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










Publication year range
1.
Brain Sci ; 13(2)2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36831872

ABSTRACT

Transplantation of immature dopaminergic neurons or neural precursors derived from embryonic stem cells (ESCs) into the substantia nigra pars compacta (SNpc) is a potential therapeutic approach for functional restitution of the nigrostriatal pathway in Parkinson's disease (PD). However, further studies are needed to understand the effects of the local microenvironment on the transplanted cells to improve survival and specific differentiation in situ. We have previously reported that the adult SNpc sustains a neurogenic microenvironment. Non-neuralized embryoid body cells (EBCs) from mouse ESCs (mESCs) overexpressing the dopaminergic transcription factor Lmx1a gave rise to many tyrosine hydroxylase (Th+) cells in the intact and damaged adult SNpc, although only for a short-term period. Here, we extended our study by transplanting EBCs from genetically engineered naive human ESC (hESC), overexpressing the dopaminergic transcription factors LMX1A, FOXA2, and OTX2 (hESC-LFO), in the SNpc. Unexpectedly, no graft survival was observed in wild-type hESC EBCs transplants, whereas hESC-LFO EBCs showed viability in the SNpc. Interestingly, neural rosettes, a developmental hallmark of neuroepithelial tissue, emerged at 7- and 15-days post-transplantation (dpt) from the hESC-LFO EBCs. Neural rosettes expressed specification dopaminergic markers (Lmx1a, Otx2), which gave rise to several Th+ cells at 30 dpt. Our results suggest that the SNpc enables the robust initiation of neural differentiation of transplanted human EBCs prompted to differentiate toward the midbrain dopaminergic phenotype.

2.
Int J Mol Sci ; 24(4)2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36834679

ABSTRACT

Parkinson's disease (PD) is the most common α-synucleinopathy worldwide. The pathognomonic hallmark of PD is the misfolding and propagation of the α-synuclein (α-syn) protein, observed in post-mortem histopathology. It has been hypothesized that α-synucleinopathy triggers oxidative stress, mitochondrial dysfunction, neuroinflammation, and synaptic dysfunction, leading to neurodegeneration. To this date, there are no disease-modifying drugs that generate neuroprotection against these neuropathological events and especially against α-synucleinopathy. Growing evidence suggests that peroxisome proliferator-activated receptor (PPAR) agonists confer neuroprotective effects in PD, however, whether they also confer an anti-α-synucleinopathy effect is unknown. Here we analyze the reported therapeutic effects of PPARs, specifically the gamma isoform (PPARγ), in preclinical PD animal models and clinical trials for PD, and we suggest possible anti-α-synucleinopathy mechanisms acting downstream from these receptors. Elucidating the neuroprotective mechanisms of PPARs through preclinical models that mimic PD as closely as possible will facilitate the execution of better clinical trials for disease-modifying drugs in PD.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Synucleinopathies , Animals , Parkinson Disease/metabolism , Peroxisome Proliferator-Activated Receptors , Neuroprotective Agents/therapeutic use , Neuroprotection , Disease Models, Animal
3.
Methods Mol Biol ; 2565: 17-33, 2023.
Article in English | MEDLINE | ID: mdl-36205884

ABSTRACT

Chromaffin cells are neuroendocrine cells that synthesize and release catecholamines and neuroactive molecules. They have been used experimentally in animal models and preclinical studies as a source for cell replacement therapy in Parkinson's disease. The long-term cell survival of these cells in the nervous system is limited, and the observed motor improvements are highly variable. An alternative source for transplantation is chromaffin progenitor cells. These cells have the capacity of self-renewal and to form spheres under low attachment conditions. They release higher quantities of dopamine than chromaffin cells and can differentiate into dopaminergic-like neurons in vitro. The transplantation of these cells into Parkinson's disease animal models has shown to induce stronger motor improvements and better survival rates than chromaffin cells. However, several aspects of chromaffin progenitor cell transplantation remain to be elucidated. Here, we describe methods to isolate and culture chromaffin and chromaffin progenitor cells from the adult cattle adrenal glands. We also describe the procedure for their transplantation into the nervous system and give recommendations for their histological analysis.


Subject(s)
Dopamine , Parkinson Disease , Animals , Catecholamines , Cattle , Central Nervous System , Stem Cell Transplantation
4.
Cells ; 11(21)2022 10 31.
Article in English | MEDLINE | ID: mdl-36359841

ABSTRACT

The complexity and overall burden of Parkinson's disease (PD) require new pharmacological approaches to counteract the symptomatology while reducing the progressive neurodegeneration of affected dopaminergic neurons. Since the pathophysiological signature of PD is characterized by the loss of physiological levels of dopamine (DA) and the misfolding and aggregation of the alpha-synuclein (α-syn) protein, new proposals seek to restore the lost DA and inhibit the progressive damage derived from pathological α-syn and its impact in terms of oxidative stress. In this line, nanomedicine (the medical application of nanotechnology) has achieved significant advances in the development of nanocarriers capable of transporting and delivering basal state DA in a controlled manner in the tissues of interest, as well as highly selective catalytic nanostructures with enzyme-like properties for the elimination of reactive oxygen species (responsible for oxidative stress) and the proteolysis of misfolded proteins. Although some of these proposals remain in their early stages, the deepening of our knowledge concerning the pathological processes of PD and the advances in nanomedicine could endow for the development of potential treatments for this still incurable condition. Therefore, in this paper, we offer: (i) a brief summary of the most recent findings concerning the physiology of motor regulation and (ii) the molecular neuropathological processes associated with PD, together with (iii) a recapitulation of the current progress in controlled DA release by nanocarriers and (iv) the design of nanozymes, catalytic nanostructures with oxidoreductase-, chaperon, and protease-like properties. Finally, we conclude by describing the prospects and knowledge gaps to overcome and consider as research into nanotherapies for PD continues, especially when clinical translations take place.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/metabolism , Nanomedicine , Dopaminergic Neurons/metabolism , Dopamine/metabolism , Drug Delivery Systems
5.
Methods Mol Biol ; 2520: 215-232, 2022.
Article in English | MEDLINE | ID: mdl-34611820

ABSTRACT

Human embryonic stem cell (hESC) and human-induced pluripotent stem cell (hiPSC) technologies have a critical role in regenerative strategies for personalized medicine. Both share the ability to differentiate into almost any cell type of the human body. The study of their properties and clinical applications requires the development of robust and reproducible cell culture paradigms that direct cell differentiation toward a specific phenotype in vitro and in vivo. Our group evaluated the potential of mouse ESCs (mESCs), hESCs, and hiPSCs (collectively named pluripotent stem cells, PSCs) to analyze brain microenvironments through the use of embryoid body (EB)-derived cells from these cell sources. EB are cell aggregates in 3D culture conditions that recapitulate embryonic development. Our approach focuses on studying the midbrain dopaminergic phenotype and transplanting EB into the substantia nigra pars compacta (SNpc) in a Parkinson's disease rodent model. Here, we describe cell culture protocols for EB generation from PSCs that show significant in vivo differentiation toward dopaminergic neurons.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Animals , Cell Differentiation/genetics , Embryoid Bodies , Embryonic Stem Cells , Humans , Mesencephalon , Mice
6.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830348

ABSTRACT

Dysfunction of cellular homeostasis can lead to misfolding of proteins thus acquiring conformations prone to polymerization into pathological aggregates. This process is associated with several disorders, including neurodegenerative diseases, such as Parkinson's disease (PD), and endoplasmic reticulum storage disorders (ERSDs), like alpha-1-antitrypsin deficiency (AATD) and hereditary hypofibrinogenemia with hepatic storage (HHHS). Given the shared pathophysiological mechanisms involved in such conditions, it is necessary to deepen our understanding of the basic principles of misfolding and aggregation akin to these diseases which, although heterogeneous in symptomatology, present similarities that could lead to potential mutual treatments. Here, we review: (i) the pathological bases leading to misfolding and aggregation of proteins involved in PD, AATD, and HHHS: alpha-synuclein, alpha-1-antitrypsin, and fibrinogen, respectively, (ii) the evidence linking each protein aggregation to the stress mechanisms occurring in the endoplasmic reticulum (ER) of each pathology, (iii) a comparison of the mechanisms related to dysfunction of proteostasis and regulation of homeostasis between the diseases (such as the unfolded protein response and/or autophagy), (iv) and clinical perspectives regarding possible common treatments focused on improving the defensive responses to protein aggregation for diseases as different as PD, and ERSDs.


Subject(s)
Afibrinogenemia/genetics , Fibrinogen/chemistry , Parkinson Disease/genetics , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin/chemistry , alpha-Synuclein/chemistry , Afibrinogenemia/drug therapy , Afibrinogenemia/metabolism , Afibrinogenemia/pathology , Animals , Autophagy/drug effects , Autophagy/genetics , Coagulants/therapeutic use , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Fibrinogen/genetics , Fibrinogen/metabolism , Gene Expression Regulation , Humans , Liver/metabolism , Liver/pathology , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protease Inhibitors/therapeutic use , Protein Aggregates/drug effects , Protein Folding/drug effects , Unfolded Protein Response/drug effects , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/drug therapy , alpha 1-Antitrypsin Deficiency/metabolism , alpha 1-Antitrypsin Deficiency/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
7.
Stem Cells Dev ; 28(16): 1104-1115, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31140356

ABSTRACT

Transplantation of dopaminergic (DA) cells into the striatum can rescue from dopamine deficiency in a Parkinson's disease condition, but this is not a suitable procedure for regaining the full control of motor activity. The minimal condition toward recovering the nigrostriatal pathway is the proper innervation of transplanted DA neurons or their precursors from the substancia nigra pars compacta (SNpc) to their target areas. However, functional integration of transplanted cells would require first that the host SNpc is suitable for their survival and/or differentiation. We recently reported that the intact adult SNpc holds a strong neurogenic environment, but primed embryonic stem cells (ie, embryoid body cells, EBCs) could not derive into DA neurons. In this study, we transplanted into the intact or lesioned SNpc, EBCs derived from embryonic stem cells that were prompt to differentiate into DA neurons by the forced expression of Lmx1a in neural precursor cells (R1B5/NesE-Lmx1a). We observed that, 6 days posttransplantation (dpt), R1B5 or R1B5/NesE-Lmx1a EBCs gave rise to Nes+ and Dcx+ cells within the host SNpc, but a large number of Th+ cells derived only from EBCs exogenously expressing Lmx1a. In contrast, when transplantation was carried out into the 6-hydroxidopamine-lesioned SNpc, the emergence of Th+ cells from EBCs was independent of exogenous Lmx1a expression, although these cells were not found by 15 dpt. These results suggest that the adult SNpc is not only a permissive niche for initiation of DA differentiation of non-neuralized cells but also releases factors upon damage that promote the acquisition of DA characteristics by transplanted EBCs.


Subject(s)
Cell Differentiation/physiology , Dopamine/metabolism , Embryonic Stem Cells/cytology , Substantia Nigra/cytology , Animals , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Doublecortin Protein , Embryonic Stem Cells/metabolism , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Transcription Factors/metabolism
8.
Neuroscience ; 408: 308-326, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31034794

ABSTRACT

Neurogenesis in the substantia nigra (SN) has been a controversial issue. Here we report that neurogenesis can be induced in the adult rodent SN by transplantation of embryoid body cells (EBCs) derived from mouse embryonic stem cells. The detection of Sox2+ dividing (BrdU+) putative host neural precursor cells (NPCs) between 1 and 6 days post-transplantation (dpt) supported the neurogenic capacity of the adult SN. In agreement with the awakening of NPCs by EBCs, only host cells from implant-bearing SN were able to generate neurosphere-like aggregates in the presence of Egf and Fgf2. Later, at 15 dpt, a significant number of SN Dcx+ neuroblasts were detected. However, a continuous BrdU administration after transplantation showed that only a fraction (about 20-30%) of those host Dcx+ progeny derived from dividing cells and few BrdU+ cells, some of them NeuN+, survived up to 30 dpt. Unexpectedly, 25-30% of Dcx+ or Psa-Ncam+ cells at 15 dpt displayed astrocytic markers such as Gfap and S100b. Using a genetic lineage tracing strategy, we demonstrated that a large proportion of host Dcx+ and/or Tubb3+ neuroblasts originated from Gfap+ cells. Remarkably, new blood vessels formed in association with the neurogenic process that, when precluded, caused a reduction in neuroblast production. Accordingly, two proteins secreted by EBCs, Fgf2 and Vegf, were able to promote the emergence of Dcx+/Psa-Ncam+, Tubb3+ and NeuN+/BrdU+ cells in vivo in the absence of EBCs. We propose that the adult SN is a mostly silent neurogenic niche with the ability to generate new neurons by typical and atypical mechanisms.


Subject(s)
Mouse Embryonic Stem Cells/transplantation , Neurogenesis/physiology , Neurons/physiology , Substantia Nigra/physiology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Doublecortin Protein , Fibroblast Growth Factor 2/pharmacology , Mice , Mouse Embryonic Stem Cells/drug effects , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , Rats , Rats, Wistar , Substantia Nigra/cytology , Substantia Nigra/drug effects , Vascular Endothelial Growth Factor A/pharmacology
9.
Front Neurosci ; 13: 1399, 2019.
Article in English | MEDLINE | ID: mdl-32038126

ABSTRACT

Alpha-synuclein (α-syn) is localized in cellular organelles of most neurons, but many of its physiological functions are only partially understood. α-syn accumulation is associated with Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy as well as other synucleinopathies; however, the exact pathomechanisms that underlie these neurodegenerative diseases remain elusive. In this review, we describe what is known about α-syn function and pathophysiological changes in different cellular structures and organelles, including what is known about its behavior as a prion-like protein. We summarize current knowledge of α-syn and its pathological forms, covering its effect on each organelle, including aggregation and toxicity in different model systems, with special interest on the mitochondria due to its relevance during the apoptotic process of dopaminergic neurons. Moreover, we explore the effect that α-syn exerts by interacting with chromatin remodeling proteins that add or remove histone marks, up-regulate its own expression, and resume the impairment that α-syn induces in vesicular traffic by interacting with the endoplasmic reticulum. We then recapitulate the events that lead to Golgi apparatus fragmentation, caused by the presence of α-syn. Finally, we report the recent findings about the accumulation of α-syn, indirectly produced by the endolysosomal system. In conclusion, many important steps into the understanding of α-syn have been made using in vivo and in vitro models; however, the time is right to start integrating observational studies with mechanistic models of α-syn interactions, in order to look at a more complete picture of the pathophysiological processes underlying α-synucleinopathies.

10.
World J Transplant ; 7(3): 179-192, 2017 Jun 24.
Article in English | MEDLINE | ID: mdl-28698835

ABSTRACT

Cell grafting has been considered a therapeutic approach for Parkinson's disease (PD) since the 1980s. The classical motor symptoms of PD are caused by the loss of dopaminergic neurons in the substantia nigra pars compacta, leading to a decrement in dopamine release in the striatum. Consequently, the therapy of cell-transplantation for PD consists in grafting dopamine-producing cells directly into the brain to reestablish dopamine levels. Different cell sources have been shown to induce functional benefits on both animal models of PD and human patients. However, the observed motor improvements are highly variable between individual subjects, and the sources of this variability are not fully understood. The purpose of this review is to provide a general overview of the pioneering studies done in animal models of PD that established the basis for the first clinical trials in humans, and compare these with the latest findings to identify the most relevant aspects that remain unanswered to date. The main focus of the discussions presented here will be on the mechanisms associated with the survival and functionality of the transplants. These include the role of the dopamine released by the grafts and the capacity of the grafted cells to extend fibers and to integrate into the motor circuit. The complete understanding of these aspects will require extensive research on basic aspects of molecular and cellular physiology, together with neuronal network function, in order to uncover the real potential of cell grafting for treating PD.

11.
Dev Biol ; 429(1): 56-70, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28733161

ABSTRACT

The ventral mesencephalic neural precursor cells (vmNPCs) that give rise to dopaminergic (DA) neurons have been identified by the expression of distinct genes (e.g., Lmx1a, Foxa2, Msx1/2). However, the commitment of these NPCs to the mesencephalic DA neuronal fate has not been functionally determined. Evaluation of the plasticity of vmNPCs suggests that their commitment occurs after E10.5. Here we show that E9.5 vmNPCs implanted in an ectopic area of E10.5 mesencephalic explants, retained their specification marker Lmx1a and efficiently differentiated into neurons but did not express the gene encoding tyrosine hydroxylase (Th), the limiting enzyme for dopamine synthesis. A proportion of E10.5-E11.5 implanted vmNPCs behaved as committed, deriving into Th+ neurons in ectopic sites. Interestingly, implanted cells from E12.5 embryos were unable to give rise to a significant number of Th+ neurons. Concomitantly, differentiation assays in culture and in mesencephalic explants treated with Fgf2+LIF detected vmNPCs with astrogenic potential since E11.5. Despite this, a full suspension of E12.5 vmNPCs give rise to DA neurons in a similar proportion as those of E10.5 when they were transplanted into adult brain, but astrocytes were only detected with the former population. These data suggest that the subventricular postmitotic progenitors present in E12.5 ventral mesencephalon are unable to implant in embryonic explants and are the source of DA neurons in the transplanted adult brain. Based on our findings we propose that during DA differentiation committed vmNPCs emerge at E10.5 and they exhaust their neurogenic capacity with the rise of NPCs with astrogenic potential.


Subject(s)
Cell Differentiation , Dopaminergic Neurons/cytology , Mesencephalon/cytology , Neural Stem Cells/cytology , Neurogenesis , Animals , Astrocytes/cytology , Cell Lineage , Cell Proliferation , Cell Survival , Dopaminergic Neurons/metabolism , Embryo, Mammalian/cytology , Female , Hedgehog Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Male , Mice, Transgenic , Models, Biological , Neural Stem Cells/metabolism , Rats, Wistar , Stem Cell Niche , Transcription Factors/metabolism
12.
PLoS One ; 11(8): e0160854, 2016.
Article in English | MEDLINE | ID: mdl-27525967

ABSTRACT

Cell replacement therapy in Parkinson's disease (PD) aims at re-establishing dopamine neurotransmission in the striatum by grafting dopamine-releasing cells. Chromaffin cell (CC) grafts produce some transitory improvements of functional motor deficits in PD animal models, and have the advantage of allowing autologous transplantation. However, CC grafts have exhibited low survival, poor functional effects and dopamine release compared to other cell types. Recently, chromaffin progenitor-like cells were isolated from bovine and human adult adrenal medulla. Under low-attachment conditions, these cells aggregate and grow as spheres, named chromospheres. Here, we found that bovine-derived chromosphere-cell cultures exhibit a greater fraction of cells with a dopaminergic phenotype and higher dopamine release than CC. Chromospheres grafted in a rat model of PD survived in 57% of the total grafted animals. Behavioral tests showed that surviving chromosphere cells induce a reduction in motor alterations for at least 3 months after grafting. Finally, we found that compared with CC, chromosphere grafts survive more and produce more robust and consistent motor improvements. However, further experiments would be necessary to determine whether the functional benefits induced by chromosphere grafts can be improved, and also to elucidate the mechanisms underlying the functional effects of the grafts.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Chromaffin Cells/cytology , Chromaffin Cells/transplantation , Neostriatum/metabolism , Oxidopamine/pharmacology , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Animals , Cattle , Chromaffin Cells/metabolism , Disease Models, Animal , Dopamine/metabolism , Male , Motor Activity , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phenotype , Rats , Rats, Wistar , Stem Cell Transplantation , Survival Analysis
13.
Biol Open ; 5(3): 336-47, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26912775

ABSTRACT

Notch signalling is a well-established pathway that regulates neurogenesis. However, little is known about the role of Notch signalling in specific neuronal differentiation. Using Dll1 null mice, we found that Notch signalling has no function in the specification of mesencephalic dopaminergic neural precursor cells (NPCs), but plays an important role in regulating their expansion and differentiation into neurons. Premature neuronal differentiation was observed in mesencephalons of Dll1-deficient mice or after treatment with a Notch signalling inhibitor. Coupling between neurogenesis and dopaminergic differentiation was indicated from the coincident emergence of neuronal and dopaminergic markers. Early in differentiation, decreasing Notch signalling caused a reduction in NPCs and an increase in dopaminergic neurons in association with dynamic changes in the proportion of sequentially-linked dopaminergic NPCs (Msx1/2+, Ngn2+, Nurr1+). These effects in differentiation caused a significant reduction in the number of dopaminergic neurons produced. Accordingly, Dll1 haploinsufficient adult mice, in comparison with their wild-type littermates, have a consistent reduction in neuronal density that was particularly evident in the substantia nigra pars compacta. Our results are in agreement with a mathematical model based on a Dll1-mediated regulatory feedback loop between early progenitors and their dividing precursors that controls the emergence and number of dopaminergic neurons.

14.
Stem Cells ; 33(2): 491-502, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25267362

ABSTRACT

A neurogenic niche can be identified by the proliferation and differentiation of its naturally residing neural stem cells. However, it remains unclear whether "silent" neurogenic niches or regions suitable for neural differentiation, other than the areas of active neurogenesis, exist in the adult brain. Embryoid body (EB) cells derived from embryonic stem cells (ESCs) are endowed with a high potential to respond to specification and neuralization signals of the embryo. Hence, to identify microenvironments in the postnatal and adult rat brain with the capacity to support neuronal differentiation, we transplanted dissociated EB cells to conventional neurogenic and non-neurogenic regions. Our results show a neuronal differentiation pattern of EB cells that was dependent on the host region. Efficient neuronal differentiation of EB cells occurred within an adjacent region to the rostral migratory stream. EB cell differentiation was initially patchy and progressed toward an even distribution along the graft by 15-21 days post-transplantation, giving rise mostly to GABAergic neurons. EB cells in the striatum displayed a lower level of neuronal differentiation and derived into a significant number of astrocytes. Remarkably, when EB cells were transplanted to the striatum of adult rats after a local ischemic stroke, increased number of neuroblasts and neurons were observed. Unexpectedly, we determined that the adult substantia nigra pars compacta, considered a non-neurogenic area, harbors a robust neurogenic environment. Therefore, neurally uncommitted cells derived from ESCs can detect regions that support neuronal differentiation within the adult brain, a fundamental step for the development of stem cell-based replacement therapies.


Subject(s)
Cell Differentiation , Corpus Striatum/metabolism , Embryonic Stem Cells/metabolism , GABAergic Neurons/metabolism , Stem Cell Niche , Stem Cell Transplantation , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/therapy , Cell Line , Corpus Striatum/pathology , Embryonic Stem Cells/pathology , GABAergic Neurons/pathology , Heterografts , Male , Mice , Rats , Rats, Wistar , Stroke/metabolism , Stroke/pathology , Stroke/therapy
15.
CNS Neurol Disord Drug Targets ; 11(7): 897-906, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23131151

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNc) and its projections. Reports show a lower incidence of PD in smokers compared to nonsmokers. Nicotine reduce motor symptoms of patients already diagnosed with PD. However, the mechanisms underlying the effects of nicotine in the dopamine (DA) depleted striatum remain elusive. This study evaluates the effects of chronic nicotine administration on PD motor symptoms in an attempt to mimic the chronic self-administration of nicotine in smokers. To achieve this, we used the 6-OHDA hemiparkinson rat model evaluating the amphetamine/apomorphine induced circling behavior, in rats whose daily water intake included nicotine. We found that chronic nicotine reduced amphetamine (AMPH) induced circling behavior by 40%, whereas apomorphine (APO) increased this behavior by 230%. High-performance liquid chromatography (HPLC) revealed that AMPH produced a 50% decrease of DA release in the intact hemisphere, while on the striatum of the lesioned side, receptor binding assays showed an increased affinity to D1 receptors and a concurrent decrease in D2 receptors. c-Fos activity showed through double labeling, that cell types involved in nicotine action were low threshold (LTS) and fast spiking (FS) inter-neurons, which increased in the DA-depleted striatum. We also observed an increase in the activity of D1 medium spiny neurons (D1 MSN), a striatal population with a major role in motor control. Our results show that chronic nicotine does not specifically protect against degeneration, but rather modifies DA receptor dynamics, suggesting that it could be used as a therapeutic element in PD pathology.


Subject(s)
Antiparkinson Agents/therapeutic use , Corpus Striatum/drug effects , Disease Models, Animal , Interneurons/drug effects , Neuroprotective Agents/therapeutic use , Nicotine/therapeutic use , Parkinson Disease/prevention & control , Amphetamine/adverse effects , Animals , Apomorphine/therapeutic use , Behavior, Animal/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine/metabolism , Dopamine Agents/therapeutic use , Dopamine D2 Receptor Antagonists , Dopamine Uptake Inhibitors/adverse effects , Interneurons/metabolism , Male , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Oxidopamine , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rats , Rats, Wistar , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology
16.
Front Neurol ; 2: 26, 2011.
Article in English | MEDLINE | ID: mdl-21541306

ABSTRACT

Narcolepsy is a chronic neurodegenerative disease caused by a deficiency of orexin-producing neurons in the lateral hypothalamus. It is clinically characterized by excessive daytime sleepiness and by intrusions into wakefulness of physiological aspects of rapid eye movement sleep such as cataplexy, sleep paralysis, and hypnagogic hallucinations. The major pathophysiology of narcolepsy has been recently described on the bases of the discovery of the neuropeptides named orexins (hypocretins) in 1998; considerable evidence, summarized below, demonstrates that narcolepsy is the result of alterations in the genes involved in the pathology of the orexin ligand or its receptor. Deficient orexin transmission is sufficient to produce narcolepsy, as we describe here, animal models with dysregulated orexin signaling exhibit a narcolepsy-like phenotype. Remarkably, these narcoleptic models have different alterations of the orexinergic circuit, this diversity provide us with the means for making comparison, and have a better understanding of orexin-cell physiology. It is of particular interest that the most remarkable findings regarding this sleep disorder were fortuitous and due to keen observations. Sleep is a highly intricate and regulated state, and narcolepsy is a disorder that still remains as one of the unsolved mysteries in science. Nevertheless, advances and development of technology in neuroscience will provide us with the necessary tools to unravel the narcolepsy puzzle in the near future. Through an evaluation of the scientific literature we traced an updated picture of narcolepsy and orexins in order to provide insight into the means by which neurobiological knowledge is constructed.

17.
BMC Genomics ; 12: 222, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21569245

ABSTRACT

BACKGROUND: During murine hypothalamic development, different neuroendocrine cell phenotypes are generated in overlapping periods; this suggests that cell-type specific developmental programs operate to achieve complete maturation. A balance between programs that include cell proliferation, cell cycle withdrawal as well as epigenetic regulation of gene expression characterizes neurogenesis. Thyrotropin releasing hormone (TRH) is a peptide that regulates energy homeostasis and autonomic responses. To better understand the molecular mechanisms underlying TRH neuron development, we performed a genome wide study of its transcriptome during fetal hypothalamic development. RESULTS: In primary cultures, TRH cells constitute 2% of the total fetal hypothalamic cell population. To purify these cells, we took advantage of the fact that the segment spanning -774 to +84 bp of the Trh gene regulatory region confers specific expression of the green fluorescent protein (GFP) in the TRH cells. Transfected TRH cells were purified by fluorescence activated cell sorting, various cell preparations pooled, and their transcriptome compared to that of GFP- hypothalamic cells. TRH cells undergoing the terminal phase of differentiation, expressed genes implicated in protein biosynthesis, intracellular signaling and transcriptional control. Among the transcription-associated transcripts, we identified the transcription factors Klf4, Klf10 and Atf3, which were previously uncharacterized within the hypothalamus. CONCLUSION: To our knowledge, this is one of the first reports identifying transcripts with a potentially important role during the development of a specific hypothalamic neuronal phenotype. This genome-scale study forms a rational foundation for identifying genes that might participate in the development and function of hypothalamic TRH neurons.


Subject(s)
Fetus/cytology , Fetus/metabolism , Gene Expression Profiling , Hypothalamus/metabolism , Neurons/metabolism , Thyrotropin-Releasing Hormone/genetics , Animals , Embryo, Mammalian , Hypothalamus/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Neurons/cytology , Oligonucleotide Array Sequence Analysis , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
18.
J Neurosci Methods ; 127(2): 179-92, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12906947

ABSTRACT

Analysis of gene regulatory sequences in primary cultures of neurons has been hampered by inefficient transfection of post-mitotic neurons with reporter plasmids. We describe detailed conditions that allowed a significant improvement of transfection efficiency in primary cultures of serum-supplemented rat fetal hypothalamic cells. Transfected cells expressed the green fluorescent protein (GFP) under the control of the strong but non-cell-specific cytomegalo virus (CMV) promoter or under the thyrotropin-releasing hormone (TRH) promoter, to direct expression only in TRH neurons. Using the CMV promoter-GFP plasmid, we tested several commercially available transfection reagents; the best results were obtained with polyethylenimine (PEI) and Lipofectamine 2000. We optimized the transfection procedure with PEI because it rendered more reproducible results. Transfection with PEI was optimal when cells were transfected at a cellular density of 2.9 x 10(6) cells in 35-mm dishes, with 10 microg of DNA, a PEI/DNA ratio of 8.8 and PEI pH of 6.9. Using these conditions, we were able to detect GFP positive neurons after transfecting the TRH promoter-GFP plasmid. GFP positive cells were successfully purified by FACS. This opens the possibility to use transfection of mammalian CNS post-mitotic neurons for new applications including the purification of specific neuronal subtypes.


Subject(s)
Hypothalamus/cytology , Neurons/drug effects , Polyethyleneimine/pharmacology , Transfection/methods , 3T3 Cells , Animals , Cell Count , Cell Size , Cell Survival , Cells, Cultured , Cytomegalovirus/metabolism , Embryo, Mammalian , Female , Flow Cytometry/instrumentation , Flow Cytometry/methods , Green Fluorescent Proteins , Hypothalamus/virology , In Situ Hybridization/methods , Luciferases/metabolism , Luminescent Proteins/metabolism , Mice , Neurons/physiology , Neurons/virology , Pregnancy , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Thyrotropin-Releasing Hormone/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...