Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Signal ; 8(406): ra125, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26645582

ABSTRACT

Glioblastomas are aggressive astrocytomas characterized by endothelial cell proliferation and abnormal vasculature, which can cause brain edema and increase patient morbidity. We identified the heparin-binding cytokine pleiotrophin as a driver of vascular abnormalization in glioma. Pleiotrophin abundance was greater in high-grade human astrocytomas and correlated with poor survival. Anaplastic lymphoma kinase (ALK), which is a receptor that is activated by pleiotrophin, was present in mural cells associated with abnormal vessels. Orthotopically implanted gliomas formed from GL261 cells that were engineered to produce pleiotrophin showed increased microvessel density and enhanced tumor growth compared with gliomas formed from control GL261 cells. The survival of mice with pleiotrophin-producing gliomas was shorter than that of mice with gliomas that did not produce pleiotrophin. Vessels in pleiotrophin-producing gliomas were poorly perfused and abnormal, a phenotype that was associated with increased deposition of vascular endothelial growth factor (VEGF) in direct proximity to the vasculature. The growth of pleiotrophin-producing GL261 gliomas was inhibited by treatment with the ALK inhibitor crizotinib, the ALK inhibitor ceritinib, or the VEGF receptor inhibitor cediranib, whereas control GL261 tumors did not respond to either inhibitor. Our findings link pleiotrophin abundance in gliomas with survival in humans and mice, and show that pleiotrophin promotes glioma progression through increased VEGF deposition and vascular abnormalization.


Subject(s)
Astrocytoma/metabolism , Astrocytoma/mortality , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Carrier Proteins/metabolism , Cytokines/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/mortality , Anaplastic Lymphoma Kinase , Animals , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Cytokines/genetics , Disease-Free Survival , Female , Humans , Male , Mice , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Survival Rate , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
Neuroimage ; 61(1): 228-39, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22406356

ABSTRACT

Large increases in regional cerebral blood flow (rCBF) have been measured in patients with Parkinson's disease (PD) following the administration of L-DOPA, but the underlying mechanisms have remained unknown. In this study, rats with unilateral 6-hydroxydopamine (6-OHDA) lesions were used to compare patterns of rCBF and regional cerebral glucose utilisation (rCGU) in chronically L-DOPA-treated subjects following a final injection of L-DOPA or saline. The same animal model was used to the leakage of a blood-brain barrier (BBB) tracer molecule at 60 min vs. 24h following the last L-DOPA injection of a chronic treatment. All the parameters under investigation were examined with brain autoradiography following intravenous injections of specific radiotracers in awake animals ([14C]-iodoantipyrine for rCBF, [14C]-2-deoxyglucose for rCGU, and [14C]-α-aminoisobutyric acid for BBB leakage). Significant changes in rCBF and rCGU on the side ipsilateral to the 6-OHDA lesion relative to the non-lesioned side were seen at 60 min ("ON") but not 24h ("OFF") following L-DOPA administration. These changes were not seen in sham-operated rats. In the output nuclei of the basal ganglia (the entopeduncular nucleus and the substantia nigra pars reticulata) both rCBF and rCGU were elevated both in acutely L-DOPA-treated rats and chronically L-DOPA-treated rats displaying dyskinesia, but did not change significantly in chronically L-DOPA-treated non-dyskinetic cases. Acutely and chronically L-DOPA-treated rats with dyskinesia exhibited increases in rCBF "ON L-DOPA" also in the motor cortex, the striatum, and the globus pallidus, but the corresponding changes in rCGU did not show the same direction, magnitude, and/or relative group differences. The uptake of a BBB tracer (studied in the striatum and the substantia nigra reticulata in chronically L-DOPA treated rats) was significantly higher ON vs. OFF L-DOPA. The present results are the first to show that the administration of L-DOPA is followed by transient and robust increases in rCBF in the dopamine-denervated basal ganglia. This effect occurs already upon acute L-DOPA treatment and persists upon repeated drug administration in animals that develop dyskinesia. Increases in rCBF ON L-DOPA are not necessarily accompanied by enhanced glucose utilisation in the affected regions, pointing to altered mechanisms of neurovascular coupling. Finally, our results show that increases in rCBF ON L-DOPA may be accompanied by BBB hyperpermeability in the most affected regions.


Subject(s)
Antiparkinson Agents/pharmacology , Basal Ganglia/metabolism , Cerebrovascular Circulation/drug effects , Levodopa/pharmacology , Parkinson Disease, Secondary/drug therapy , Parkinson Disease, Secondary/physiopathology , Algorithms , Animals , Autoradiography , Basal Ganglia/drug effects , Behavior, Animal/drug effects , Benserazide/pharmacology , Blood-Brain Barrier/drug effects , Capillaries/metabolism , Dyskinesias/physiopathology , Female , Glucose/metabolism , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Sympatholytics , Tyrosine 3-Monooxygenase/metabolism
3.
Brain ; 134(Pt 8): 2339-57, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21771855

ABSTRACT

Angiogenesis and increased permeability of the blood-brain barrier have been reported to occur in animal models of Parkinson's disease and l-dopa-induced dyskinesia, but the significance of these phenomena has remained unclear. Using a validated rat model of l-dopa-induced dyskinesia, this study demonstrates that chronic treatment with l-dopa dose dependently induces the expression of vascular endothelial growth factor in the basal ganglia nuclei. Vascular endothelial growth factor was abundantly expressed in astrocytes and astrocytic processes in the proximity of blood vessels. When co-administered with l-dopa, a small molecule inhibitor of vascular endothelial growth factor signalling significantly attenuated the development of dyskinesia and completely blocked the angiogenic response and associated increase in blood-brain barrier permeability induced by the treatment. The occurrence of angiogenesis and vascular endothelial growth factor upregulation was verified in post-mortem basal ganglia tissue from patients with Parkinson's disease with a history of dyskinesia, who exhibited increased microvascular density, microvascular nestin expression and an upregulation of vascular endothelial growth factor messenger ribonucleic acid. These congruent findings in the rat model and human patients indicate that vascular endothelial growth factor is implicated in the pathophysiology of l-dopa-induced dyskinesia and emphasize an involvement of the microvascular compartment in the adverse effects of l-dopa pharmacotherapy in Parkinson's disease.


Subject(s)
Antiparkinson Agents/adverse effects , Brain/drug effects , Dyskinesia, Drug-Induced/etiology , Levodopa/adverse effects , Parkinsonian Disorders/pathology , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism , Aged , Aged, 80 and over , Analysis of Variance , Animals , Antigens, CD/metabolism , Antigens, Surface/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Behavior, Animal/drug effects , Benserazide/adverse effects , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Brain/cytology , Bromodeoxyuridine/metabolism , Cell Count , Cells, Cultured , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/pathology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Enzyme-Linked Immunosorbent Assay , Female , Humans , Laminin/metabolism , Male , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/drug therapy , Nerve Tissue Proteins/metabolism , Parkinsonian Disorders/drug therapy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Vascular Endothelial Growth Factor A/genetics
4.
Neuropsychopharmacology ; 34(12): 2477-88, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19606087

ABSTRACT

Angiogenesis occurs in the brains of Parkinson's disease patients, but the effects of dopamine replacement therapy on this process have not been examined. Using rats with 6-hydroxydopamine lesions, we have compared angiogenic responses induced in the basal ganglia by chronic treatment with either L-DOPA, or bromocriptine, or a selective D1 receptor agonist (SKF38393). Moreover, we have asked whether L-DOPA-induced angiogenesis can be blocked by co-treatment with either a D1- or a D2 receptor antagonist (SCH23390 and eticlopride, respectively), or by an inhibitor of extracellular signal-regulated kinases 1 and 2 (ERK1/2) (SL327). L-DOPA, but not bromocriptine, induced dyskinesia, which was associated with endothelial proliferation, upregulation of immature endothelial markers (nestin) and downregulation of endothelial barrier antigen in the striatum and its output structures. At a dose inducing dyskinesia (1.5 mg/kg/day), SKF38393 elicited angiogenic changes similar to L-DOPA. Antagonism of D1- but not D2 class receptors completely suppressed both the development of dyskinesia and the upregulation of angiogenesis markers. In fact, L-DOPA-induced endothelial proliferation was markedly exacerbated by low-dose D2 antagonism (0.01 mg/kg eticlopride). Inhibition of ERK1/2 by SL327 attenuated L-DOPA-induced dyskinesia and completely inhibited all markers of angiogenesis. These results highlight the specific link between treatment-induced dyskinesias and microvascular remodeling in the dopamine-denervated brain. L-DOPA-induced angiogenesis requires stimulation of D1 receptors and activation of ERK1/2, whereas the stimulation of D2 receptors seems to oppose this response.


Subject(s)
Antiparkinson Agents/pharmacology , Levodopa/pharmacology , Neovascularization, Pathologic/chemically induced , Parkinsonian Disorders/physiopathology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Animals , Basal Ganglia/blood supply , Basal Ganglia/drug effects , Basal Ganglia/physiology , Benzazepines/pharmacology , Bromocriptine/pharmacology , Disease Models, Animal , Dopamine Agents/pharmacology , Dopamine D2 Receptor Antagonists , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology , Oxidopamine , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D2/agonists , Salicylamides/pharmacology
5.
Parkinsonism Relat Disord ; 15 Suppl 4: S13-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20123549

ABSTRACT

Treatment-induced motor complications represent a major clinical problem in Parkinson's disease (PD). Pharmacological dopamine (DA) replacement with l-dopa causes motor fluctuations and abnormal involuntary movements (dyskinesia) in the vast majority of the patients. Intrastriatal grafts of embryonic dopaminergic neurons can cause dyskinesia too, as shown by clinical trials of neural transplantation in PD. Animals models of these complications can be produced in rats and mice in which the nigrostriatal DA pathway has been severely damaged. Rodent models allow investigators to explore mechanistic hypotheses at the cellular and molecular level. Moreover, the rat model of L-dopa-induced abnormal involuntary movements shows both face validity and predictive validity relative to the corresponding disorder in primates, and provides a cost effective tool to evaluate novel antidyskinetic interventions. This article reviews the strategies that have been used to reproduce different motor complications of PD treatment in rodents, and comments on their range of applicability.


Subject(s)
Brain Tissue Transplantation/adverse effects , Disease Models, Animal , Dyskinesia, Drug-Induced/etiology , Levodopa/adverse effects , Motor Skills Disorders/etiology , Parkinson Disease/drug therapy , Animals , Brain Tissue Transplantation/physiology , Dyskinesia, Drug-Induced/physiopathology , Humans , Mice , Motor Skills Disorders/chemically induced , Motor Skills Disorders/physiopathology , Parkinson Disease/physiopathology , Rats
6.
Parkinsonism Relat Disord ; 15 Suppl 3: S59-63, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20083009

ABSTRACT

The development of L-DOPA-induced dyskinesia (LID) is attributed to plastic responses triggered by dopamine (DA) receptor stimulation in the parkinsonian brain. This article reviews studies that have uncovered different levels of maladaptive plasticity in animal models of LID. Rats developing dyskinesia on chronic L-DOPA treatment show abnormal patterns of signaling pathway activation and synaptic plasticity in striatal neurons. In addition, these animals show a gene expression profile indicative of structural cellular plasticity, including pronounced upregulation of genes involved in extracellular matrix remodeling, neurite extension, synaptic vesicle trafficking, and endothelial and cellular proliferation. Structural changes of neurons and microvessels within the basal ganglia are currently being unraveled by detailed morphological analyses. The structural and functional adaptations induced by L-DOPA in the brain can be viewed as an attempt to meet increased metabolic demands and to boost cellular defense mechanisms. These homeostatic responses, however, also predispose to the appearance of dyskinesia and other complications during the course of the treatment.


Subject(s)
Antiparkinson Agents/pharmacology , Basal Ganglia/drug effects , Dyskinesia, Drug-Induced/etiology , Levodopa/pharmacology , Neuronal Plasticity/drug effects , Animals , Antiparkinson Agents/adverse effects , Basal Ganglia/pathology , Brain-Derived Neurotrophic Factor/metabolism , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/pathology , Humans , Levodopa/adverse effects , Microvessels/drug effects , Microvessels/physiology , Parkinson Disease/drug therapy , Serotonin/metabolism
7.
Behav Brain Res ; 177(1): 150-9, 2007 Feb 12.
Article in English | MEDLINE | ID: mdl-17157933

ABSTRACT

L-DOPA-induced motor complications can be modelled in rats with 6-hydroxydopamine (6-OHDA) lesions by chronic injections of L-DOPA. We have compared the sensitisation and duration of rotational responses, and the occurrence of dose-failure episodes and abnormal involuntary movements (AIMs) in 6-OHDA-lesioned rats with regard to the dose and route of administration of L-DOPA. Rats were treated with either low (6mg/kg) or high (25mg/kg) doses of L-DOPA twice daily for 21 days whereas control animals received injections of either saline or bromocriptine (2.5mg/kg). A dose-dependent and gradual development of AIMs and contralateral turning was observed in rats treated chronically with l-DOPA. Rats treated with bromocriptine exhibited rotational sensitisation but no AIMs. A shortening of motor response duration was not seen in any of the drug-treated groups. In contrast, dose-failure episodes occurred frequently in both L-DOPA- and bromocriptine-treated animals. Changing the route of L-DOPA administration from intraperitoneal to subcutaneous completely abolished failures in motor response without affecting the development of dyskinesia. Based on the hypothesis that higher doses of L-DOPA may be toxic to dopaminoceptive structures, we compared the total number of neurons and the levels of activated microglia in the striatum. No signs of neurodegenerative changes could be seen in any of the treatment groups. In conclusion, both body AIMs and rotations were dose-dependently evoked by L-DOPA. Only AIMs, however, provided a specific measure of dyskinesia since rotations also were induced by bromocriptine, a drug with low dyskinesiogenic potential. Dose-failure episodes were not specific to L-DOPA treatment and could be attributed to erratic drug absorption from the peritoneal route.


Subject(s)
Brain Injuries/chemically induced , Dopamine Agents/administration & dosage , Levodopa/administration & dosage , Movement Disorders/drug therapy , Oxidopamine , Analysis of Variance , Animals , Brain Injuries/complications , Bromocriptine/administration & dosage , Cell Count , Dopamine Agents/blood , Dose-Response Relationship, Drug , Drug Administration Routes , Female , Immunohistochemistry/methods , Levodopa/blood , Lipopolysaccharides/administration & dosage , Movement Disorders/etiology , Rats , Rats, Sprague-Dawley , Rotarod Performance Test/methods , Time Factors , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...