Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(6)2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36982650

ABSTRACT

Antiparkinsonian carotid body (CB) cell therapy has been proven to be effective in rodent and nonhuman primate models of Parkinson's disease (PD), exerting trophic protection and restoration of the dopaminergic nigrostriatal pathway. These neurotrophic actions are mediated through the release of high levels of glial-cell-line-derived neurotrophic factor (GDNF) by the CB transplant. Pilot clinical trials have also shown that CB autotransplantation can improve motor symptoms in PD patients, although its effectiveness is affected by the scarcity of the grafted tissue. Here, we analyzed the antiparkinsonian efficacy of in vitro-expanded CB dopaminergic glomus cells. Intrastriatal xenografts of rat CB neurospheres were shown to protect nigral neurons from degeneration in a chronic MPTP mouse PD model. In addition, grafts performed at the end of the neurotoxic treatment resulted in the repair of striatal dopaminergic terminals through axonal sprouting. Interestingly, both neuroprotective and reparative effects induced by in vitro-expanded CB cells were similar to those previously reported by the use of CB transplants. This action could be explained because stem-cell-derived CB neurospheres produce similar amounts of GDNF compared to native CB tissue. This study provides the first evidence that in vitro-expanded CB cells could be a clinical option for cell therapy in PD.


Subject(s)
Carotid Body , Parkinson Disease , Mice , Rats , Humans , Animals , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Carotid Body/metabolism , Parkinson Disease/therapy , Parkinson Disease/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Cell Transplantation , Substantia Nigra/metabolism , Disease Models, Animal , Corpus Striatum/metabolism
2.
Cell Stem Cell ; 29(4): 528-544.e9, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35276096

ABSTRACT

The autonomic nervous system is a master regulator of homeostatic processes and stress responses. Sympathetic noradrenergic nerve fibers decrease bone mass, but the role of cholinergic signaling in bone has remained largely unknown. Here, we describe that early postnatally, a subset of sympathetic nerve fibers undergoes an interleukin-6 (IL-6)-induced cholinergic switch upon contacting the bone. A neurotrophic dependency mediated through GDNF-family receptor-α2 (GFRα2) and its ligand, neurturin (NRTN), is established between sympathetic cholinergic fibers and bone-embedded osteocytes, which require cholinergic innervation for their survival and connectivity. Bone-lining osteoprogenitors amplify and propagate cholinergic signals in the bone marrow (BM). Moderate exercise augments trabecular bone partly through an IL-6-dependent expansion of sympathetic cholinergic nerve fibers. Consequently, loss of cholinergic skeletal innervation reduces osteocyte survival and function, causing osteopenia and impaired skeletal adaptation to moderate exercise. These results uncover a cholinergic neuro-osteocyte interface that regulates skeletogenesis and skeletal turnover through bone-anabolic effects.


Subject(s)
Interleukin-6 , Osteogenesis , Cholinergic Agents , Cholinergic Fibers , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology
4.
Blood ; 133(3): 224-236, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30361261

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) and leukocytes circulate between the bone marrow (BM) and peripheral blood following circadian oscillations. Autonomic sympathetic noradrenergic signals have been shown to regulate HSPC and leukocyte trafficking, but the role of the cholinergic branch has remained unexplored. We have investigated the role of the cholinergic nervous system in the regulation of day/night traffic of HSPCs and leukocytes in mice. We show here that the autonomic cholinergic nervous system (including parasympathetic and sympathetic) dually regulates daily migration of HSPCs and leukocytes. At night, central parasympathetic cholinergic signals dampen sympathetic noradrenergic tone and decrease BM egress of HSPCs and leukocytes. However, during the daytime, derepressed sympathetic noradrenergic activity causes predominant BM egress of HSPCs and leukocytes via ß3-adrenergic receptor. This egress is locally supported by light-triggered sympathetic cholinergic activity, which inhibits BM vascular cell adhesion and homing. In summary, central (parasympathetic) and local (sympathetic) cholinergic signals regulate day/night oscillations of circulating HSPCs and leukocytes. This study shows how both branches of the autonomic nervous system cooperate to orchestrate daily traffic of HSPCs and leukocytes.


Subject(s)
Cell Movement , Cholinergic Agents/pharmacology , Circadian Rhythm , Hematopoietic Stem Cells/physiology , Leukocytes/physiology , Parasympathetic Nervous System/physiology , Sympathetic Nervous System/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Adhesion , Cells, Cultured , Chemotaxis , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Leukocytes/cytology , Leukocytes/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Adrenergic, beta-2 , Receptors, Adrenergic, beta-3/physiology , Receptors, G-Protein-Coupled/physiology
5.
Xenotransplantation ; 25(6): e12410, 2018 11.
Article in English | MEDLINE | ID: mdl-29932254

ABSTRACT

BACKGROUND: The use of long-term immunosuppressive treatments on neural transplantation has been controversial during the last decades. Although nowadays there is a consensus about the necessity of maintaining a permanent state of immunosuppression to preserve the survival of cerebral grafts, little is known about the effects that chronic immunosuppression produces both on the neurodegenerative process and on transplants function. METHODS: Here, we establish a new immunosuppressive protocol, based on the discontinuous administration of CsA (15 mg/kg; s.c.) and prednisone (20 mg/kg; s.c.), to produce long-term immunosuppression in mice. Using this treatment, we analyse the effects that long-term immunosuppression induces in a chronic 1-methyl-4-phenyl-1,2,3,6,-tetrahydropyridine (MPTP) model of parkinsonism and on the neuroprotective and neurorestorative anti-parkinsonian actions exerted by rat carotid body (CB) xenografts. RESULTS: This protocol preserves the survival of rat CB xenotransplants maintaining the general wellness of the grafted mice. Although permanent immunosuppression does not prevent the MPTP-induced cell death of nigral neurons and the consequent degeneration of dopaminergic striatal innervation, allowing for its use as Parkinson's disease (PD) model, it reduces the microglial activation and slightly declines the striatal damage. Moreover, we reported that chronic administration of immunosuppressant drugs does not alter the neuroprotective and restorative anti-parkinsonian actions of rat CB xenografts into parkinsonian mice. CONCLUSIONS: This new immunosuppressive protocol provides a new murine model to assay the long-term effects of cerebral xenografts and offer a pharmacological alternative to the commonly used genetic immunodeficient mice, allowing the use of genetically modified mice as hosts. In addition, it will permit the experimental analysis of the effects produced by human CB xenografts in the chronic PD murine model, with the final aim of using CB allografts as an option of cell therapy in PD patients.


Subject(s)
Carotid Body/pathology , Cell- and Tissue-Based Therapy , Heterografts/drug effects , Immunosuppression Therapy , Transplantation, Heterologous , Animals , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/metabolism , Immunosuppression Therapy/methods , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/pathology
6.
Glia ; 66(1): 191-205, 2018 01.
Article in English | MEDLINE | ID: mdl-29024008

ABSTRACT

Neuroinflammation mediated by chronically activated microglia, largely caused by abnormal accumulation of misfolded α-synuclein (αSyn) protein, is known to contribute to the pathophysiology of Parkinson's disease (PD). In this work, based on the immunomodulatory activities displayed by particular heat-shock proteins (HSPs), we tested a novel vaccination strategy that used a combination of αSyn and Grp94 (HSPC4 or Gp96) chaperone and a murine PD model. We used two different procedures, first, the adoptive transfer of splenocytes from αSyn/Grp94-immunized mice to recipient animals, and second, direct immunization with αSyn/Grp94, to study the effects in a chronic mouse MPTP-model of parkinsonism. We found that both approaches promoted a distinct profile in the peripheral system-supported by humoral and cellular immunity-consisting of a Th1-shifted αSyn-specific response accompanied by an immune-regulatory/Th2-skewed general phenotype. Remarkably, this mixed profile sustained by αSyn/Grp94 immunization led to strong suppression of microglial activation in the substantia nigra and striatum, pointing to a newly described positive effect of anti-αSyn Th1-responses in the context of PD. This strategy is the first to target αSyn and report the suppression of PD-associated microgliosis. Overall, we show that the αSyn/Grp94 combination supports a distinct and long-lasting immune profile in the peripheral system, which has an impact at the CNS level by suppressing chronic microglial activation in an MPTP model of PD. Furthermore, our study demonstrates that reshaping peripheral immunity by vaccination with appropriate misfolding protein/HSP combinations could be highly beneficial as a treatment for neurodegenerative misfolding diseases.


Subject(s)
Gliosis/etiology , Gliosis/therapy , Immunization/methods , MPTP Poisoning , Membrane Glycoproteins/immunology , alpha-Synuclein/immunology , Adoptive Transfer , Analysis of Variance , Animals , CD4 Antigens/metabolism , Chronic Disease , Cytokines/metabolism , Disease Models, Animal , MPTP Poisoning/chemically induced , MPTP Poisoning/complications , MPTP Poisoning/immunology , MPTP Poisoning/therapy , Male , Mice , Mice, Inbred C57BL , Microglia/pathology , Substantia Nigra/metabolism , Substantia Nigra/pathology , T-Lymphocytes, Regulatory/metabolism
7.
Stem Cells Transl Med ; 5(6): 804-15, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27075764

ABSTRACT

UNLABELLED: In models of Parkinson's disease (PD), Rho kinase (ROCK) inhibitors have antiapoptotic and axon-stabilizing effects on damaged neurons, decrease the neuroinflammatory response, and protect against dopaminergic neuron death and axonal retraction. ROCK inhibitors have also shown protective effects against apoptosis induced by handling and dissociation of several types of stem cells. However, the effect of ROCK inhibitors on dopaminergic cell grafts has not been investigated. In the present study, treatment of dopaminergic cell suspension with ROCK inhibitors yielded significant decreases in the number of surviving dopaminergic neurons, in the density of graft-derived dopaminergic fibers, and in graft vascularization. Dopaminergic neuron death also markedly increased in primary mesencephalic cultures when the cell suspension was treated with ROCK inhibitors before plating, which suggests that decreased angiogenesis is not the only factor leading to cell death in grafts. Interestingly, treatment of the host 6-hydroxydopamine-lesioned rats with ROCK inhibitors induced a slight, nonsignificant increase in the number of surviving neurons, as well as marked increases in the density of graft-derived dopaminergic fibers and the size of the striatal reinnervated area. The study findings discourage treatment of cell suspensions before grafting. However, treatment of the host induces a marked increase in graft-derived striatal reinnervation. Because ROCK inhibitors have also exerted neuroprotective effects in several models of PD, treatment of the host with ROCK inhibitors, currently used against vascular diseases in clinical practice, before and after grafting may be a useful adjuvant to cell therapy in PD. SIGNIFICANCE: Cell-replacement therapy is one promising therapy for Parkinson's disease (PD). However, many questions must be addressed before widespread application. Rho kinase (ROCK) inhibitors have been used in a variety of applications associated with stem cell research and may be an excellent strategy for improving survival of grafted neurons and graft-derived dopaminergic innervation. The present results discourage the treatment of suspensions of dopaminergic precursors with ROCK inhibitors in the pregrafting period. However, treatment of the host (patients with PD) with ROCK inhibitors, currently used against vascular diseases, may be a useful adjuvant to cell therapy in PD.


Subject(s)
Cell- and Tissue-Based Therapy , Corpus Striatum/pathology , Parkinson Disease/therapy , rho-Associated Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Corpus Striatum/transplantation , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/transplantation , Enzyme Inhibitors/administration & dosage , Humans , Mesencephalon/drug effects , Mesencephalon/pathology , Neuroprotective Agents/administration & dosage , Parkinson Disease/enzymology , Parkinson Disease/genetics , Rats , Transplants/drug effects , Transplants/growth & development , rho-Associated Kinases/metabolism
8.
J Neurochem ; 136(2): 373-87, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26500044

ABSTRACT

Despite the different animal models of Parkinson's disease developed during the last years, they still present limitations modelling the slow and progressive process of neurodegeneration. Here, we undertook a histological, neurochemical and behavioural analysis of a new chronic parkinsonian mouse model generated by the subcutaneous administration of low doses of MPTP (20 mg/kg, 3 times per week) for 3 months, using both young adult and aged mice. The MPTP-induced nigrostriatal neurodegeneration was progressive and was accompanied by a decrease in striatal dopamine levels and motor impairment. We also demonstrated the characteristic neuroinflammatory changes (microglial activation and astrogliosis) associated with the neurodegenerative process. Aged animals showed both a faster time course of neurodegeneration and an altered neuroinflammatory response. The long-term systemic application of low MPTP doses did not induce any increase in mortality in either young adult or aged mice and better resembles the slow evolution of the neurodegenerative process. This treatment could be useful to model different stages of Parkinson's disease, providing a better understanding of the pathophysiology of the disease and facilitating the testing of both protective and restorative treatments. Here, we show a new chronic and progressive parkinsonian mouse model, in young and aged mice. This model produces a stable degeneration of the dopaminergic nigrostriatal pathway, continuous neuroinflammatory reaction and motor deficits. Aged animals showed a faster neurodegeneration and an altered neuroinflammatory response. This treatment could be useful to model different stages of PD and to test both protective and restorative therapeutic approaches.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Aging , MPTP Poisoning , Age Factors , Animals , Catecholamines/metabolism , Chronic Disease , Corpus Striatum/pathology , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Inflammation/etiology , MPTP Poisoning/chemically induced , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Muscle Strength/drug effects , Nerve Degeneration/etiology , Nerve Tissue Proteins/metabolism , Psychomotor Performance/drug effects , Tyrosine 3-Monooxygenase/metabolism
9.
FASEB J ; 30(2): 564-77, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26443817

ABSTRACT

We have investigated the potential role of molecular chaperones as modulators of the immune response by using α-synuclein (αSyn) as an aggregation-prone model protein. We first performed an in vitro immunoscreening with 21 preselected candidate chaperones and selected 2 from this set as displaying immunological activity with differential profiles, Grp94/Gp96 and FKBP4/52. We then immunized mice with both chaperone/α-synuclein combinations using monomeric or oligomeric α-synuclein (MαSyn or OαSyn, respectively), and we characterized the immune response generated in each case. We found that Grp94 promoted αSyn-specific T-helper (Th)1/Th17 and IgG1 antibody responses (up to a 3-fold increase) with MαSyn and OαSyn, respectively, coupled to a Th2-type general phenotype (generating 2.5-fold higher IgG1/IgG2 levels). In addition, we observed that FKBP4 favored a Th1-skewed phenotype with MαSyn but strongly supported a Th2-type phenotype with OαSyn (with a 3-fold higher IL-10/IFN-γ serum levels). Importantly, results from adoptive transfer of splenocytes from immunized animals in a Parkinson's disease mouse model indicates that these effects are robust, stable in time, and physiologically relevant. Taken together, Grp94 and FKBP4 are able to generate differential immune responses to α-synuclein-based immunizations, depending both on the nature of the chaperone and on the aggregation state of α-synuclein. Our work reveals that several chaperones are potential modulators of the immune response and suggests that different chaperones could be exploited to redirect the amyloid-elicited immunity both for basic studies of the immunological processes associated with neurodegeneration and for immunotherapy of pathologies associated with protein misfolding and aggregation.


Subject(s)
Membrane Glycoproteins/metabolism , Molecular Chaperones/physiology , Tacrolimus Binding Proteins/metabolism , alpha-Synuclein/metabolism , Adaptive Immunity , Animals , Gene Expression Regulation , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunity, Innate , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Protein Folding , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/immunology , alpha-Synuclein/genetics
10.
Immun Inflamm Dis ; 2(4): 226-38, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25866630

ABSTRACT

α-Synuclein (αSyn) is a 140-residue amyloid-forming protein whose aggregation is linked to Parkinson's disease (PD). It has also been found to play a critical role in the immune imbalance that accompanies disease progression, a characteristic that has prompted the search for an effective αSyn-based immunotherapy. In this study, we have simultaneously exploited two important features of certain heat-shock proteins (HSPs): their classical "chaperone" activities and their recently discovered and diverse "immunoactive" properties. In particular, we have explored the immune response elicited by immunization of C57BL/6 mice with an αSyn/Hsp70 protein combination in the absence of added adjuvant. Our results show differential effects for mice immunized with the αSyn/Hsp70 complex, including a restrained αSyn-specific (IgM and IgG) humoral response as well as minimized alterations in the Treg (CD4(+)CD25(+)Foxp3(+)) and Teff (CD4(+)Foxp3(-)) cell populations, as opposed to significant changes in mice immunized with αSyn and Hsp70 alone. Furthermore, in vitro-stimulated splenocytes from immunized mice showed the lowest relative response against αSyn challenge for the "αSyn/Hsp70" experimental group as measured by IFN-γ and IL-17 secretion, and higher IL-10 levels when stimulated with LPS. Finally, serum levels of Th1-cytokine IFN-γ and immunomodulatory IL-10 indicated a unique shift toward an immunomodulatory/immunoprotective phenotype in mice immunized with the αSyn/Hsp70 complex. Overall, we propose the use of functional "HSP-chaperoned amyloid/aggregating proteins" generated with appropriate HSP-substrate protein combinations, such as the αSyn/Hsp70 complex, as a novel strategy for immune-based intervention against synucleinopathies and other amyloid or "misfolding" neurodegenerative disorders.

11.
Neurobiol Aging ; 34(3): 902-15, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22743091

ABSTRACT

Intrastriatal transplantation of dopaminergic carotid body (CB) cells ameliorates parkinsonism in animal models and, with less efficacy, in Parkinson's disease patients. CB-based cell therapy was initially proposed because of its high dopamine content. However, later studies suggested that its beneficial effect might be due to a trophic action exerted on nigrostriatal neurons. Compatible with this concept are the high levels of neurotrophic factors encountered in CB cells. To test experimentally this idea, unilateral striatal transplants were performed with a sham graft in the contralateral striatum, as a robust internal control. Thereafter, the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6, -tetrahydropyridine was injected during 3 months. CB grafts protected from degeneration ipsilateral nigral dopaminergic neurons projecting to the transplant in a dose-dependent manner regarding size and glial cell line-derived neurotrophic factor expression. Grafts performed at different times after the onset of the neurotoxic treatment demonstrated with histological and behavioral methods protection and repair of the nigrostriatal pathway by CB transplants. This study provides a mechanistic explanation for the action of CB transplants on parkinsonian models. It should also help to improve cell therapy approaches to Parkinson's disease.


Subject(s)
Carotid Body , Dopaminergic Neurons/transplantation , Parkinsonian Disorders , RNA, Messenger/analysis , Animals , Carotid Body/cytology , Carotid Body/transplantation , Corpus Striatum/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/surgery , Real-Time Polymerase Chain Reaction
12.
Regen Med ; 7(3): 309-22, 2012 May.
Article in English | MEDLINE | ID: mdl-22594325

ABSTRACT

AIMS: A major limiting factor for cell therapy in Parkinson's disease is that the survival of grafted dopaminergic neurons is very poor, which may be improved by administration of GDNF, for which the carotid body is a good source. MATERIALS & METHODS: Rats with total unilateral dopaminergic denervation were grafted with a cell suspension of rat dopaminergic neuroblasts with or without cell aggregates from the rat carotid body. At 1, 2 and 3 months after grafting, the rats were tested in the cylinder and the rotometer and killed 4 months after grafting. RESULTS: We observed that the survival of dopaminergic neurons and graft-derived dopaminergic innervation were higher in rats that received mixed grafts. Both grafted groups showed complete recovery in the amphetamine-induced rotation test. However, rats with cografts performed significantly better in the cylinder test. CONCLUSION: Cografting of carotid body cells may constitute a useful strategy for cell therapy in Parkinson's disease.


Subject(s)
Carotid Body/cytology , Carotid Body/transplantation , Dopaminergic Neurons/physiology , Dopaminergic Neurons/transplantation , Nerve Fibers/metabolism , Amphetamine , Animals , Cell Aggregation , Cell Survival , Dopaminergic Neurons/cytology , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mesencephalon/cytology , Mesencephalon/transplantation , Motor Activity/physiology , Neostriatum/cytology , Neostriatum/transplantation , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Rotation , Substantia Nigra/cytology , Time Factors , Tyrosine 3-Monooxygenase/metabolism
13.
Pflugers Arch ; 459(5): 775-83, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20049482

ABSTRACT

In this study, we explored the presence of aquaporins (AQPs), a family of membrane water channel proteins, in carotid body (CB) type I chemoreceptor cells. The CB is a polymodal chemoreceptor whose major function is to detect changes in arterial O2 tension to elicit hyperventilation during hypoxia. The CB has also been proposed to function as a systemic osmoreceptor, thus we hypothesized that the presence of AQPs in type I cell membrane may confer higher sensitivity to osmolarity changes and hence accelerate the activation of chemoreceptor cells. We detected the expression of AQP1, AQP7, and AQP8 in the CB and confirmed the location of AQP1 in type I cells. We have also shown that inhibition of AQP1 expression clearly reduced type I cell swelling after a hyposmotic shock, demonstrating that AQP1 has a major contribution in transmembrane water movement in these chemoreceptor cells. Interestingly, CB AQP1 expression levels change during postnatal development, increasing during the first postnatal weeks as the organ matures. In conclusion, in this study, we report the novel observation that AQPs are expressed in the CB. We also show that AQP1 mediates water transport across the cell membrane of type I cells, supporting the contribution of this protein to the osmoreception function of the CB.


Subject(s)
Aquaporin 1/metabolism , Biological Transport/physiology , Carotid Body/physiology , Water/metabolism , Animals , Aquaporin 1/genetics , Gene Expression Regulation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
14.
J Biol Chem ; 282(41): 30207-15, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17673462

ABSTRACT

O(2) is essential for aerobic life, and the classic view is that it diffuses freely across the plasma membrane. However, measurements of O(2) permeability of lipid bilayers have indicated that it is much lower than previously thought, and therefore, the existence of membrane O(2) channels has been suggested. We hypothesized that, besides its role as a water channel, aquaporin-1 (AQP-1) could also work as an O(2) transporter, because this transmembrane protein appears to be CO(2)-permeable and is highly expressed in cells with rapid O(2) turnover (erythrocytes and microvessel endothelium). Here we show that in mammalian cells overexpressing AQP-1 and exposed to hypoxia, the loss of cytosolic O(2), as well as stabilization of the O(2)-dependent hypoxia-inducible transcription factor and expression of its target genes, is accelerated. In normoxic endothelial cells, knocking down AQP-1 produces induction of hypoxia-inducible genes. Moreover, lung AQP-1 is markedly up-regulated in animals exposed to hypoxia. These data suggest that AQP-1 has O(2) permeability and thus could facilitate O(2) diffusion across the cell membrane.


Subject(s)
Aquaporin 1/biosynthesis , Cytosol/metabolism , Gene Expression Regulation , Hypoxia-Inducible Factor 1/metabolism , Hypoxia , Up-Regulation , Animals , Aquaporin 1/chemistry , Carbon Dioxide/chemistry , Cell Membrane/metabolism , Lung/metabolism , Microscopy, Confocal , Models, Biological , Nitroimidazoles/pharmacology , Oxygen/metabolism , PC12 Cells , Permeability , Rats
15.
J Neurol Neurosurg Psychiatry ; 78(8): 825-31, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17220289

ABSTRACT

BACKGROUND: Carotid body (CB) glomus cells are highly dopaminergic and express the glial cell line derived neurotrophic factor. The intrastriatal grafting of CB cell aggregates exerts neurotrophic actions on nigrostriatal neurons in animal models of Parkinson disease (PD). OBJECTIVE: We conducted a phase I-II clinical study to assess the feasibility, long term safety, clinical and neurochemical effects of intrastriatal CB autotransplantation in patients with PD. METHODS: Thirteen patients with advanced PD underwent bilateral stereotactic implantation of CB cell aggregates into the striatum. They were assessed before surgery and up to 1-3 years after surgery according to CAPIT (Core Assessment Programme for Intracerebral Transplantation) and CAPSIT-PD (Core Assessment Programme for Surgical Interventional Therapies in Parkinson's Disease) protocols. The primary outcome measure was the change in video blinded Unified Parkinson's Disease Rating Scale III score in the off-medication state. Seven patients had 18F-dopa positron emission tomography scans before and 1 year after transplantation. RESULTS: Clinical amelioration in the primary outcome measure was observed in 10 of 12 blindly analysed patients, which was maximal at 6-12 months after transplantation (5-74%). Overall, mean improvement at 6 months was 23%. In the long term (3 years), 3 of 6 patients still maintained improvement (15-48%). None of the patients developed off-period dyskinesias. The main predictive factors for motor improvement were the histological integrity of the CB and a milder disease severity. We observed a non-significant 5% increase in mean putaminal 18F-dopa uptake but there was an inverse relationship between clinical amelioration and annual decline in putaminal 18F-dopa uptake (r = -0.829; p = 0.042). CONCLUSIONS: CB autotransplantation may induce clinical effects in patients with advanced PD which seem partly related to the biological properties of the implanted glomus cells.


Subject(s)
Carotid Body/cytology , Cell Transplantation , Parkinson Disease/therapy , Adult , Corpus Striatum , Dopamine/metabolism , Female , Humans , Male , Middle Aged , Parkinson Disease/diagnostic imaging , Positron-Emission Tomography , Transplantation, Autologous , Treatment Outcome
16.
J Neurosci ; 26(17): 4500-8, 2006 Apr 26.
Article in English | MEDLINE | ID: mdl-16641229

ABSTRACT

Oxidative damage to dopaminergic nigrostriatal (DNS) neurons plays a central role in the pathogenesis of Parkinson's disease (PD). Glucose-6-phosphate dehydrogenase (G6PD) is a key cytoprotective enzyme that provides NADPH, the major source of the reducing equivalents of a cell. Mutations of this enzyme are the most common enzymopathies worldwide. We have studied in vivo the role of G6PD overexpressed specifically in the DNS pathway and show that the increase of G6PD activity in the soma and axon terminals of DNS neurons, separately from other neurons or glial cells, protects them from parkinsonism. Analysis of DNS neurons by histological, neurochemical, and functional methods showed that even a moderate increase of G6PD activity rendered transgenic mice more resistant than control littermates to the toxic effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). The neuroprotective action of G6PD was also observed in aged animals despite that they had a greater susceptibility to MPTP. Therefore, overexpression of G6PD in dopaminergic neurons or pharmacological activation of the native enzyme should be considered as potential therapeutic strategies to PD.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Glucosephosphate Dehydrogenase/metabolism , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Substantia Nigra/metabolism , Animals , Animals, Genetically Modified , Cells, Cultured , Corpus Striatum/pathology , Glucosephosphate Dehydrogenase/genetics , Mice/genetics , Neuroprotective Agents/metabolism , Recombinant Proteins/metabolism , Substantia Nigra/pathology
17.
J Neurosci ; 25(28): 6631-40, 2005 Jul 13.
Article in English | MEDLINE | ID: mdl-16014724

ABSTRACT

We studied the participation of adrenal medulla (AM) chromaffin cells in hypercapnic chemotransduction. Using amperometric recordings, we measured catecholamine (CAT) secretion from cells in AM slices of neonatal and adult rats perfused with solutions bubbled with different concentrations of CO2. The secretory activity augmented from 1.74 +/- 0.19 pC/min at 5% CO2 to 6.36 +/- 0.77 pC/min at 10% CO2. This response to CO2 was dose dependent and appeared without changes in extracellular pH, although it was paralleled by a drop in intracellular pH. Responsiveness to hypercapnia was higher in neonatal than in adult slices. The secretory response to hypercapnia required extracellular Ca2+ influx. Both the CO2-induced internal pH drop and increase in CAT secretion were markedly diminished by methazolamide (2 microm), a membrane-permeant carbonic anhydrase (CA) inhibitor. We detected the presence of two CA isoforms (CAI and CAII) in neonatal AM slices by in situ hybridization and real-time PCR. The expression of these enzymes decreased in adult AM together with the disappearance of responsiveness to CO2. In patch-clamped chromaffin cells, hypercapnia elicited a depolarizing receptor potential, which led to action potential firing, extracellular Ca2+ influx, and CAT secretion. This receptor potential (inhibited by methazolamide) was primarily attributable to activation of a resting cationic conductance. In addition, voltage-gated K+ current amplitude was also decreased by high CO2. The CO2-sensing properties of chromaffin cells may be of physiologic relevance, particularly for the adaptation of neonates to extrauterine life, before complete maturation of peripheral and central chemoreceptors.


Subject(s)
Adrenal Medulla/physiology , Carbon Dioxide/analysis , Carbonic Anhydrases/genetics , Chromaffin Cells/physiology , Hypercapnia/physiopathology , Animals , Animals, Newborn , Calcium/metabolism , Calcium/pharmacology , Carbon Dioxide/administration & dosage , Carbon Dioxide/pharmacology , Carbonic Anhydrases/biosynthesis , Catecholamines/metabolism , Chromaffin Cells/drug effects , Chromaffin Cells/metabolism , Dose-Response Relationship, Drug , Enzyme Induction , Extracellular Fluid/metabolism , Hydrogen-Ion Concentration , Intracellular Fluid/metabolism , Ion Transport , Isoenzymes/biosynthesis , Isoenzymes/genetics , Methazolamide/pharmacology , Patch-Clamp Techniques , Polymerase Chain Reaction , Potassium/metabolism , Potassium/pharmacology , Rats , Rats, Wistar , Sodium/metabolism , Sodium/pharmacology , Tetrodotoxin/pharmacology
18.
J Neurosci ; 25(16): 4091-8, 2005 Apr 20.
Article in English | MEDLINE | ID: mdl-15843611

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) exerts a notable protective effect on dopaminergic neurons in rodent and primate models of Parkinson's disease (PD). The clinical applicability of this therapy is, however, hampered by the need of a durable and stable GDNF source allowing the safe and continuous delivery of the trophic factor into the brain parenchyma. Intrastriatal carotid body (CB) autografting is a neuroprotective therapy potentially useful in PD. It induces long-term recovery of parkinsonian animals through a trophic effect on nigrostriatal neurons and causes amelioration of symptoms in some PD patients. Moreover, the adult rodent CB has been shown to express GDNF. Here we show, using heterozygous GDNF/lacZ knock-out mice, that unexpectedly CB dopaminergic glomus, or type I, cells are the source of CB GDNF. Among the neural or paraneural cells tested, glomus cells are those that synthesize and release the highest amount of GDNF in the adult rodent (as measured by standard and in situ ELISA). Furthermore, GDNF expression by glomus cells is maintained after intrastriatal grafting and in CB of aged and parkinsonian 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated animals. Thus, glomus cells appear to be prototypical abundant sources of GDNF, ideally suited to be used as biological pumps for the endogenous delivery of trophic factors in PD and other neurodegenerative diseases.


Subject(s)
Carotid Body/cytology , Corpus Striatum/transplantation , Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , MPTP Poisoning/therapy , Neurons/transplantation , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Carotid Body/metabolism , Carotid Body/ultrastructure , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry/methods , MPTP Poisoning/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Neurons/metabolism , Neurons/ultrastructure , PC12 Cells , Rats , Rats, Wistar , Time Factors , Tyrosine 3-Monooxygenase/metabolism
19.
J Neurosci ; 23(1): 141-8, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12514210

ABSTRACT

We studied the mechanisms underlying long-term functional recovery of hemiparkinsonian rats grafted intrastriatally with carotid body (CB) cell aggregates. Amelioration of their motor syndrome is a result of the trophic actions of these grafts on the remaining ipsilateral substantia nigra neurons rather than of the release of dopamine from the CB grafts. The grafts maintain a stable morphological appearance and differentiated cell phenotype for the duration of the life of the host. Adult CB expresses high levels of glial cell line-derived neurotrophic factor (GDNF) and the multicomponent GDNF receptor complex. These properties may contribute to the trophic actions of the CB transplants on nigrostriatal neurons and to their extraordinary longevity. We show that CB glomus cells, although highly dopaminergic, are protected from dopamine-mediated oxidative damage because of the absence of the high-affinity dopamine transporter. Thus, intrastriatal CB grafts are uniquely suited for long-term delivery of trophic factors capable of promoting restoration of the nigrostriatal pathway.


Subject(s)
Carotid Body/transplantation , Corpus Striatum/physiology , Membrane Glycoproteins , Nerve Tissue Proteins , Parkinson Disease/therapy , Substantia Nigra/physiology , Animals , Behavior, Animal , Carotid Body/metabolism , Cell Survival , Corpus Striatum/cytology , Corpus Striatum/pathology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins , Female , Glial Cell Line-Derived Neurotrophic Factor , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Growth Factors/metabolism , Neural Pathways , Neurons/drug effects , Neurons/metabolism , Neurotoxins/pharmacology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rats , Rats, Wistar , Substantia Nigra/cytology , Substantia Nigra/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...