Skip to main content
Erschienen in: Brain Structure and Function 2/2013

Open Access 01.03.2013 | Original Article

Expression of angiotensinogen and receptors for angiotensin and prorenin in the monkey and human substantia nigra: an intracellular renin–angiotensin system in the nigra

verfasst von: Pablo Garrido-Gil, Rita Valenzuela, Begoña Villar-Cheda, Jose L. Lanciego, Jose L. Labandeira-Garcia

Erschienen in: Brain Structure and Function | Ausgabe 2/2013

Abstract

We have previously obtained in rodents a considerable amount of data suggesting a major role for the brain renin–angiotensin system (RAS) in dopaminergic neuron degeneration and potentially in Parkinson’s disease. However, the presence of a local RAS has not been demonstrated in the monkey or the human substantia nigra compacta (SNc). The present study demonstrates the presence of major RAS components in dopaminergic neurons, astrocytes and microglia in both the monkey and the human SNc. Angiotensin type 1 and 2 and renin–prorenin receptors were located at the surface of dopaminergic neurons and glial cells, as expected for a tissular RAS. However, angiotensinogen and receptors for angiotensin and renin–prorenin were also observed at the cytoplasm and nuclear level, which suggests the presence of an intracrine or intracellular RAS in monkey and human SNc. Although astrocytes and microglia were labeled for angiotensin and prorenin receptors in the normal SNc, most glial cells appeared less immunoreactive than the dopaminergic neurons. However, our previous studies in rodent models of PD and studies in other animal models of brain diseases suggest that the RAS activity is significantly upregulated in glial cells in pathological conditions. The present results together with our previous findings in rodents suggest a major role for the nigral RAS in the normal functioning of the dopaminergic neurons, and in the progression of the dopaminergic degeneration.
Abkürzungen
3n
Third cranial nerve
ACE
Angiotensin converting enzyme
AngII
Angiotensin II
Ang (RD)
Angiotensinogen (R&D Systems)
Ang (SC)
Angiotensinogen (Santa Cruz Biotechnology)
AT1R
Angiotensin II type 1 receptor
AT2R
Angiotensin II type 2 receptor
BSA
Bovine serum albumin
GFAP
Glial fibrillary acidic protein
HLA-DR
Human leukocyte antigen DR
KPBS
Potassium phosphate buffer saline
MBRF
Midbrain reticular formation
PB
Phosphate buffer
PD
Parkinson’s disease
PRR
Prorenin/renin receptor
RAS
Renin–angiotensin system
RN
Red nucleus
RT
Room temperature
SNc
Substantia nigra compacta
SNr
Substantia nigra pars reticulata
TH
Tyrosine hydroxylase
VTA
Ventral tegmental area

Introduction

The renin–angiotensin system (RAS) has long been considered as a circulating humoral system that regulates blood pressure and water homeostasis. Angiotensin II (AngII) is the most important effector peptide, and is formed by the sequential action of two enzymes, renin and angiotensin converting enzyme (ACE), on the precursor glycoprotein angiotensinogen. The actions of AngII are mediated by two main cell receptors: AngII type 1 and 2 (AT1 and AT2; Allen et al. 1998; Unger et al. 1996). AT1 receptors mediate most of the classical peripheral actions of AngII. However, AngII (via AT2 receptor) exerts actions directly opposed to those mediated by AT1 receptors thus antagonizing many of the effects of the latter (Chabrashvili et al. 2003; Jones et al. 2008). It is now generally accepted that in addition to the “classical” humoral RAS there exist local RAS in many tissues, including brain tissue (Ganong 1994; Re 2004), and that locally formed AngII regulates processes such as cell growth/apoptosis and inflammation (Ruiz-Ortega et al. 2001; Suzuki et al. 2003). Local AngII (via AT1 receptors) activates NADPH-dependent oxidases (Garrido and Griendling 2009; Touyz 2004), which are a major source of superoxide and are upregulated in major aging-related diseases such as hypertension, diabetes and atherosclerosis (Benigni et al. 2009; Griendling et al. 2000; Münzel and Keany 2001). However, activation of AT2 receptors inhibits NADPH-oxidase activation. Emerging aspects of the RAS, such as the identification of a specific receptor for renin and its precursor prorenin (PRR) (Nguyen 2011; Nguyen et al. 2002) and the identification of a functional intracellular/intracrine RAS in several types of cells, in addition to the “classical” humoral RAS and the local or tissue RAS (Baker et al. 2004; Re 2003), are contributing to a better understanding of the RAS and RAS-related diseases (Kumar et al. 2007, 2009).
The brain has an independent local RAS, which was initially associated with the central control of blood pressure. Over the last two decades, several components of the classical RAS have been identified in the brain (Cuadra et al. 2010; Phillips and de Oliveira 2008; Saavedra 2005; Wright and Harding 2011). Circulating AngII does not cross the brain–blood barrier, and the astroglia is considered as the main site of angiotensinogen synthesis in the brain (Milsted et al. 1990; Stornetta et al. 1988), although several authors have suggested that angiotensinogen is probably produced at lower levels in neurons (Kumar et al. 1988; Thomas et al. 1992). The existence of brain renin (Ganten et al. 1971) was initially controversial matter, probably due to the low expression levels of renin in comparison with the brain levels of AngII. However, the recent location of the new prorenin/renin receptor (PRR) in brain tissue has helped to clarify this issue (Nguyen et al. 2002).
More recent studies have involved the brain RAS in several brain disorders, including stroke (Li et al. 2005; Lou et al. 2004) and Alzheimer’s disease (Kehoe and Wilcock 2007; Mogi and Horiuchi 2009; Savaskan 2005). Over the last few years, we have carried out numerous experiments with rodents and have obtained a considerable amount of data suggesting a major role for the brain RAS in dopaminergic neuron degeneration and potentially in Parkinson’s disease (PD). We have observed all major components of the RAS system in dopaminergic neurons and glial cells of rat and mouse substantia nigra compacta (SNc), (Joglar et al. 2009; Rodriguez-Pallares et al. 2008). We have observed that in rodents RAS hyperactivation exacerbates NADPH-oxidase activity, oxidative stress and the microglial inflammatory response, which all contribute to the progression of dopaminergic neuron loss (Joglar et al. 2009; Lopez-Real et al. 2005; Muñoz et al. 2006; Rey et al. 2007; Rodriguez-Pallares et al. 2008). However, the presence of the major components of the RAS has not been demonstrated in primate or human SNc, which would provide further support for the involvement of brain RAS in PD. In the present study, we have demonstrated the location of the major components of the RAS in dopaminergic neurons and glial cells in the SNc of monkeys and humans, as well as the existence of an intracellular RAS in the SNc.

Materials and methods

Human postmortem and nonhuman primate tissue preparation

The human postmortem samples used in the present study were obtained from the Neurological Brain Bank of Navarra (Hospital of Navarra, Pamplona, Spain). Brains were dissected at autopsy from donors who had given informed consent in accordance with the Brain Donation Program of the Government of Navarra (Government directive 23/2001). The samples were obtained from four adult men (33.75 ± 6.4 years old) with no history or histological findings supporting any neurological disease. Following autopsy, brain slices including ventral mesencephalon were obtained, immediately frozen at −80°C and stored until processing. Postmortem times varied from 2.5 to 6 h.
Brain blocks including the mesencephalon were fixed by immersion in phosphate-buffered 4% paraformaldehyde for 24 h and cryoprotected for 48 h in a solution containing 20% of glycerin and 2% DMSO in 0.125 M phosphate buffer (PB), pH 7.4. Frozen coronal sections (40 µm thick) were then cut with a sliding microtome. Sections at different rostrocaudal levels of the SNc were processed by immunohistochemistry or immunofluorescence.
Nonhuman primate tissue was obtained from three adult male cynomolgus monkeys (Macaca fascicularis) (4.5–5 years old; body weight ranging from 3.8 to 4.5 kg). Animal handling was conducted in accordance with European Council Directive 86/609/EEC, and with the Society for Neuroscience Policy on the Use of Animals in Neuroscience Research. The experimental design was approved by the Ethical Committee for Animal Testing of the University of Navarra (ref: 019/2008) as well as by the Department of Health of the Government of Navarra (ref: NA-UNAV-04-08). Animals were anesthetized with an overdose of chloral hydrate and perfused transcardially with a fixative solution containing 4% paraformaldehyde and 0.1% glutaraldehyde in 0.125 M PB, pH 7.4. This very low amount of glutaraldehyde did not affect immunostaining and the brain sections can be used for additional studies with tracers that require a minimum amount of glutaraldehyde, and minimizes the number of monkeys used for research. Once perfusion was completed, the skull was opened, and the brain was removed and stored for 48 h in a cryoprotectant solution containing 20% of glycerin and 2% DMSO in 0.125 M PB, pH 7.4. Coronal tissue sections (40 µm thick) were then cut with a sliding microtome and collected in cryoprotectant solution. Sections at different rostrocaudal levels of the SNc were processed by immunohistochemistry or immunofluorescence.

Histological processing: immunoperoxidase

A free-floating immunoperoxidase labeling method was used to evaluate the expression of angiotensinogen/angiotensin, AT1R, AT2R and PRR immunoreactivity in monkey and human SNc. Firstly, endogen peroxidase activity was inhibited in SNc sections with a solution of hydrogen peroxide (H2O2; 3%; Merck) diluted in potassium phosphate buffer saline (KPBS). Antigen retrieval was achieved in human SNc sections by incubation with 10 mM sodium citrate buffer (pH = 3.5) for 30 min at 37°C. Tissue sections were then pre-incubated in KPBS containing bovine serum albumin (BSA, 1%; Sigma), normal swine or horse serum (4%; Vector Laboratories) and Triton X-100 (0.05%; Sigma) for 60 min at room temperature (RT). Tissue sections were subsequently incubated with different polyclonal antibodies raised against major RAS components (Table 1): angiotensinogen (goat IgG; 1:200; AF3156, R&D Systems, RD; detects human angiotensinogen), angiotensin (Angiotensin N-10, goat IgG; 1:1,000; sc-7419, Santa Cruz Biotechnology, SC; detects human angiotensinogen, angiotensin I, II and III), AT1R (rabbit IgG; 1:100; sc-579, Santa Cruz Biotechnology), AT2R (rabbit IgG; 1:100; sc-9040, Santa Cruz Biotechnology) and PRR (rabbit IgG; 1:100, ab40790; Abcam) for 48 h at 4°C in a dilution of KPBS–BSA (1%) containing 2% normal swine or horse serum. Following primary antibody labeling, tissue sections were first incubated for 60 min at RT in the corresponding biotinylated secondary antibodies (biotinylated swine anti-rabbit IgG, 1:200, Dako; biotinylated horse anti-goat IgG, 1:200, Vector laboratories). Tissue sections were then incubated with the avidin–biotin-peroxidase complex (1:70; Vectastain Elite ABC kit; Vector Laboratories) for 60 min at RT. Staining for peroxidase was performed in KPBS with 0.05% 3-3′ diaminobenzidine tetrahydrochloride (Sigma) and 0.04% H2O2. Tissue sections were mounted on gelatin-coated slides and coverslipped using DPX (Panreac). Finally, sections were visualized with a Nikon Optiphot-2 microscope connected to a digital camera (Nikon DXM1200). No staining was observed in control sections in which primary antibodies were omitted from the incubation solution. We previously confirmed the specificity of the antibodies used in the present study by Western blot analysis with the corresponding peptide-preabsorbed antibody (Rodriguez-Perez et al. 2010; Valenzuela et al. 2010).
Table 1
Primary antibodies
Antibody specificity
Host species
Type (clone)
Isotype
Dilution
Manufacturer (cat. no.)
Angiotensin II type 1 receptor
Rabbit
Polyclonal
IgG
IP: 1:100
Santa cruz biotechnology (sc-579)
IF: 1:50
Angiotensin II type 2 receptor
Rabbit
Polyclonal
IgG
IP: 1:100
Santa cruz biotechnology (sc-9040)
IF: 1:50
Angiotensinogen
Goat
Polyclonal
IgG
IP: 1:200
R&D systems (AF3156)
IF: 1:100
Angiotensinogen, angiotensin I, II and III
Goat
Polyclonal
IgG
IP: 1:1,000
Santa cruz biotechnology (sc-7419)
IF: 1:500
Glial fibrillary acidic protein
Mouse
Monoclonal (GA5)
IgG1
IF: 1:500
Millipore (MAB360)
HLA-DR
Mouse
Monoclonal (169-1B5)
IgG2b
IF: 1:50
MP biomedicals (68549)
HLA-DR
Mouse
Monoclonal (LN-3)
IgG2b
IF: 1:50
Novocastra (NCL-LN3)
Renin/prorenin receptor
Rabbit
Polyclonal
IgG
IP: 1:100
Abcam (ab40790)
IF: 1:50
Tyrosine hydroxylase
Mouse
Monoclonal (TH-16)
IgG
IF: 1:5,000
Sigma (T2928)
IP immunoperoxidase labeling method, IF immunofluorescence labeling method

Histological processing: double immunofluorescence labeling

Double immunofluorescence labeling was performed to identify the cells that expressed angiotensinogen/angiotensin, AT1R, AT2R and PRR in the human and monkey SNc. Angiotensinogen/angiotensin, AT1R, AT2R and PRR antibodies were combined with antibodies against tyrosine hydroxylase (TH; as a marker of dopaminergic neurons), glial fibrillary acidic protein (GFAP; as a marker of astrocytes), and human HLA class II-DR (as a marker of both resting and reactive microglia; Miles and Chou 1988; Verina et al. 2011). Neuromelanin granules detected by bright-field microscopy co-localized with TH in human SNc sections, and were also used as markers of dopaminergic neurons. Free-floating tissue sections containing SNc were pre-incubated in KPBS-1% BSA with 4% normal donkey serum (Sigma) and 0.05% Triton X-100 for 60 min at RT. Antigen retrieval was required for human SNc sections. Tissue sections were then incubated for 66–72 h at 4°C in primary antibodies (Table 1) raised against angiotensinogen (RD; 1:100), angiotensin (SC; 1:500), AT1R (1:50), AT2R (1:50), PRR (1:50), TH (1:5,000; mouse monoclonal; T2928, Sigma), GFAP (1:500; mouse monoclonal; MAB360, Millipore), HLA-DR (1:50; mouse monoclonal; 68549, MP Biomedicals) or HLA-DR (1:50; mouse monoclonal; NCL-LN3, Novocastra) diluted in KPBS-1% BSA with 2% normal donkey or rabbit serum. The immunoreaction was visualized with the fluorescent secondary antibodies: Alexa Fluor 568-conjugated donkey anti-rabbit IgG (1:200; Molecular Probes) or Alexa Fluor 488-conjugated donkey anti-mouse IgG (1:200; Molecular Probes) or Cy3-conjugated rabbit anti-goat IgG (1:200; Millipore). Finally, tissue sections were incubated for 30 min at RT with the DNA-binding dye Hoechst 33342 (3 × 10−5 M in KPBS), mounted on gelatin-coated slides and coverslipped with Immumount (Thermo-Shandon).
Tissue sections were visualized with a confocal laser-scanning microscope (TCS-SP2; Leica Microsystems Heidelberg GmbH, Mannheim, Germany). Confocal images were obtained by a sequential scan method and three different laser lines to avoid simultaneous excitation and possible overlap. Emission from the blue diode at 405 nm was detected in a spectral range of 422–457 nm and color-coded in blue. Emission from the argon laser at 488 nm was detected in a spectral range of 500–535 nm and color-coded in green. Finally, a spectral range of 581–625 nm was used to visualize the emission from the DPS diode at 561 nm, which was color-coded in red. Co-localization analysis was subsequently performed with the captured images in order to detect double-labeled cells. Series of confocal images were obtained every 0.7 μm in the Z-axis level by use of a sequential scan method. Analysis of the photographs at central cell levels revealed whether the labeling was located as a peripheral ring (suggesting membrane labeling) or throughout the cytoplasm. The presence of immunofluorescent labeling in neurons containing neuromelanin was analyzed using images obtained with the TCS-SP2 laser confocal microscope. To study co-localization of neuromelanin and immunofluorescence, emission from the DPS diode at 561 nm was detected in a spectral range of 581–625 nm and color-coded in green, and the bright-field illumination was acquired using an independent detection canal and color-coded in the gray scale.

Results

Single immunoperoxidase stains of sections from monkey and human ventral mesencephalon revealed intense immunoreactivity for angiotensinogen/angiotensin, AT1 and AT2 and PRR receptors in a large number of cells in the SNc, as well as in the ventral tegmental area (VTA) and other areas of the monkey and human ventral mesencephalon (Figs. 1, 2). The specificity of the labeling was confirmed using control sections in which the primary antibodies were omitted. The control sections did not exhibit immunoreactive labeling, and only neuronal profiles with neuromelanin granules were observed in the human SNc, thus revealing the location of the dopaminergic neurons.
Double immunofluorescence and confocal microscopy were used to identify the SNc cells that expressed angiotensinogen/angiotensin, AT1, AT2 receptors and PPR. Dopaminergic neurons were identified by their TH-immunoreactivity in monkey sections (Figs. 3, 4) and TH-immunoreactivity or the presence neuromelanin granules in human sections (Fig. 5). Co-localization of TH-immunofluorescence and neuromelanin granules was confirmed in human SNc neurons by confocal microscopy (Fig. 5a–c). Ultrastructural analysis is required to confirm unequivocally that labeling is located in the cytoplasm or membrane. However, series of confocal images were obtained every 0.7 μm in the Z-axis level by use of a sequential scan method. Analysis of the photographs at central cell levels revealed whether the labeling was located as a peripheral ring (suggesting membrane labeling) or throughout the cytoplasm. In the monkey, dopaminergic neurons were intensely immunopositive for all major RAS components studied. Angiotensinogen/angiotensin was located in both the cytoplasm and neuronal nucleus (Figs. 3a–c, 4a–c). Labeling for AT1 receptors was intense at the periphery of the neurons (Figs 3d–f, 4d–f), suggesting the presence of receptors at the cell surface. However, AT1R labeling was also intense within the neurons, particularly at nuclear level, as revealed by the use of Hoechst stain as a nuclear marker (Fig. 4d–f). AT2 receptor labeling appeared also located at the cell surface and the neuronal nucleus, and appeared particularly intense in the cytoplasm (Figs. 3g–i, 4g–i). PPR labeling was particularly intense in the neuronal nucleus (Figs. 3j–l, 4j–l), although weaker labeling was distinguished at the cell surface and in the cytoplasm. Intense labeling for angiotensinogen/angiotensin, AT1R, AT2R and PRR was also observed in human dopaminergic neurons. Labeling for the different receptors was apparently located at the cell surface, and in the cytoplasm and nucleus as described above for monkeys (Fig. 5).
All major RAS components studied (angiotensinogen/angiotensin, AT1R, AT2R and PRR) were also located in glial cells in the SNc of monkeys and humans (Figs. 6, 7, 8). In monkey and human SNc, astrocytes were identified by GFAP immunofluorescence, and exhibited intense labeling for angiotensinogen/angiotensin located in the cytoplasm, without a clear labeling at the nuclear level (Fig. 6a–h). AT1 receptors were mainly located at the cell surface and in the cytoplasm, although weak labeling for AT1R was occasionally observed at the nuclear level (Fig. 7a–h). AT2 receptor labeling was also observed in monkey and human astrocytes, particularly concentrated at nuclear and cytoplasmatic levels (Fig. 7i–p). Most astrocytes identified with GFAP did not show clear labeling for PRR in monkey and human SNc. However, a few astrocytes with unequivocal labeling for PRR were also observed revealing PRR in nuclear (Fig. 7q–t) and cytoplasmic (Fig. 7u–x) location.
Microglial cells were identified in human and monkey SNc sections by their HLA-DR immunoreactivity. In microglial cells of normal substantia nigra labeling for angiotensinogen/angiotensin was weak and mainly located at the cell surface (Fig. 6i–t). Immunoreactivity for AT1 receptors was observed at the cell surface and in the cytoplasm, and was also co-localized with the nuclear marker Hoechst (Fig. 8a–h). Human and monkey microglial cells were also immunoreactive for AT2 receptors, which was more intense in the cytoplasm at the perinuclear level (Fig. 8i–p). Immunoreactivity for PRR was also observed in monkey and human microglia (Fig. 8q–x). In most microglial cells PRR labeling appeared to be concentrated at the cell surface, although perinuclear or weak nuclear labeling was observed in some cells.

Discussion

In a series of recent studies (Joglar et al. 2009; Rodriguez-Pallares et al. 2008; Valenzuela et al. 2010), we demonstrated the presence of angiotensin receptors in nigral dopaminergic neurons and glial cells in rodents, as well as in rat primary mesencephalic cultures (Joglar et al. 2009; Rodriguez-Pallares et al. 2004, 2008). The present study shows the location of major components of the RAS in dopaminergic neurons, astrocytes and microglial cells of the monkey and human substantia nigra. AT1R and AT2R and PRR were located at the cell surface, as may be expected for a local or tissular RAS. However, angiotensinogen and angiotensin and PRR receptors were also observed at the cytoplasmic or nuclear level, which suggests the presence of an intracrine or intracellular RAS in monkey and human SNc. Several transmembrane receptors are known to accumulate in nuclei, particularly in nuclear membranes. Cells such as cardiomyocytes possess AngII receptors that couple to nuclear signaling pathways and regulate transcription. The observed intracellular location supports the possibility of an intracellular function for AngII, in addition to the effects induced by activation of cell surface AT1 and AT2 receptors. Extracellular AngII may act intracellularly by binding to AT1 receptors, which are subsequently internalized, or AngII may be synthesized within the cell. AT1R-dependent internalization of AngII has been described in a number of different cell types (Chen et al. 2000; Eggena et al. 1996; Lu et al. 1998). However, the demonstration of an intracellular location for PRR receptors in DA neurons suggests that some AngII may be formed intraneuronally, as previously suggested for heart cells (Baker et al. 2004). Intracellular AngII has been suggested to induce transcription of angiotensinogen and renin in response to binding to nuclear AT1 receptors in some cell types (Eggena et al. 1996). Therefore, our observation of intracellular PRR receptors supports the existence of an intracellular/intracrine RAS in the brain, and in the SNc in particular, as previously been suggested for other cell types (Lavoie et al. 2004; Re 2003).
Several studies have reported the presence of RAS components in the human basal ganglia (Allen et al. 1992; Quinlan and Phillips 1981). Early autoradiographic studies reported AT1 receptors in dopaminergic neurons, both in cell bodies in the SNc and in their terminal fields in the striatum of different mammals, including humans (Allen et al. 1992, 1998; Lenkei et al. 1997; Simonnet et al. 1981), and suggested that the density of AT1 receptors may be higher in human striatum and SNc than in rats and other mammals (Allen et al., 1992, 1998). However, the existence of brain renin (and therefore a local brain RAS) was initially controversial, largely because of low levels of renin expression, which were below the detection threshold of some standard assays. However, Immunoreactivity for renin has also been reported in other studies in neurons and glial cells in several areas of the rodent (Dzau et al. 1982; Fuxe et al. 1980) and human (Slater et al. 1980) brains. The existence of a local brain RAS was also questioned because brain levels of AngII may be much higher than the levels of renin detected. This may be now explained by the recent location of PRR receptors in the brain. High levels of PRR mRNA expression were initially observed in brain tissue homogenates (Nguyen et al. 2002), and we have observed abundant PRR receptors in neurons and glial cells in the monkey and human brain. PRR activates a catalytic activity similar to that of renin by binding of prorenin (previously considered inactive precursor of renin), and the prorenin to renin ratios are 5–10 times higher (and even up to 20–200 times higher in pathological conditions; Luetscher et al. 1985).
In the present study, intense immunoreactivity against the RAS components under study was observed in the monkey and human dopaminergic neurons. RAS appears to be involved in several functions in the normal brain. However, the interaction between AngII and dopamine is particularly interesting. An important interaction between dopamine and AngII receptors in peripheral tissues has been demonstrated in several recent studies, particularly with regard to the regulation of renal sodium excretion and cardiovascular function (Gildea 2009; Khan et al. 2008; Zeng et al. 2006). Recent evidence suggests that dopamine and AngII systems directly counterregulate each other in renal cells (Gildea 2009) and that abnormal counterregulatory interactions between dopamine and AngII play a major role in degenerative changes and hypertension (Li et al. 2008). In a recent study (Villar-Cheda et al. 2010), we have shown similar functional interactions and counterregulatory mechanisms in the striatum and SNc of rodents, and the present data suggest that similar interactions and counterregulatory mechanisms take place in humans. However, the present study not only shows that the tissular RAS is well developed in the SNc, but also that there is an important intracellular RAS in the dopaminergic neurons. The role of the intraneuronal RAS and its interactions with the tissular RAS are unknown, and are currently under study in our laboratory.
Although SNc astrocytes and microglial cells were labeled for angiotensin or prorenin receptors, most glial cells appeared less intensely immunoreactive than the dopaminergic neurons. However, it is important to note that we studied the SNc of normal human and monkey brains. Nonetheless, our previous studies in the SNc of rodent models of PD and studies in other animal models of brain diseases (Lanz et al. 2010; Lou et al. 2004; Stegbauer et al. 2009) suggest that the RAS activity is significantly upregulated in glial cells in lesioned brains. In rats lesioned with the dopaminergic neurotoxin 6-OHDA, we have observed a significant increase in the expression of AT1 and AT2 receptors in the SNc, apparently due to glial upregulation since the dopaminergic neurons had been eliminated by the toxin (Villar-Cheda et al. 2010). This issue was also confirmed in cultures of microglial cells, as activated microglial cells showed more intense nuclear labeling for AT1R, AT2R and PRR than that observed in the normal SNc in the present study (Joglar et al. 2009; Rodriguez-Pallares et al. 2008; Valenzuela et al. 2010). Together our studies suggest that RAS hyperactivity enhances microglial derived oxidative stress and neuroinflammation, which play a major role in degeneration of dopaminergic neurons and possibly the progression of PD. We have shown that AngII, via AT1 receptors, exacerbates dopaminergic cell death and that a RAS-induced increase in the glial response plays a major role (Joglar et al. 2009; Lopez-Real et al. 2005; Muñoz et al. 2006; Rey et al. 2007; Rodriguez-Pallares et al. 2008). This is consistent with recent findings showing that neuroinflammation and oxidative stress play a pivotal role at least in the progression of PD, and also with numerous studies showing that the local RAS plays a key role in the initiation and perpetuation of inflammation and oxidative damage in different tissues (Ruiz-Ortega et al. 2001; Suzuki et al. 2003; Zalba et al. 2001). However, it is not yet known whether the pro-oxidative and pro-inflammatory effects of the tissular RAS is completed or enhanced by the intracellular RAS, or whether the intracellular RAS plays a different role, or whether the intracellular RAS has different functions in neurons and glial cells.
The mechanisms responsible for PD have not been totally clarified (González-Hernández et al. 2010; Smith and Villalba 2008). However, some other data also suggest the potential involvement of RAS in PD. A marked reduction in AT1 receptors has been observed in the striatum of PD patients in comparison with normal brains, and attributed to the loss of dopaminergic terminals in PD (Allen et al. 1992, 1998). However, the decrease in AT1 receptor expression was possibly more closely related to the L-dopa treatment that the PD patients had received (Villar-Cheda et al. 2010), in accordance with our experimental data from rodents. Furthermore, increased ACE activity in the cerebrospinal fluid of patients with PD has been reported (Konings et al. 1994), as well as an association between genetic polymorphism of the ACE gene and PD (Lin et al. 2002). Finally, a major role of the brain RAS in the dopaminergic system function and dopaminergic degeneration is also supported by several recent studies in which we have shown hyperactivation of the nigral RAS in several animal models with increased vulnerability of dopaminergic neurons to degeneration (i.e. increased risk for PD). Higher nigral RAS activity has been observed in aged male rats than in young male rats (Villar-Cheda et al. 2012), in menopausal rats (either surgical or natural menopause) than in female rats with estrogen (Rodriguez-Perez et al. 2012), and in young male rats in comparison with young female rats (Rodriguez-Perez et al. 2011, 2012). We have confirmed that in aged male rats and menopausal females, aging and lack of estrogen enhances the dopaminergic cell death induced by dopaminergic neurotoxins, and that an increased nigral RAS activity is involved; we observed increased AT1 receptor expression, increased activation of the NADPH-oxidase complex and increased levels of the pro-inflammatory cytokines, which indicate a pro-oxidative, pro-inflammatory state in the substantia nigra. Increased RAS activity and dopaminergic vulnerability were reduced by treatment with the AT1R antagonists such as candesartan.
In conclusion, the present study reveals the presence of a local/tissue RAS and an intracellular RAS in the SNc of monkeys and humans, which suggests a major role for the nigral RAS in the normal function of the dopaminergic neurons. Our previous findings in mouse and rat models of PD suggest a major role for the nigral RAS in progression of the dopaminergic degeneration, and the present results confirm the presence of a well developed nigral RAS in monkeys and humans. A better comprehension of the role of the RAS in the SNc, and the subsequent RAS manipulation may lead to an effective neuroprotective strategy against PD as previously observed in cardiovascular and renal diseases.

Acknowledgments

The authors thank Pilar Aldrey, Iria Novoa and Jose A. Trillo for their excellent technical assistance. Funding: Spanish Ministry of Science and Innovation, Spanish Ministry of Health (RD06/0010/0013 and CIBERNED), Galician Government (XUGA) and European Regional Development Fund (FEDER).

Conflict of interest

We declare no conflicts of interest.

Open Access

This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 2.0 International License (https://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
Zurück zum Zitat Allen AM, MacGregor DP, Chai SY, Donnan GA, Kaczymarzyk S, Richardson K, Kalnins R, Ireton J, Mendelsohn FAO (1992) Angiotensin II receptor binding associated with nigrostriatal dopaminergic neurons in human basal ganglia. Ann Neurol 32:339–344PubMedCrossRef Allen AM, MacGregor DP, Chai SY, Donnan GA, Kaczymarzyk S, Richardson K, Kalnins R, Ireton J, Mendelsohn FAO (1992) Angiotensin II receptor binding associated with nigrostriatal dopaminergic neurons in human basal ganglia. Ann Neurol 32:339–344PubMedCrossRef
Zurück zum Zitat Allen AM, Moeller I, Jenkins A, Zhuo J, Aldred GP, Chai SY, Mendelsohn FAO (1998) Angiotensin receptors in the nervous system. Brain Res Bull 47:17–18PubMedCrossRef Allen AM, Moeller I, Jenkins A, Zhuo J, Aldred GP, Chai SY, Mendelsohn FAO (1998) Angiotensin receptors in the nervous system. Brain Res Bull 47:17–18PubMedCrossRef
Zurück zum Zitat Baker KM, Chernin MI, Schreiber T, Sanghi S, Haiderzaidi S, Booz GW, Dostal DE, Kumar R (2004) Evidence of a novel intracrine mechanism in angiotensin II-induced cardiac hypertrophy. Regul Pept 120:5–13PubMedCrossRef Baker KM, Chernin MI, Schreiber T, Sanghi S, Haiderzaidi S, Booz GW, Dostal DE, Kumar R (2004) Evidence of a novel intracrine mechanism in angiotensin II-induced cardiac hypertrophy. Regul Pept 120:5–13PubMedCrossRef
Zurück zum Zitat Benigni A, Corna D, Zoja C, Sonzogni A, Latini R, Salio M, Conti S, Rottoli D, Longaretti L, Cassis P, Morigi M, Coffman TM, Remuzzi G (2009) Disruption of the Ang II type 1 receptor promotes longevity in mice. J Clin Invest 119:524–530PubMedCrossRef Benigni A, Corna D, Zoja C, Sonzogni A, Latini R, Salio M, Conti S, Rottoli D, Longaretti L, Cassis P, Morigi M, Coffman TM, Remuzzi G (2009) Disruption of the Ang II type 1 receptor promotes longevity in mice. J Clin Invest 119:524–530PubMedCrossRef
Zurück zum Zitat Chabrashvili T, Kitiyakara C, Blau J, Karber A, Islam S, Welch WJ, Wilcox CF (2003) Effect of Ang II type 1 and 2 receptors on oxidative stress, renal NAD(P)H oxidase, and SOD expression. Am J Physiol Regu Integr Comp Physiol 285:R117–R124 Chabrashvili T, Kitiyakara C, Blau J, Karber A, Islam S, Welch WJ, Wilcox CF (2003) Effect of Ang II type 1 and 2 receptors on oxidative stress, renal NAD(P)H oxidase, and SOD expression. Am J Physiol Regu Integr Comp Physiol 285:R117–R124
Zurück zum Zitat Chen R, Mukhin YV, Garnovskaya MN, Thielen TE, Iijima Y, Huang C, Raymond JR, Ullian ME, Paul RV (2000) A functional angiotensin II receptor-GFP fusion protein: evidence for agonist-dependent nuclear translocation. Am J Physiol Renal Physiol 279:F440–F448PubMed Chen R, Mukhin YV, Garnovskaya MN, Thielen TE, Iijima Y, Huang C, Raymond JR, Ullian ME, Paul RV (2000) A functional angiotensin II receptor-GFP fusion protein: evidence for agonist-dependent nuclear translocation. Am J Physiol Renal Physiol 279:F440–F448PubMed
Zurück zum Zitat Cuadra AE, Shan Z, Sumners C, Raizada MK (2010) A current view of brain renin–angiotensin system: Is the (pro)renin receptor the missing link? Pharmacol Ther 125:27–38PubMedCrossRef Cuadra AE, Shan Z, Sumners C, Raizada MK (2010) A current view of brain renin–angiotensin system: Is the (pro)renin receptor the missing link? Pharmacol Ther 125:27–38PubMedCrossRef
Zurück zum Zitat Dzau VJ, Brenner A, Emmett NL (1982) Evidence for renin in rat brain: differentiation from other renin like enzymes. Am J Physiol 242:E292–E297PubMed Dzau VJ, Brenner A, Emmett NL (1982) Evidence for renin in rat brain: differentiation from other renin like enzymes. Am J Physiol 242:E292–E297PubMed
Zurück zum Zitat Eggena P, Zhu JH, Sereevinyayut S, Giordani M, Clegg K, Andersen PC, Hyun P, Barrett JD (1996) Hepatic angiotensin II nuclear receptors and transcription of growth-related factors. J Hypertens 14:961–968PubMedCrossRef Eggena P, Zhu JH, Sereevinyayut S, Giordani M, Clegg K, Andersen PC, Hyun P, Barrett JD (1996) Hepatic angiotensin II nuclear receptors and transcription of growth-related factors. J Hypertens 14:961–968PubMedCrossRef
Zurück zum Zitat Fuxe K, Ganten D, Hökfelt T, Locatelli V, Poulsen K, Stock G, Rix E, Taugner R (1980) Renin-like immunocytochemical activity in the rat and mouse brain. Neurosci Lett 18:245–250PubMedCrossRef Fuxe K, Ganten D, Hökfelt T, Locatelli V, Poulsen K, Stock G, Rix E, Taugner R (1980) Renin-like immunocytochemical activity in the rat and mouse brain. Neurosci Lett 18:245–250PubMedCrossRef
Zurück zum Zitat Ganong WF (1994) Origin of the angiotensin II secreted by cells. Proc Soc Exp Biol Med 205:213–219PubMed Ganong WF (1994) Origin of the angiotensin II secreted by cells. Proc Soc Exp Biol Med 205:213–219PubMed
Zurück zum Zitat Ganten D, Minnich JL, Granger P, Hayduk K, Brecht HM, Barbeau A, Boucher R, Genest J (1971) Angiotensin-forming enzyme in brain tissue. Science 173(991):64–65PubMedCrossRef Ganten D, Minnich JL, Granger P, Hayduk K, Brecht HM, Barbeau A, Boucher R, Genest J (1971) Angiotensin-forming enzyme in brain tissue. Science 173(991):64–65PubMedCrossRef
Zurück zum Zitat Garrido AM, Griendling KK (2009) NADPH oxidases and angiotensin II receptor signaling. Mol Cell Endocrinol 302(2):148–158PubMedCrossRef Garrido AM, Griendling KK (2009) NADPH oxidases and angiotensin II receptor signaling. Mol Cell Endocrinol 302(2):148–158PubMedCrossRef
Zurück zum Zitat Gildea JJ (2009) Dopamine and angiotensin as renal counterregulatory systems controlling sodium balance. Curr Opin Nephrol Hypertens 18:28–32PubMedCrossRef Gildea JJ (2009) Dopamine and angiotensin as renal counterregulatory systems controlling sodium balance. Curr Opin Nephrol Hypertens 18:28–32PubMedCrossRef
Zurück zum Zitat González-Hernández T, Cruz-Muros I, Afonso-Oramas D, Salas-Hernandez J, Castro-Hernandez J (2010) Vulnerability of mesostriatal dopaminergic neurons in Parkinson’s disease. Front Neuroanat 4:140PubMedCrossRef González-Hernández T, Cruz-Muros I, Afonso-Oramas D, Salas-Hernandez J, Castro-Hernandez J (2010) Vulnerability of mesostriatal dopaminergic neurons in Parkinson’s disease. Front Neuroanat 4:140PubMedCrossRef
Zurück zum Zitat Griendling KK, Sorescu D, Ushio-Fukai M (2000) NADPH oxidase. Role in cardiovascular biology and disease. Circ Res 86:494–501PubMedCrossRef Griendling KK, Sorescu D, Ushio-Fukai M (2000) NADPH oxidase. Role in cardiovascular biology and disease. Circ Res 86:494–501PubMedCrossRef
Zurück zum Zitat Joglar B, Rodriguez-Pallares J, Rodríguez-Perez AI, Rey P, Guerra MJ, Labandeira-Garcia JL (2009) The inflammatory response in the MPTP model of Parkinson’s disease is mediated by brain angiotensin: relevance to progression of the disease. J Neurochem 109:656–669PubMedCrossRef Joglar B, Rodriguez-Pallares J, Rodríguez-Perez AI, Rey P, Guerra MJ, Labandeira-Garcia JL (2009) The inflammatory response in the MPTP model of Parkinson’s disease is mediated by brain angiotensin: relevance to progression of the disease. J Neurochem 109:656–669PubMedCrossRef
Zurück zum Zitat Jones ES, Vinh A, McCarthy CA, Gaspari TA, Widdop RE (2008) AT2 receptors: functional relevance in cardiovascular disease. Pharmacol Ther 120(3):292–316PubMedCrossRef Jones ES, Vinh A, McCarthy CA, Gaspari TA, Widdop RE (2008) AT2 receptors: functional relevance in cardiovascular disease. Pharmacol Ther 120(3):292–316PubMedCrossRef
Zurück zum Zitat Kehoe PG, Wilcock GK (2007) Is inhibition of the renin–angiotensin system a new treatment option for Alzheimer’s disease? Lancet Neurol 6(4):373–378PubMedCrossRef Kehoe PG, Wilcock GK (2007) Is inhibition of the renin–angiotensin system a new treatment option for Alzheimer’s disease? Lancet Neurol 6(4):373–378PubMedCrossRef
Zurück zum Zitat Khan F, Spicarová Z, Zelenin S, Holtbäck U, Scott L, Aperia A (2008) Negative reciprocity between angiotensin II type 1 and dopamine D1 receptors in rat renal proximal tubule cells. Am J Physiol Renal Physiol 295:F1110–F1116PubMedCrossRef Khan F, Spicarová Z, Zelenin S, Holtbäck U, Scott L, Aperia A (2008) Negative reciprocity between angiotensin II type 1 and dopamine D1 receptors in rat renal proximal tubule cells. Am J Physiol Renal Physiol 295:F1110–F1116PubMedCrossRef
Zurück zum Zitat Konings CH, Kuiper MA, Bergmans PL, Grijpma AM, van Kamp GJ, Wolters EC (1994) Increased angiotensin-converting enzyme activity in cerebrospinal fluid of treated patients with Parkinson’s disease. Clin Chim Acta 23:101–106CrossRef Konings CH, Kuiper MA, Bergmans PL, Grijpma AM, van Kamp GJ, Wolters EC (1994) Increased angiotensin-converting enzyme activity in cerebrospinal fluid of treated patients with Parkinson’s disease. Clin Chim Acta 23:101–106CrossRef
Zurück zum Zitat Kumar A, Rassoli A, Raizada MK (1988) Angiotensinogen gene expression in neuronal and glial cells in primary cultures of rat brain. J Neurosci Res 19:287–290PubMedCrossRef Kumar A, Rassoli A, Raizada MK (1988) Angiotensinogen gene expression in neuronal and glial cells in primary cultures of rat brain. J Neurosci Res 19:287–290PubMedCrossRef
Zurück zum Zitat Kumar R, Singh VP, Baker KM (2007) The intracellular renin–angiotensin system: a new paradigm. Trends Endocrinol Metab 18(5):208–214PubMedCrossRef Kumar R, Singh VP, Baker KM (2007) The intracellular renin–angiotensin system: a new paradigm. Trends Endocrinol Metab 18(5):208–214PubMedCrossRef
Zurück zum Zitat Kumar R, Singh VP, Baker KM (2009) The intracellular renin–angiotensin system in the heart. Curr Hypertens Rep 11(2):104–110PubMedCrossRef Kumar R, Singh VP, Baker KM (2009) The intracellular renin–angiotensin system in the heart. Curr Hypertens Rep 11(2):104–110PubMedCrossRef
Zurück zum Zitat Lanz TV, Ding Z, Ho PP, Luo J, Agrawal AN, Srinagesh H, Axtell R, Zhang H, Platten M, Wyss-Coray T, Steinman L (2010) Angiotensin II sustains brain inflammation in mice via TGF-beta. J Clin Invest 120(8):2782–2794PubMedCrossRef Lanz TV, Ding Z, Ho PP, Luo J, Agrawal AN, Srinagesh H, Axtell R, Zhang H, Platten M, Wyss-Coray T, Steinman L (2010) Angiotensin II sustains brain inflammation in mice via TGF-beta. J Clin Invest 120(8):2782–2794PubMedCrossRef
Zurück zum Zitat Lavoie JL, Cassell MD, Gross KW, Sigmund CD (2004) Localization of renin expressing cells in the brain, by use of a REN-eGFP transgenic model. Physiol Genomics 16:240–246PubMed Lavoie JL, Cassell MD, Gross KW, Sigmund CD (2004) Localization of renin expressing cells in the brain, by use of a REN-eGFP transgenic model. Physiol Genomics 16:240–246PubMed
Zurück zum Zitat Lenkei Z, Palkovits M, Corvol P, Llorens-Cortes C (1997) Expression of angiotensin type-1 (AT1) and type 2 (AT2) receptor mRNAs in the adult rat brain: a functional neuroanatomical review. Front Neuroendocrinol 18:383–439PubMedCrossRef Lenkei Z, Palkovits M, Corvol P, Llorens-Cortes C (1997) Expression of angiotensin type-1 (AT1) and type 2 (AT2) receptor mRNAs in the adult rat brain: a functional neuroanatomical review. Front Neuroendocrinol 18:383–439PubMedCrossRef
Zurück zum Zitat Li J, Culman J, Hortnagl H, Zhao Y, Gerova N, Timm M, Blume A, Zimmermann M, Seidel K, Dirnagl U, Unger T (2005) Angiotensin AT2 receptor protects against cerebral ischemia-induced neuronal injury. FASEB J 19:617–619PubMed Li J, Culman J, Hortnagl H, Zhao Y, Gerova N, Timm M, Blume A, Zimmermann M, Seidel K, Dirnagl U, Unger T (2005) Angiotensin AT2 receptor protects against cerebral ischemia-induced neuronal injury. FASEB J 19:617–619PubMed
Zurück zum Zitat Li H, Armando I, Yu P, Escano C, Mueller SC, Asico L, Pascua A, Lu Q, Wang X, Villar VA, Jones JE, Wang Z, Periasamy A, Lau YS, Soares-da-Silva P, Creswell K, Guillemette G, Sibley DR, Eisner G, Gildea JJ, Felder RA, Jose PA (2008) Dopamine 5 receptor mediates Ang II type 1 receptor degradation via a ubiquitin-proteasome pathway in mice and human cells. J Clin Invest 118:2180–2189PubMedCrossRef Li H, Armando I, Yu P, Escano C, Mueller SC, Asico L, Pascua A, Lu Q, Wang X, Villar VA, Jones JE, Wang Z, Periasamy A, Lau YS, Soares-da-Silva P, Creswell K, Guillemette G, Sibley DR, Eisner G, Gildea JJ, Felder RA, Jose PA (2008) Dopamine 5 receptor mediates Ang II type 1 receptor degradation via a ubiquitin-proteasome pathway in mice and human cells. J Clin Invest 118:2180–2189PubMedCrossRef
Zurück zum Zitat Lin JJ, Yueh KC, Chang DC, Lin SZ (2002) Association between genetic polymorphism of angiotensin-converting enzyme gene and Parkinson disease. J Neurol Sci 199:25–29PubMedCrossRef Lin JJ, Yueh KC, Chang DC, Lin SZ (2002) Association between genetic polymorphism of angiotensin-converting enzyme gene and Parkinson disease. J Neurol Sci 199:25–29PubMedCrossRef
Zurück zum Zitat Lopez-Real A, Rey P, Soto-Otero R, Mendez-Alvarez E, Labandeira-Garcia JL (2005) Angiotensin-converting enzyme inhibitors reduce oxidative stress and protect dopaminergic neurons in a 6-hydroxydopamine rat model of parkinsonism. J Neurosci Res 81:865–873PubMedCrossRef Lopez-Real A, Rey P, Soto-Otero R, Mendez-Alvarez E, Labandeira-Garcia JL (2005) Angiotensin-converting enzyme inhibitors reduce oxidative stress and protect dopaminergic neurons in a 6-hydroxydopamine rat model of parkinsonism. J Neurosci Res 81:865–873PubMedCrossRef
Zurück zum Zitat Lou M, Blume A, Zhao Y, Gohlke P, Deuschl G, Herdegen T, Culman J (2004) Sustained blockade of brain AT1 receptors before and after focal cerebral ischemia alleviates neurologic deficits and reduces neuronal injury, apoptosis, and inflammatory responses in the rat. J Cereb Blood Flow Metab 24:536–547PubMedCrossRef Lou M, Blume A, Zhao Y, Gohlke P, Deuschl G, Herdegen T, Culman J (2004) Sustained blockade of brain AT1 receptors before and after focal cerebral ischemia alleviates neurologic deficits and reduces neuronal injury, apoptosis, and inflammatory responses in the rat. J Cereb Blood Flow Metab 24:536–547PubMedCrossRef
Zurück zum Zitat Lu D, Yang H, Shaw G, Raizada MK (1998) Angiotensin II-induced nuclear targeting of the angiotensin type 1 (AT1) receptor in brain neurons. Endocrinology 139:365–375PubMedCrossRef Lu D, Yang H, Shaw G, Raizada MK (1998) Angiotensin II-induced nuclear targeting of the angiotensin type 1 (AT1) receptor in brain neurons. Endocrinology 139:365–375PubMedCrossRef
Zurück zum Zitat Luetscher JA, Kraemer FB, Wilson DM, Schwartz HC, Bryer-Ash M (1985) Increased plasma inactive renin in diabetes mellitus. A marker of microvascular complications. N Engl J Med 312:1412–1417PubMedCrossRef Luetscher JA, Kraemer FB, Wilson DM, Schwartz HC, Bryer-Ash M (1985) Increased plasma inactive renin in diabetes mellitus. A marker of microvascular complications. N Engl J Med 312:1412–1417PubMedCrossRef
Zurück zum Zitat Miles JM, Chou SM (1988) A new immunoperoxidase marker for microglia in paraffin section. J Neuropathol Exp Neurol 47(6):579–587PubMedCrossRef Miles JM, Chou SM (1988) A new immunoperoxidase marker for microglia in paraffin section. J Neuropathol Exp Neurol 47(6):579–587PubMedCrossRef
Zurück zum Zitat Milsted A, Barna BP, Ransohoff RM, Brosnihan KB, Ferrario CM (1990) Astrocyte cultures derived from human brain tissue express angiotensinogen mRNA. Proc Natl Acad Sci USA 87:5720–5723PubMedCrossRef Milsted A, Barna BP, Ransohoff RM, Brosnihan KB, Ferrario CM (1990) Astrocyte cultures derived from human brain tissue express angiotensinogen mRNA. Proc Natl Acad Sci USA 87:5720–5723PubMedCrossRef
Zurück zum Zitat Mogi M, Horiuchi M (2009) Effects of angiotensin II receptor blockers on dementia. Hypertens Res 32(9):738–740PubMedCrossRef Mogi M, Horiuchi M (2009) Effects of angiotensin II receptor blockers on dementia. Hypertens Res 32(9):738–740PubMedCrossRef
Zurück zum Zitat Muñoz A, Rey P, Guerra MJ, Mendez-Alvarez E, Soto-Otero R, Labandeira-Garcia JL (2006) Reduction of dopaminergic degeneration and oxidative stress by inhibition of angiotensin converting enzyme in a MPTP model of parkinsonism. Neuropharmacology 51:112–120PubMedCrossRef Muñoz A, Rey P, Guerra MJ, Mendez-Alvarez E, Soto-Otero R, Labandeira-Garcia JL (2006) Reduction of dopaminergic degeneration and oxidative stress by inhibition of angiotensin converting enzyme in a MPTP model of parkinsonism. Neuropharmacology 51:112–120PubMedCrossRef
Zurück zum Zitat Münzel T, Keany JF (2001) Are ACE inhibitors a “magic bullet” against oxidative stress? Circulation 104:1571–1577PubMedCrossRef Münzel T, Keany JF (2001) Are ACE inhibitors a “magic bullet” against oxidative stress? Circulation 104:1571–1577PubMedCrossRef
Zurück zum Zitat Nguyen G (2011) Renin, (pro)renin and receptor: an update. Clin Sci (Lond) 120(5):169–178CrossRef Nguyen G (2011) Renin, (pro)renin and receptor: an update. Clin Sci (Lond) 120(5):169–178CrossRef
Zurück zum Zitat Nguyen G, Delarue F, Burcklé C, Bouzhir L, Giller T, Sraer JD (2002) Pivotal role of the renin/prorenin receptor in angiotensin II production and cellular responses to renin. J Clin Invest 109:1417–1427PubMed Nguyen G, Delarue F, Burcklé C, Bouzhir L, Giller T, Sraer JD (2002) Pivotal role of the renin/prorenin receptor in angiotensin II production and cellular responses to renin. J Clin Invest 109:1417–1427PubMed
Zurück zum Zitat Phillips MI, de Oliveira EM (2008) Brain renin angiotensin in disease. J Mol Med 86:715–722PubMedCrossRef Phillips MI, de Oliveira EM (2008) Brain renin angiotensin in disease. J Mol Med 86:715–722PubMedCrossRef
Zurück zum Zitat Quinlan JT, Phillips MI (1981) Immunoreactivity for angiotensin II-like peptide in the human brain. Brain Res 205:212–218PubMedCrossRef Quinlan JT, Phillips MI (1981) Immunoreactivity for angiotensin II-like peptide in the human brain. Brain Res 205:212–218PubMedCrossRef
Zurück zum Zitat Re RN (2003) Intracellular renin and the nature of intracrine enzymes. Hypertension 42:117–122PubMedCrossRef Re RN (2003) Intracellular renin and the nature of intracrine enzymes. Hypertension 42:117–122PubMedCrossRef
Zurück zum Zitat Rey P, Lopez-Real A, Sanchez-Iglesias S, Muñoz A, Soto-Otero R, Labandeira-Garcia JL (2007) Angiotensin type-1-receptor antagonists reduce 6-hydroxydopamine toxicity for dopaminergic neurons. Neurobiol Aging 28:555–567PubMedCrossRef Rey P, Lopez-Real A, Sanchez-Iglesias S, Muñoz A, Soto-Otero R, Labandeira-Garcia JL (2007) Angiotensin type-1-receptor antagonists reduce 6-hydroxydopamine toxicity for dopaminergic neurons. Neurobiol Aging 28:555–567PubMedCrossRef
Zurück zum Zitat Rodriguez-Pallares J, Quiroz CR, Parga JA, Guerra MJ, Labandeira-Garcia JL (2004) Angiotensin II increases differentiation of dopaminergic neurons from mesencephalic precursors via angiotensin type 2 receptors. Eur J Neurosci 20:1489–1498PubMedCrossRef Rodriguez-Pallares J, Quiroz CR, Parga JA, Guerra MJ, Labandeira-Garcia JL (2004) Angiotensin II increases differentiation of dopaminergic neurons from mesencephalic precursors via angiotensin type 2 receptors. Eur J Neurosci 20:1489–1498PubMedCrossRef
Zurück zum Zitat Rodriguez-Pallares J, Rey P, Parga JA, Muñoz A, Guerra MJ, Labandeira-Garcia JL (2008) Brain angiotensin enhances dopaminergic cell death via microglial activation and NADPH-derived ROS. Neurobiol Dis 31:58–73PubMedCrossRef Rodriguez-Pallares J, Rey P, Parga JA, Muñoz A, Guerra MJ, Labandeira-Garcia JL (2008) Brain angiotensin enhances dopaminergic cell death via microglial activation and NADPH-derived ROS. Neurobiol Dis 31:58–73PubMedCrossRef
Zurück zum Zitat Rodriguez-Perez AI, Valenzuela R, Villar-Cheda B, Guerra MJ, Lanciego JL, Labandeira-Garcia JL (2010) Estrogen and angiotensin interaction in the substantia nigra. Relevance to postmenopausal Parkinson’s disease. Exp Neurol 224:517–526PubMedCrossRef Rodriguez-Perez AI, Valenzuela R, Villar-Cheda B, Guerra MJ, Lanciego JL, Labandeira-Garcia JL (2010) Estrogen and angiotensin interaction in the substantia nigra. Relevance to postmenopausal Parkinson’s disease. Exp Neurol 224:517–526PubMedCrossRef
Zurück zum Zitat Rodriguez-Perez AI, Valenzuela R, Joglar B, Garrido-Gil P, Guerra MJ, Labandeira-Garcia JL (2011) Renin angiotensin system and gender differences in dopaminergic degeneration. Mol Neurodegener 6(1):58PubMedCrossRef Rodriguez-Perez AI, Valenzuela R, Joglar B, Garrido-Gil P, Guerra MJ, Labandeira-Garcia JL (2011) Renin angiotensin system and gender differences in dopaminergic degeneration. Mol Neurodegener 6(1):58PubMedCrossRef
Zurück zum Zitat Rodriguez-Perez AI, Valenzuela R, Villar-Cheda B, Guerra MJ, Labandeira-Garcia JL (2012) Dopaminergic neuroprotection of hormonal replacement therapy in young and aged menopausal rats. Role of the brain angiotensin system. Brain 135:124–138 Rodriguez-Perez AI, Valenzuela R, Villar-Cheda B, Guerra MJ, Labandeira-Garcia JL (2012) Dopaminergic neuroprotection of hormonal replacement therapy in young and aged menopausal rats. Role of the brain angiotensin system. Brain 135:124–138
Zurück zum Zitat Ruiz-Ortega M, Lorenzo O, Ruperez M, Suzuki Y, Egido J (2001) Proinflammatory actions of angiotensin II. Curr Opin Nephrol Hypertens 10:321–329PubMedCrossRef Ruiz-Ortega M, Lorenzo O, Ruperez M, Suzuki Y, Egido J (2001) Proinflammatory actions of angiotensin II. Curr Opin Nephrol Hypertens 10:321–329PubMedCrossRef
Zurück zum Zitat Saavedra JM (2005) Brain angiotensin II: new developments, unanswered questions and therapeutic opportunities. Cell Mol Neurobiol 25:485–512PubMedCrossRef Saavedra JM (2005) Brain angiotensin II: new developments, unanswered questions and therapeutic opportunities. Cell Mol Neurobiol 25:485–512PubMedCrossRef
Zurück zum Zitat Savaskan E (2005) The role of the brain renin–angiotensin system in neurodegenerative disorders. Curr Alzheimer Res 2(1):29–35PubMedCrossRef Savaskan E (2005) The role of the brain renin–angiotensin system in neurodegenerative disorders. Curr Alzheimer Res 2(1):29–35PubMedCrossRef
Zurück zum Zitat Simonnet G, Giorguieff-Chesselet MF, Carayon A, Bioulac B, Cesselin F, Glowinski J, Vincent JD (1981) Angiotensin II and the nigrostriatal system. J Physiol (Paris) 77:71–79 Simonnet G, Giorguieff-Chesselet MF, Carayon A, Bioulac B, Cesselin F, Glowinski J, Vincent JD (1981) Angiotensin II and the nigrostriatal system. J Physiol (Paris) 77:71–79
Zurück zum Zitat Slater EE, Defendini R, Zimmerman EA (1980) Wide distribution of immunoreactive renin in nerve cells of human brain. Proc Natl Acad Sci USA 77:5458–5460PubMedCrossRef Slater EE, Defendini R, Zimmerman EA (1980) Wide distribution of immunoreactive renin in nerve cells of human brain. Proc Natl Acad Sci USA 77:5458–5460PubMedCrossRef
Zurück zum Zitat Smith Y, Villalba R (2008) Striatal and extrastriatal dopamine in the basal ganglia: an overview of its anatomical organization in normal and Parkinsonian brains. Mov Disord 23(Suppl 3):S534–S547PubMedCrossRef Smith Y, Villalba R (2008) Striatal and extrastriatal dopamine in the basal ganglia: an overview of its anatomical organization in normal and Parkinsonian brains. Mov Disord 23(Suppl 3):S534–S547PubMedCrossRef
Zurück zum Zitat Stegbauer J, Lee DH, Seubert S, Ellrichmann G, Manzel A, Kvakan H, Muller DN, Gaupp S, Rump LC, Gold R, Linker RA (2009) Role of the renin–angiotensin system in autoimmune inflammation of the central nervous system. Proc Natl Acad Sci USA 106(35):14942–14947PubMedCrossRef Stegbauer J, Lee DH, Seubert S, Ellrichmann G, Manzel A, Kvakan H, Muller DN, Gaupp S, Rump LC, Gold R, Linker RA (2009) Role of the renin–angiotensin system in autoimmune inflammation of the central nervous system. Proc Natl Acad Sci USA 106(35):14942–14947PubMedCrossRef
Zurück zum Zitat Stornetta RL, Hawelu-Johnson CL, Guyenet PG, Lynch KR (1988) Astrocytes synthesize angiotensinogen in brain. Science 242:1444–1446PubMedCrossRef Stornetta RL, Hawelu-Johnson CL, Guyenet PG, Lynch KR (1988) Astrocytes synthesize angiotensinogen in brain. Science 242:1444–1446PubMedCrossRef
Zurück zum Zitat Suzuki Y, Ruiz-Ortega M, Lorenzo O, Ruperez M, Esteban V, Egido J (2003) Inflammation and angiotensin II. Int J Biochem Cell Biol 35:881–900PubMedCrossRef Suzuki Y, Ruiz-Ortega M, Lorenzo O, Ruperez M, Esteban V, Egido J (2003) Inflammation and angiotensin II. Int J Biochem Cell Biol 35:881–900PubMedCrossRef
Zurück zum Zitat Thomas WG, Greenland KJ, Shinkel TA, Sernia C (1992) Angiotensinogen is secreted by pure rat neuronal cell cultures. Brain Res 588:191–200PubMedCrossRef Thomas WG, Greenland KJ, Shinkel TA, Sernia C (1992) Angiotensinogen is secreted by pure rat neuronal cell cultures. Brain Res 588:191–200PubMedCrossRef
Zurück zum Zitat Touyz RM (2004) Reactive oxygen species and angiotensin II signaling in vascular cells-implications in cardiovascular disease. Braz J Med Biol Res 37:1263–1273PubMedCrossRef Touyz RM (2004) Reactive oxygen species and angiotensin II signaling in vascular cells-implications in cardiovascular disease. Braz J Med Biol Res 37:1263–1273PubMedCrossRef
Zurück zum Zitat Unger T, Chung O, Csikos T, Culman J, Gallinat S, Gohlke P, Höhle S, Meffert S, Stoll M, Stroth U, Zhu Y-Z (1996) Angiotensin receptors. J Hypertens Suppl 14(5):S95–S103PubMed Unger T, Chung O, Csikos T, Culman J, Gallinat S, Gohlke P, Höhle S, Meffert S, Stoll M, Stroth U, Zhu Y-Z (1996) Angiotensin receptors. J Hypertens Suppl 14(5):S95–S103PubMed
Zurück zum Zitat Valenzuela R, Barroso-Chinea P, Muñoz A, Joglar B, Villar-Cheda B, Lanciego JL, Labandeira-Garcia JL (2010) Location of prorenin receptors in primate substantia nigra. Effects on dopaminergic cell death. J Neuropathol Exp Neurol 69(11):1130–1142PubMedCrossRef Valenzuela R, Barroso-Chinea P, Muñoz A, Joglar B, Villar-Cheda B, Lanciego JL, Labandeira-Garcia JL (2010) Location of prorenin receptors in primate substantia nigra. Effects on dopaminergic cell death. J Neuropathol Exp Neurol 69(11):1130–1142PubMedCrossRef
Zurück zum Zitat Verina T, Kiihl SF, Schneider JS, Guilarte TR (2011) Manganese exposure induces microglia activation and dystrophy in the substantia nigra of non-human primates. Neurotoxicology 32(2):215–226PubMedCrossRef Verina T, Kiihl SF, Schneider JS, Guilarte TR (2011) Manganese exposure induces microglia activation and dystrophy in the substantia nigra of non-human primates. Neurotoxicology 32(2):215–226PubMedCrossRef
Zurück zum Zitat Villar-Cheda B, Rodríguez-Pallares J, Muñoz A, Valenzuela R, Guerra MJ, Baltatu OC, Labandeira-Garcia JL (2010) Nigral and striatal regulation of angiotensin receptor expression by dopamine and angiotensin in rodents: implications for progression of Parkinson’s disease. Eur J Neurosci 32:1695–1706PubMedCrossRef Villar-Cheda B, Rodríguez-Pallares J, Muñoz A, Valenzuela R, Guerra MJ, Baltatu OC, Labandeira-Garcia JL (2010) Nigral and striatal regulation of angiotensin receptor expression by dopamine and angiotensin in rodents: implications for progression of Parkinson’s disease. Eur J Neurosci 32:1695–1706PubMedCrossRef
Zurück zum Zitat Villar-Cheda B, Valenzuela R, Rodriguez-Perez AI, Guerra MJ, Labandeira-Garcia JL (2012) Aging-related changes in the nigral angiotensin system enhances proinflammatory and pro-oxidative markers and 6-OHDA-induced dopaminergic degeneration. Neurobiol Aging 33(1):204.e1–11 Villar-Cheda B, Valenzuela R, Rodriguez-Perez AI, Guerra MJ, Labandeira-Garcia JL (2012) Aging-related changes in the nigral angiotensin system enhances proinflammatory and pro-oxidative markers and 6-OHDA-induced dopaminergic degeneration. Neurobiol Aging 33(1):204.e1–11
Zurück zum Zitat Wright JW, Harding JW (2011) Brain renin–angiotensin—a new look at an old system. Prog Neurobiol 95:49–67PubMedCrossRef Wright JW, Harding JW (2011) Brain renin–angiotensin—a new look at an old system. Prog Neurobiol 95:49–67PubMedCrossRef
Zurück zum Zitat Zalba G, San Jose G, Moreno MU, Fortuño MA, Fortuño A, Beaumont FJ, Diez J et al (2001) Oxidative stress in arterial hypertension. Role of NADPH oxidase. Hypertension 38:1395–1399PubMedCrossRef Zalba G, San Jose G, Moreno MU, Fortuño MA, Fortuño A, Beaumont FJ, Diez J et al (2001) Oxidative stress in arterial hypertension. Role of NADPH oxidase. Hypertension 38:1395–1399PubMedCrossRef
Zurück zum Zitat Zeng C, Liu Y, Wang Z, He D, Huang L, Yu P, Zheng S, Jones JE, Asico LD, Hopfer U, Eisner GM, Felder RA, Jose PA (2006) Activation of D3 dopamine receptor decreases angiotensin II type 1 receptor expression in rat renal proximal tubule cells. Circ Res 99:494–500PubMedCrossRef Zeng C, Liu Y, Wang Z, He D, Huang L, Yu P, Zheng S, Jones JE, Asico LD, Hopfer U, Eisner GM, Felder RA, Jose PA (2006) Activation of D3 dopamine receptor decreases angiotensin II type 1 receptor expression in rat renal proximal tubule cells. Circ Res 99:494–500PubMedCrossRef
Metadaten
Titel
Expression of angiotensinogen and receptors for angiotensin and prorenin in the monkey and human substantia nigra: an intracellular renin–angiotensin system in the nigra
verfasst von
Pablo Garrido-Gil
Rita Valenzuela
Begoña Villar-Cheda
Jose L. Lanciego
Jose L. Labandeira-Garcia
Publikationsdatum
01.03.2013
Verlag
Springer-Verlag
Erschienen in
Brain Structure and Function / Ausgabe 2/2013
Print ISSN: 1863-2653
Elektronische ISSN: 1863-2661
DOI
https://doi.org/10.1007/s00429-012-0402-9

Weitere Artikel der Ausgabe 2/2013

Brain Structure and Function 2/2013 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.