZFP) motifs. We tested a panel of custom-designed ZFP for their ability to activate the utrophin A promoter. Expression of one such ZFP efficiently increased, in a time-dependent manner, utrophin transcript and protein levels both in vitro and in vivo. In dystrophic mouse (mdx) muscles, administration of adenoviral vectors expressing this ZFP led to significant enhancement of muscle function with decreased necrosis, restoration of the dystrophin-associated proteins, and improved resistance to eccentric contractions. These studies provide evidence that specifically designed ZFPs can act as strong transcriptional activators of the utrophin A promoter. These may thus serve as attractive therapeutic agents for dystrophin deficiency states such as Duchenne muscular dystrophy.


Subject(s)
Gene Expression Regulation , Muscles/pathology , Promoter Regions, Genetic , Transcription Factors/metabolism , Up-Regulation , Utrophin/genetics , Animals , Humans , Mice , Mice, Transgenic , Muscle Contraction , Muscles/metabolism , Muscular Dystrophy, Duchenne/metabolism , Necrosis , Time Factors
17.
Cytometry A ; 73(10): 940-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18773455

ABSTRACT

Side population (SP) analyses and CD133 expression have identified cells with stem-like potential in normal and cancerous tissue. Whether stem-like cells exist in cancer cell lines is hotly debated. We have interrogated the DAOY medulloblastoma cell line with respect to stem-like potential. Vital staining for Hoechst 33342 efflux capacity and CD133 immunophenotyping were performed on DAOY cells to assess the presence of the SP and the CD133 stem cell markers, respectively. SP/non-SP and CD133(+)/CD133(-) DAOY cells were sorted into separate fractions for limiting dilution analysis (tumor sphere assay) and asymmetric division assessment. SP/non-SP cells were also sorted separately for viability (XTT assay), cell size, cell cycle status, and proliferative capacity (carboxyfluorescein succinimidyl ester (CFSE)) evaluation. A minor proportion of cells displayed either the SP or the CD133(+) phenotypes. CD133 expression mapped to both the SP and non-SP compartments, with CD133(+) cells being enriched almost fourfold within the non-SP gate. The SP, non-SP, CD133(+), and CD133(-) fractions were all capable of reconstituting the original parental DAOY population. Slight clonogenic enrichment was observed in only the SP fraction; however, both CD133(+) and CD133(-) cells displayed equivalent stem cell-like frequencies. SP cells were resistant to Hoechst 33342-mediated toxicity relative to the parental population and differed from the non-SP cells with respect to increased cell size, decreased S-phase, and slightly decreased proliferative capacity. The multiparametric strategy described in this study revealed that the SP and CD133(+) subset may be two independent compartments. Our results highlight the need for new reliable specific cancer stem cell marker(s) as Hoechst 33342 efflux and CD133 expression might not be suitable for selectively isolating cancer stem-like cells from cell lines, as shown for the DAOY cells. As such, care must be used in interpreting therapeutic studies targeting the stem cell compartment of cancer cell lines.


Subject(s)
Antigens, CD/analysis , Biomarkers, Tumor/analysis , Flow Cytometry , Glycoproteins/analysis , Medulloblastoma/pathology , Neoplastic Stem Cells/pathology , Peptides/analysis , AC133 Antigen , Antigens, CD/biosynthesis , Benzimidazoles/analysis , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Cell Separation/methods , Flow Cytometry/methods , Fluoresceins/metabolism , Glycoproteins/biosynthesis , Humans , Medulloblastoma/metabolism , Neoplastic Stem Cells/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Succinimides/metabolism
18.
Hum Gene Ther ; 19(2): 133-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18067405

ABSTRACT

Adenoviral vectors that use the coxsackievirus and adenovirus receptor do not transduce mature muscle efficiently. Group B adenoviruses use CD46 as their cell attachment receptor. To evaluate the utility of vectors based on group B adenoviruses for gene transfer to human skeletal muscle, we assessed the expression of CD46 in biopsied normal skeletal muscle samples and in muscles from patients with Duchenne muscular dystrophy. Transcript levels of CD46 were extremely low in mature muscle and CD46 immunoreactivity was detected only on blood vessels in the muscle sections. Although myoblasts cultured from biopsied samples had robust cell surface CD46 expression by flow cytometry, CD46 transcript levels were barely detectable after differentiation of the myoblasts into myotubes. The myotubes were also much less susceptible to infection with an adenoviral vector carrying the fiber of serotype 35 adenovirus (AdF35). These results suggest that for skeletal muscle, vectors derived from group B adenoviruses may not be a suitable alternative to the commonly used Ad5 vectors.


Subject(s)
Adenoviridae/metabolism , Cell Differentiation , Down-Regulation/genetics , Membrane Cofactor Protein/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Transduction, Genetic/methods , Biopsy , Cell Membrane Permeability , Cells, Cultured , Flow Cytometry , Humans , Muscle Fibers, Skeletal , Muscular Dystrophy, Duchenne , Myoblasts , Reverse Transcriptase Polymerase Chain Reaction , beta-Galactosidase
19.
Mol Ther ; 15(10): 1767-74, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17667948

ABSTRACT

Helper-dependent adenovirus vector (AdV)-mediated full-length dystrophin expression leads to significant mitigation of the dystrophic phenotype of the mdx mouse. However, dystrophin, as a neoantigen, elicits antibody formation. As an alternative approach, we evaluated gene transfer of full-length murine utrophin, a functional homologue of dystrophin that is normally present only at the neuromuscular junction. A single injection in the tibialis anterior (TA) muscle of the helper-dependent adenovirus vector encoding utrophin provided very good transduction, with 58% of fibers demonstrating sarcolemmal utrophin expression in the neonates, and 35% utrophin-positive (Utr(+)) fibers in adults. The presence of utrophin prevented extensive necrosis in the neonates, halted further necrosis in the adults, and led to restoration of sarcolemmal expression of dystrophin-associated proteins up to 1 year after injection. Marked physiological improvement was observed in both neonates and adults. Neither increased humoral responses nor cellular immune responses were evident. However, there was a time-related decline of the initial high utrophin expression. Although viral DNA persisted in animals that were injected in the neonatal stage, viral DNA levels decreased in muscles of adult mice. These results demonstrate that although utrophin gene transfer leads to amelioration of the dystrophic phenotype, the effects are not sustained upon loss of utrophin expression.


Subject(s)
Adenoviridae/genetics , Dystrophin/genetics , Utrophin/genetics , Animals , Animals, Newborn , Antibody Formation , DNA, Viral/metabolism , Immunity, Cellular , Mice , Mice, Mutant Strains , Muscle, Skeletal/metabolism , Polymerase Chain Reaction , Transduction, Genetic , Utrophin/administration & dosage , Utrophin/immunology
20.
Cancer Res ; 67(14): 6700-7, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17638880

ABSTRACT

In solid tumors, cancer cells subjected to ischemic conditions trigger distinct signaling pathways contributing to angiogenic stimulation and tumor development. Characteristic features of tumor ischemia include hypoxia and glucose deprivation, leading to the activation of hypoxia-inducible factor-1-dependent signaling pathways and to complex signaling events known as the unfolded protein response. Here, we show that the activation of the endoplasmic reticulum stress sensor IRE1 is a common determinant linking hypoxia- and hypoglycemia-dependent responses to the up-regulation of vascular endothelial growth factor-A (VEGF-A). Tumor cells expressing a dominant-negative IRE1 transgene as well as Ire1alpha-null mouse embryonic fibroblasts were unable to trigger VEGF-A up-regulation upon either oxygen or glucose deprivation. These data correlated with a reduction of tumor angiogenesis and growth in vivo. Our results therefore suggest an essential role for IRE1-dependent signaling pathways in response to ischemia and identify this protein as a potential therapeutic target to control both the angiogenic switch and tumor development.


Subject(s)
Endoribonucleases/physiology , Gene Expression Regulation, Neoplastic , Ischemia , Membrane Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Brain/metabolism , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Genes, Dominant , Humans , Mice , Neovascularization, Pathologic , Oxygen/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...