Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cereb Cortex ; 25(11): 4259-72, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25596590

ABSTRACT

Corticospinal motor neurons (CSMN) receive, integrate, and relay cerebral cortex's input toward spinal targets to initiate and modulate voluntary movement. CSMN degeneration is central for numerous motor neuron disorders and neurodegenerative diseases. Previously, 5 patients with mutations in the ubiquitin carboxy-terminal hydrolase-L1 (UCHL1) gene were reported to have neurodegeneration and motor neuron dysfunction with upper motor neuron involvement. To investigate the role of UCHL1 on CSMN health and stability, we used both in vivo and in vitro approaches, and took advantage of the Uchl1(nm3419) (UCHL1(-/-)) mice, which lack all UCHL1 function. We report a unique role of UCHL1 in maintaining CSMN viability and cellular integrity. CSMN show early, selective, progressive, and profound cell loss in the absence of UCHL1. CSMN degeneration, evident even at pre-symptomatic stages by disintegration of the apical dendrite and spine loss, is mediated via increased ER stress. These findings bring a novel understanding to the basis of CSMN vulnerability, and suggest UCHL1(-/-) mice as a tool to study CSMN pathology.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Spinal Cord/cytology , Ubiquitin Thiolesterase/deficiency , Afferent Pathways/physiology , Age Factors , Animals , Animals, Newborn , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Motor Cortex/metabolism , Muscle Strength/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Ubiquitin Thiolesterase/genetics
2.
Brain Res ; 1550: 47-60, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24463035

ABSTRACT

BACKGROUND: Alpha-synuclein (SNCA) downregulation shows therapeutic potential for synucleinopathies, including Parkinson's disease (PD). Previously we showed that human (h)SNCA gene silencing using a short hairpin (sh)RNA in rat substantia nigra (SN) protects against a hSNCA-induced forelimb deficit, but not dopamine (DA) neuron loss. Furthermore, the shRNA increases cell death in vitro, but the same target sequence embedded in a microRNA30 transcript (mir30-hSNCA) does not. OBJECTIVE: Examine hSNCA gene silencing using mir30-hSNCA in vivo. METHODS: Rats were stereotaxically injected into one SN with adeno-associated virus serotype 2/8 (AAV)-hSNCA, AAV-hSNCA plus AAV-mir30-SNCA or AAV-hSNCA plus a control non-silencing mir30-embedded siRNA and DA neuron markers and associated behavior were examined. RESULTS: AAV2/8-mediated SN hSNCA expression induces a forelimb deficit and tyrosine hydroxylase-immunoreactive (TH-IR) neuron loss. hSNCA gene silencing using mir30-hSNCA protects against this forelimb deficit at 2 m and ameliorates TH-IR neuron loss. Striatal (ST) TH-IR fiber density and DA markers, assessed by western blot, are unaffected by AAV-hSNCA alone. Co-expression of either silencing vector reduces ST TH-IR fibers, panTH in SN and Ser40 phosphorylated TH in SN and ST, but does not affect vesicular monoamine transporter-2. However, hSNCA gene silencing promotes partial TH-IR fiber recovery by 2 m. Co-expression of either silencing vector also induces SN inflammation, although some recovery was observed by 2 m in hSNCA-silenced SN. CONCLUSION: hSNCA gene silencing with AAV-mir30-hSNCA has positive effects on forelimb behavior and SN DA neurons, which are compromised by inflammation and reduced TH expression, suggesting that AAV2/8-mir30-hSNCA-mediated gene silencing, although promising in vitro, is not a candidate for therapeutic translation for PD.


Subject(s)
MicroRNAs/metabolism , RNA Interference , Substantia Nigra/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dependovirus/genetics , Dopamine/metabolism , Forelimb/physiopathology , Gene Transfer Techniques , Genetic Vectors , Humans , Male , MicroRNAs/genetics , Motor Activity/physiology , Movement Disorders/metabolism , Movement Disorders/pathology , Neurons/metabolism , Neurons/pathology , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism , Vesicular Monoamine Transport Proteins/metabolism
3.
Gene ; 534(2): 400-7, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-23954875

ABSTRACT

In vitro neural differentiation of human embryonic stem cells (hESCs) is an advantageous system for studying early neural development. The process of early neural differentiation in hESCs begins by initiation of primitive neuroectoderm, which is manifested by rosette formation, with consecutive differentiation into neural progenitors and early glial-like cells. In this study, we examined the involvement of early neural markers - OTX2, PAX6, Sox1, Nestin, NR2F1, NR2F2, and IRX2 - in the onset of rosette formation, during spontaneous neural differentiation of hESC and human induced pluripotent stem cell (hiPSC) colonies. This is in contrast to the conventional way of studying rosette formation, which involves induction of neuronal differentiation and the utilization of embryoid bodies. Here we show that OTX2 is highly expressed at the onset of rosette formation, when rosettes comprise no more than 3-5 cells, and that its expression precedes that of established markers of early neuronal differentiation. Importantly, the rise of OTX2 expression in these cells coincides with the down-regulation of the pluripotency marker OCT4. Lastly, we show that cells derived from rosettes that emerge during spontaneous differentiation of hESCs or hiPSCs are capable of differentiating into dopaminergic neurons in vitro, and into mature-appearing pyramidal and serotonergic neurons weeks after being injected into the motor cortex of NOD-SCID mice.


Subject(s)
Cell Differentiation/genetics , Dopaminergic Neurons/physiology , Embryonic Stem Cells/physiology , Pluripotent Stem Cells/physiology , Rosette Formation/methods , Serotonergic Neurons/physiology , Animals , Biomarkers/metabolism , Cell Line , Dopaminergic Neurons/metabolism , Down-Regulation/genetics , Embryonic Stem Cells/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Motor Cortex/metabolism , Motor Cortex/physiology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Pluripotent Stem Cells/metabolism , Serotonergic Neurons/metabolism
4.
Endocrinology ; 154(6): 2166-73, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23584855

ABSTRACT

There is substantial evidence that age-related ovarian failure in rats is preceded by abnormal responsiveness of the neuroendocrine axis to estrogen positive feedback. Because IGF-I seems to act as a permissive factor for proper GnRH neuronal response to estrogen positive feedback and considering that the hypothalamic content of IGF-I declines in middle-aged (M-A) rats, we assessed the effectiveness of long-term IGF-I gene therapy in the mediobasal hypothalamus (MBH) of M-A female rats to extend regular cyclicity and preserve ovarian structure. We used 3 groups of M-A rats: 1 group of intact animals and 2 groups injected, at 36.2 weeks of age, in the MBH with either a bicistronic recombinant adeno-associated virus (rAAV) harboring the genes for IGF-I and the red fluorescent protein DsRed2, or a control rAAV expressing only DsRed2. Daily vaginal smears were taken throughout the study, which ended at 49.5 weeks of age. We measured serum levels of reproductive hormones and assessed ovarian histology at the end of the study. Although most of the rats injected with the IGF-I rAAV had, on the average, well-preserved estrous cyclicity as well as a generally normal ovarian histology, the intact and control rAAV groups showed a high percentage of acyclic rats at the end of the study and ovaries with numerous enlarged cysts and scarce corpora lutea. Serum LH was higher and hyperprolactinemia lower in the treated animals. These results suggest that overexpression of IGF-I in the MBH prolongs normal ovarian function in M-A female rats.


Subject(s)
Estrous Cycle/physiology , Genetic Therapy/methods , Hypothalamus/metabolism , Insulin-Like Growth Factor I/physiology , Ovary/physiology , Age Factors , Animals , Dependovirus/genetics , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HEK293 Cells , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Luteinizing Hormone/blood , Microscopy, Fluorescence , Ovary/anatomy & histology , Prolactin/blood , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
5.
Exp Neurol ; 247: 25-38, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23542820

ABSTRACT

Parkinson's disease (PD) is a prevalent neurodegenerative disorder characterized by a progressive and extensive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and their terminals in the striatum, which results in debilitating movement disorders. This devastating disease affects over 1 million individuals in the United States and is increasing in incidence worldwide. Currently available pharmacological and surgical therapies ameliorate clinical symptoms in the early stages of disease, but they cannot stop or reverse degeneration of DA neurons. Stem cell therapies have come to the forefront of the PD research field as promising regenerative therapies. The majority of preclinical stem cell studies in experimental models of PD are focused on the idea that stem cell-derived DA neurons could be developed for replacement of diseased neurons. Alternatively, our studies and the studies from other groups suggest that stem cells also have the potential to protect and stimulate regeneration of compromised DA neurons. This review is focused on strategies based on the therapeutic potential for PD of the neurotrophic and neuroregenerative properties of a subclass of stem cells, mesenchymal stem cells (MSCs).


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Nerve Degeneration/therapy , Parkinson Disease/therapy , Animals , Humans , Nerve Degeneration/epidemiology , Nerve Degeneration/etiology , Parkinson Disease/complications , Parkinson Disease/epidemiology
6.
Neurobiol Dis ; 47(2): 174-83, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22521461

ABSTRACT

Corticospinal motor neurons (CSMN) are the cortical component of motor neuron circuitry, which controls voluntary movement and degenerates in diseases such as amyotrophic lateral sclerosis, primary lateral sclerosis and hereditary spastic paraplegia. By using dual labeling combined with molecular marker analysis, we identified AAV2-2 mediated retrograde transduction as an effective approach to selectively target CSMN without affecting other neuron populations both in wild-type and hSOD1(G93A) transgenic ALS mice. This approach reveals very precise details of cytoarchitectural defects within vulnerable neurons in vivo. We report that CSMN vulnerability is marked by selective degeneration of apical dendrites especially in layer II/III of the hSOD1(G93A) mouse motor cortex, where cortical input to CSMN function is vastly modulated. While our findings confirm the presence of astrogliosis and microglia activation, they do not lend support to their direct role for the initiation of CSMN vulnerability. This study enables development of targeted gene replacement strategies to CSMN in the cerebral cortex, and reveals CSMN cortical modulation defects as a potential cause of neuronal vulnerability in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/virology , Dendrites/pathology , Dependovirus/physiology , Motor Neurons/pathology , Pyramidal Tracts/pathology , Signal Transduction/physiology , Amyotrophic Lateral Sclerosis/metabolism , Animals , Dendrites/chemistry , Dendrites/virology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/virology , Pyramidal Tracts/metabolism , Pyramidal Tracts/virology
7.
Dev Neurobiol ; 72(6): 906-17, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22021082

ABSTRACT

Experimental models of Parkinson's disease (PD) created by aberrant expression of the alpha-synuclein (SNCA) coding region have been reported. However, noncoding regions function in normal physiology and recent in vitro studies have shown that microRNAs-7 and -153 regulate SNCA expression by binding the 3'UTR. Here, effects of different hSNCA forms were examined in vivo. Adult, male rats were injected into one substantia nigra (SN) with AAV-wtSNCA, AAV-S129A hSNCA, or AAV-S129D hSNCA either with or without a portion of the native 3'UTR. DA neurons in SN that maintained striatal (ST) projections at the end of treatment were retrogradely labeled by bilateral ST fluorogold (FG) injections and FG-positive DA neurons in SN were counted. At 5 weeks, hSNCA coding vectors reduced numbers of FG-positive neurons in injected SN compared with uninjected SN (wtSNCA, p = 0.05; S129A/D hSNCA, p = 0.01). At 7 and 9 weeks, wtSNCA- and S129D hSNCA-treated rats exhibited recovery, but S129A hSNCA-injected rats did not (p = 0.01). In contrast, numbers of FG-positive neurons were unaffected by hSNCA expression when the 3'UTR was included. When FG-positive neurons were expressed as the ratio of numbers in injected to uninjected sides, the S129A hSNCA coding vector resulted in the highest decrease at 9 weeks versus wtSNCA (p = 0.05) or S129D hSNCA (p = 0.01). Inclusion of the 3'UTR resulted in no significant differences in FG-positive neuron ratios. These data suggest that inclusion of the 3'UTR protects against S129A hSNCA-induced loss of nigrostriatal-projecting DA neurons in vivo and that mis-regulation of hSNCA expression and function at noncoding regions contribute to PD pathogenesis.


Subject(s)
3' Untranslated Regions/genetics , Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , Nerve Degeneration/metabolism , Substantia Nigra/metabolism , alpha-Synuclein/genetics , Animals , Corpus Striatum/pathology , Disease Models, Animal , Dopaminergic Neurons/pathology , Genetic Therapy , Male , Nerve Degeneration/pathology , Neural Pathways/metabolism , Neural Pathways/pathology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phosphorylation , Rats , Rats, Sprague-Dawley , Substantia Nigra/pathology , alpha-Synuclein/metabolism
8.
Brain Res ; 1395: 94-107, 2011 Jun 13.
Article in English | MEDLINE | ID: mdl-21565333

ABSTRACT

Effects of silencing ectopically expressed hSNCA in rat substantia nigra (SN) were examined as a novel therapeutic approach to Parkinson's disease (PD). AAV-hSNCA with or without an AAV harboring a short-hairpin (sh)RNA targeting hSNCA or luciferase was injected into one SN. At 9weeks, hSNCA-expressing rats had reduced SN dopamine (DA) neurons and exhibited a forelimb deficit. AAV-shRNA-SNCA silenced hSNCA and protected against the forelimb deficit. However, AAV-shRNA-SNCA also led to DA neuron loss suggesting undesirable effects of chronic shRNA expression. Effects on nigrostriatal-projecting neurons were examined using a retrograde tract tracer. Loss of striatal-projecting DA neurons was evident in the vector injection site, whereas DA neurons outside this site were lost in hSNCA-expressing rats, but not in hSNCA-silenced rats. These observations suggest that high levels of shRNA-SNCA were toxic to DA neurons, while neighboring neurons exposed to lower levels were protected by hSNCA gene silencing. Also, data collected on DA levels suggest that neurons other than or in addition to nigrostriatal DA neurons contributed to protection of forelimb use. Our observations suggest that while hSNCA gene silencing in DA neurons holds promise as a novel PD therapy, further development of silencing technology is required.


Subject(s)
Dependovirus/genetics , Gene Silencing/physiology , Genetic Therapy/methods , Genetic Vectors/pharmacology , Genetic Vectors/toxicity , Parkinsonian Disorders/genetics , Parkinsonian Disorders/therapy , alpha-Synuclein/genetics , Animals , Disease Models, Animal , Dopamine/physiology , Forelimb/physiopathology , HEK293 Cells , Humans , Male , Neurons/metabolism , Neurons/pathology , Parkinsonian Disorders/physiopathology , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/genetics , RNA, Small Interfering/toxicity , Rats , Rats, Sprague-Dawley , Treatment Outcome , alpha-Synuclein/physiology
9.
Brain Res ; 1386: 15-24, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21338582

ABSTRACT

Alpha-synuclein (SNCA), an abundantly expressed presynaptic protein, is implicated in Parkinson's disease (PD). Since over-expression of human SNCA (hSNCA) leads to death of dopaminergic (DA) neurons in human, rodent and fly brain, hSNCA gene silencing may reduce levels of toxic forms of SNCA and ameliorate degeneration of DA neurons in PD. To begin to develop a gene therapy for PD based on hSNCA gene silencing, two AAV gene silencing vectors were designed, and tested for efficiency and specificity of silencing, as well as toxicity in vitro. The same hSNCA silencing sequence (shRNA) was used in both vectors, but in one vector, the shRNA was embedded in a microRNA backbone and driven by a pol II promoter, and in the other the shRNA was not embedded in a microRNA and was driven by a pol III promoter. Both vectors silenced hSNCA to the same extent in 293T cells transfected with hSNCA. In DA PC12 cells, neither vector decreased expression of rat SNCA, tyrosine hydroxylase (TH), dopamine transporter (DAT) or the vesicular monoamine transporter (VMAT). However, the mir30 embedded vector was significantly less toxic to both PC12 and SH-SY5Y cells. Our in vitro data suggest that this miRNA-embedded silencing vector may be ideal for chronic in vivo SNCA gene silencing in DA neurons.


Subject(s)
Dependovirus/genetics , Dopamine/metabolism , Gene Silencing/physiology , Genetic Vectors/toxicity , MicroRNAs/toxicity , Neurons/metabolism , alpha-Synuclein/antagonists & inhibitors , Animals , Cell Line, Tumor , Genetic Vectors/physiology , HEK293 Cells , Humans , Mice , MicroRNAs/genetics , Neurons/pathology , PC12 Cells , Parkinson Disease/genetics , Parkinson Disease/pathology , Parkinson Disease/therapy , Plasmids/genetics , Rats , alpha-Synuclein/genetics
10.
J Neurosci Res ; 88(12): 2669-81, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20544825

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive degeneration of nigrostriatal dopaminergic (DA) neurons. The therapeutic potential of glial cell line-derived neurotrophic factor (GDNF), the most potent neurotrophic factor for DA neurons, has been demonstrated in many experimental models of PD. However, chronic delivery of GDNF to DA neurons in the brain remains an unmet challenge. Here, we report the effects of GDNF-releasing Notch-induced human bone marrow-derived mesenchymal stem cells (MSC) grafted into striatum of the 6-hydroxydopamine (6-OHDA) progressively lesioned rat model of PD. Human MSC, obtained from bone marrow aspirates of young, healthy adult volunteers, were transiently transfected with the intracellular domain of the Notch1 gene (NICD) to generate SB623 cells. SB623 cells expressing GDNF and/or humanized Renilla green fluorescent protein (hrGFP) following lentiviral transduction or nontransduced cells were stereotaxically placed into rat striatum 1 week after a unilateral partial 6-OHDA striatal lesion. At 4 weeks, rats that had received GDNF-transduced SB623 cells had significantly decreased amphetamine-induced rotation compared with control rats, although this effect was not observed in rats that received GFP-transduced or nontransduced SB623 cells. At 5 weeks, rejuvenated tyrosine hydroxylase-immunoreactive (TH-IR) fibers that appeared to be host DA axons were observed in and around grafts. This effect was more prominent in rats that received GDNF-secreting cells and was not observed in controls. These observations suggest that human bone-marrow derived MSC, genetically modified to secrete GDNF, hold potential as an allogeneic or autologous stem cell therapy for PD.


Subject(s)
Bone Marrow Transplantation/methods , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Parkinsonian Disorders/pathology , Parkinsonian Disorders/therapy , Recovery of Function/physiology , Adult , Animals , Cell Differentiation/genetics , Cell Line , Disease Models, Animal , Genetic Engineering/methods , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/physiology , Graft Survival/physiology , Humans , Male , Nerve Regeneration/physiology , Parkinsonian Disorders/physiopathology , Rats , Rats, Inbred F344 , Transplantation, Autologous , Transplantation, Heterologous/methods , Transplantation, Homologous
11.
Restor Neurol Neurosci ; 28(3): 293-309, 2010.
Article in English | MEDLINE | ID: mdl-20479525

ABSTRACT

PURPOSE: The therapeutic potential of glial cell line-derived neurotrophic factor (GDNF) gene delivery was examined in a rodent model of traumatic brain injury (TBI), the controlled cortical impact (CCI). METHODS: An adenoviral vector harboring human GDNF (AdGDNF) or green fluorescent protein (AdGFP) was injected unilaterally into the forelimb sensorimotor cortex (FL-SMC) of the rat one week prior to a unilateral CCI. Tests of forelimb function and asymmetry were administered for 2 weeks post-injury. At 2 weeks post-injury, animals were sacrificed and contusion size, neuronal survival, neurodegeneration, and virally-mediated GDNF and GFP protein expression were measured. RESULTS: Rats injected with AdGDNF had significantly smaller contusions, more surviving neurons, and less neurodegeneration than AdGFP injected and uninjected injured animals. GDNF gene delivery also resulted in significantly faster recovery of forelimb coordination and a smaller initial preference for the uninjured forelimb during exploration of the walls of a platform. However, overall recovery of symmetrical forelimb use was not achieved. CONCLUSIONS: The discrepancy between neural protection and behavioral recovery suggests that while GDNF gene delivery provided a high degree of protection of damaged cortical neurons in this model of TBI, it may not have fully protected their terminals and synaptic functioning, resulting in only mild protection against behavioral deficits.


Subject(s)
Brain Injuries/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/genetics , Neuroprotective Agents/administration & dosage , Adenoviridae/genetics , Animals , Brain Injuries/genetics , Brain Injuries/pathology , Disease Models, Animal , Genetic Vectors/administration & dosage , Lameness, Animal/etiology , Lameness, Animal/physiopathology , Lameness, Animal/therapy , Male , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Degeneration/therapy , Neuroprotective Agents/metabolism , Rats , Rats, Inbred F344 , Recovery of Function/genetics
12.
Cell Transplant ; 18(7): 801-14, 2009.
Article in English | MEDLINE | ID: mdl-19796495

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disease characterized by the selective loss of dopaminergic (DA) neurons in the midbrain. Various types of stem cells that have potential to differentiate into DA neurons are being investigated as cellular therapies for PD. Stem cells also secrete growth factors and therefore also may have therapeutic effects in promoting the health of diseased DA neurons in the PD brain. To address this possibility in an experimental model of PD, bone marrow-derived neuroprogenitor-like cells were generated from bone marrow procured from healthy human adult volunteers and their potential to elicit recovery of damaged DA axons was studied in a partial lesion rat model of PD. Following collection of bone marrow, mesenchymal stem cells (MSC) were isolated and then genetically modified to create SB623 cells by transient transfection with the intracellular domain of the Notch1 gene (NICD), a modification that upregulates expression of certain neuroprogenitor markers. Ten deposits of 0.5 microl of SB623 cell suspension adjusted from 6,000 to 21,000 cells/microl in PBS or PBS alone were stereotaxically placed in the striatum 1 week after the nigrostriatal projection had been partially lesioned in adult F344 rats by injection of 6-hydroxydopamine (6-OHDA) into the striatum. At 3 weeks, a small number of grafted SB623 cells survived in the lesioned striatum as visualized by expression of the human specific nuclear matrix protein (hNuMA). In rats that received SB623 cells, but not in control rats, dense tyrosine hydroxylase immunoreactive (TH-ir) fibers were observed around the grafts. These fibers appeared to be rejuvenated host DA axons because no TH-ir in soma of surviving SB623 cells or coexpression of TH and hNuMA-ir were observed. In addition, dense serotonin immunoreactive (5-HT-ir) fibers were observed around grafted SB623 cells and these fibers also appeared to be of the host origin. Also, in some SB623 grafted rats that were sacrificed within 2 h of dl-amphetamine injection, hot spots of c-Fos-positive nuclei that coincided with rejuvenated dense TH fibers around the grafted SB623 cells were observed, suggesting increased availability of DA in these locations. Our observations suggest that NICD-transfected MSC hold potential as a readily available autologous or allogenic cellular therapy for ameliorating the degeneration of DA and 5-HT neurons in PD patients.


Subject(s)
Dopamine , Mesenchymal Stem Cell Transplantation , Nerve Degeneration/therapy , Neurons/physiology , Parkinsonian Disorders/therapy , Amphetamine/metabolism , Amphetamine/pharmacology , Animals , Bone Marrow Cells/cytology , Cell Communication , Cell Line , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Humans , Male , Mesencephalon/metabolism , Mesencephalon/pathology , Neostriatum/cytology , Nerve Fibers/metabolism , Neurons/cytology , Parkinsonian Disorders/pathology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Inbred F344 , Serotonin/metabolism , Substantia Nigra/metabolism , Substantia Nigra/pathology , Transfection , Tyrosine 3-Monooxygenase/metabolism
13.
Tissue Eng Part C Methods ; 14(3): 207-19, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18721070

ABSTRACT

Recent advances in tissue engineering that combine an extracellular matrix-like scaffold with therapeutic molecules, cells, DNA encoding therapeutic proteins, or a combination of the three hold promise for treating defects in the brain resulting from a penetrating injury or tumor resection. The purpose of this study was to investigate a porous sponge-like collagen scaffold for non-viral delivery of a plasmid encoding for glial cell line-derived neurotrophic factor (pGDNF) to rat marrow stromal stem cells (also referred to as mesenchymal stem cells, MSCs). The effects of the following parameters on GDNF synthesis in the three-dimensional (3D) constructs were evaluated and compared with results in monolayer culture: initial plasmid load (2-50 microg pGDNF), ratio of a lipid transfection reagent to plasmid (5:10), culture environment during the transfection (static and dynamic), and cell density. The level of gene expression in the collagen scaffolds achieved therapeutic levels that had previously been found to support survival of dopaminergic and trigeminal neurons in vitro. For the highest loading of plasmid (50 microg), the level of GDNF protein remained six to seven times above the control level after 2 weeks, a significant difference. Cell density in the scaffold was of importance for an early increase in GDNF production, with accumulated GDNF being approximately 60% greater after 9 days of culture when scaffolds were initially seeded with 2 million rat MSCs compared to 500,000 cells. Application of orbital shaking during the 4 h of transfection had a positive effect on the production of GDNF on 3D constructs but not of the same magnitude as reported in monolayer studies. Overall, these results demonstrate that the combination of tissue engineering and non-viral transfection of MSCs for the over-expression of GDNF is a promising approach for the long-term production of GDNF and probably for neurotrophic factors in general.


Subject(s)
Collagen/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mesenchymal Stem Cells/cytology , Animals , Cell Line , Cell Proliferation , Cells, Cultured , DNA/metabolism , Gene Transfer Techniques , In Vitro Techniques , Models, Biological , Models, Genetic , Neuroglia/cytology , Plasmids/metabolism , Rats , Rats, Inbred Lew
14.
J Neurosci Res ; 86(2): 317-25, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17787016

ABSTRACT

6-Hydroxydopamine (6-OHDA), a neurotoxin that causes the death of dopamine (DA) neurons, is commonly used to produce experimental models of Parkinson's disease (PD) in rodents. In the rat model of PD first described by Sauer and Oertel, DA neurons progressively die over several weeks following a striatal injection of 6-OHDA. It is generally assumed that DA neurons die through apoptosis after exposure to 6-OHDA, but data supporting activation of a caspase enzymatic cascade are lacking. In this study, we sought to determine if caspases involved in the intrinsic apoptotic cascade play a role in the initial stages of 6-OHDA-induced death of DA neurons in the progressively lesioned rat model of PD. We found that injection of 6-OHDA into adult rat striatum did not activate caspase-9 or caspase-3 or increase levels of caspase-dependent cleavage products in the substantia nigra at various survival times up to 7 days after the lesion, even though this paradigm produced DA neuronal loss. These data suggest that in the adult rat brain DA neurons whose terminals are challenged with 6-OHDA do not die through a classical caspase-dependent apoptotic mechanism.


Subject(s)
Adrenergic Agents/pharmacology , Caspases/drug effects , Nerve Degeneration/chemically induced , Neurons/pathology , Oxidopamine/pharmacology , Parkinsonian Disorders/pathology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain/metabolism , Brain/pathology , Caspase 3/metabolism , Caspase 9/metabolism , Caspases/metabolism , Dopamine/metabolism , Enzyme Activation/physiology , Immunohistochemistry , Male , Nerve Degeneration/pathology , Parkinsonian Disorders/metabolism , Rats , Rats, Inbred F344
15.
Mol Ther ; 14(1): 69-78, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16650807

ABSTRACT

We investigated the movement of interstitially infused macromolecules within the central nervous system (CNS) in rats with high and low blood pressure (BP)/heart rate and in rats euthanized immediately before infusion (no heart action). Adeno-associated virus 2 (AAV2), fluorescent liposomes, or bovine serum albumin was infused into rat striatum (six hemispheres per group) by convection-enhanced delivery (CED). After infusion, distribution volumes were evaluated. The rats with high BP/heart rate displayed a significantly larger distribution of the infused molecules within the injected site and more extensive transport of those molecules to the globus pallidus. This difference was particularly apparent for AAV2, for which a 16.5-fold greater distribution of viral capsids was observed in the rats with high BP/heart rate than in the rats with no heartbeat. Similar results were observed for liposomes, despite their larger diameter. The distribution of all infused molecules in all rats that had low or no blood flow was confined to the space around brain blood vessels. These findings show that fluid circulation within the CNS through the perivascular space is the primary mechanism by which viral particles and other therapeutic agents administered by CED are spread within the brain and that cardiac contractions power this process.


Subject(s)
Blood Vessels/physiology , Brain/metabolism , Drug Delivery Systems/methods , Animals , Blood Pressure/physiology , Blood Vessels/metabolism , Convection , Dependovirus/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/pharmacokinetics , Heart Rate/physiology , Liposomes/administration & dosage , Liposomes/pharmacokinetics , Magnetic Resonance Angiography , Male , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/pharmacokinetics
16.
Exp Neurol ; 198(2): 382-90, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16455076

ABSTRACT

Human alpha-synuclein overexpression and its toxic accumulation in neurons or glia are known to play key roles in the pathogenesis of Parkinson's disease and other related neurodegenerative synucleinopathies. Several single point mutations in the alpha-synuclein gene, as well as gene duplication and triplication, have been linked to familial Parkinson's disease. Moreover, genetic variability of the alpha-synuclein gene promoter is associated with idiopathic Parkinson's disease. Silencing of the human alpha-synuclein gene by vector-based RNA interference (RNAi) is a promising therapeutic approach for synucleinopathies. Here, we report identification of a 21-nucleotide sequence in the coding region of human alpha-synuclein that constitutes an effective target for robust silencing by RNAi and demonstrate allele-specific silencing of the A53T mutant of human alpha-synuclein. Furthermore, we have developed a plasmid vector-based RNAi for silencing of human alpha-synuclein in vitro. Lastly, using a dual cassette lentivirus that co-expresses an alpha-synuclein-targeting small hairpin RNA (shRNA) and enhanced green fluorescent protein (EGFP) as a marker gene, we demonstrate effective silencing of endogenous human alpha-synuclein in vitro in the human dopaminergic cell line SH-SY5Y and also of experimentally expressed human alpha-synuclein in vivo in rat brain. Our results demonstrate potent silencing of human alpha-synuclein expression in vitro and in vivo by viral vector-based RNAi and provide the tools for developing effective gene silencing therapeutics for synucleinopathies, including Parkinson's disease.


Subject(s)
Brain/metabolism , Gene Expression Regulation/physiology , Lentivirus/physiology , RNA Interference/physiology , alpha-Synuclein/metabolism , Alleles , Animals , Brain/drug effects , Brain/virology , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Male , Mutant Proteins/physiology , Phosphopyruvate Hydratase/metabolism , RNA Interference/drug effects , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Transfection/methods , alpha-Synuclein/genetics
17.
Exp Neurol ; 191 Suppl 1: S80-94, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15629764

ABSTRACT

Caspase-9 is a critical downstream effector molecule involved in apoptosis, a cell death process thought to be involved in the demise of dopamine (DA) neurons in the substantia nigra (SN) affected by Parkinson's disease (PD). In this study, we determined that a tetracycline-regulated adenovirus harboring a dominant-negative form of caspase-9 (Casp9DN) and the marker gene, enhanced green fluorescent protein (EGFP), under the control of a bidirectional promoter could each be regulated in vitro and in vivo by doxycycline. We next observed that Casp9DN gene delivery significantly protected against TNFalpha and cycloheximide-induced chromatin condensation in HeLa cells and prevented chromatin condensation and the appearance of the early apoptotic marker annexin V in 6-hydroxydopamine (6-OHDA) treated MN9D cells, a dopaminergic cell line. Effects of Casp9DN on DA neurons in vivo were also assessed. DA neurons were retrogradely labeled with fluorogold (FG) and transduced with Casp9DN and EGFP or EGFP alone. A progressive lesion of DA neurons was induced by striatal injection of 6-OHDA 1 week later. At 2 weeks post-lesion, a morphometric analysis of FG+ neurons in the SN revealed that the mean cell diameter of FG labeled neurons in the Casp9DN group was 8% and 21% larger than the EGFP and PBS groups, respectively (P <0.05). However, there was no difference among the treatment groups in the number of neurons remaining in the lesioned SN. These results suggest that while inhibiting apoptosis at the level of caspase-9 is protective in vitro, it is not protective against 6-OHDA-induced cell death in vivo.


Subject(s)
Adenoviridae/genetics , Brain/metabolism , Caspases/genetics , Gene Expression Regulation/drug effects , Parkinsonian Disorders/metabolism , Tetracycline/pharmacology , Adenoviridae/drug effects , Animals , Apoptosis , Brain/drug effects , Brain/pathology , Caspase 9 , Caspases/metabolism , Cell Line , Gene Transfer Techniques , Genes, Dominant , Genes, Reporter , Genetic Vectors/drug effects , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Kidney/cytology , Kidney/metabolism , Male , Neostriatum/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins , Oxidopamine , Parkinsonian Disorders/pathology , Parkinsonian Disorders/therapy , Rats , Rats, Inbred F344
18.
Exp Neurol ; 190(2): 263-75, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15530868

ABSTRACT

This is a commentary on the developmental and therapeutic relevance of recent studies in the glial fibrillary acid protein (GFAP)-glial cell line-derived neurotrophic factor (GDNF) transgenic mouse reported by Zhao et al. (2004). This interesting study demonstrated that increased expression of GDNF in astrocytes increases the number of neighboring motoneurons of certain motoneuron subpopulations by diminishing programmed cell death during development. In addition, astrocyte-derived GDNF was shown to protect facial motoneurons from injury-induced cell death. Since this is the first direct demonstration that secretion of GDNF from astrocytes in the CNS can affect motoneuron development in utero and motoneuron survival after axotomy, novel approaches for motor neuron disease are suggested. The known target neurons that respond to GDNF are reviewed, as are studies using GDNF gene delivery in animal models of amyotrophic lateral sclerosis (ALS). It is postulated that GDNF is a factor to which many motoneurons respond along their whole extent from soma to axon to terminal.


Subject(s)
Motor Neurons/metabolism , Nerve Growth Factors/metabolism , Animals , Astrocytes/metabolism , Glial Cell Line-Derived Neurotrophic Factor , Humans , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...