Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(10)2021 May 11.
Article in English | MEDLINE | ID: mdl-34065020

ABSTRACT

Although optical hyperthermia could be a promising anticancer therapy, the need for high concentrations of light-absorbing metal nanoparticles and high-intensity lasers, or large exposure times, could discourage its use due to the toxicity that they could imply. In this article, we explore a possible role of silica microparticles that have high biocompatibility and that scatter light, when used in combination with conventional nanoparticles, to reduce those high concentrations of particles and/or those intense laser beams, in order to improve the biocompatibility of the overall procedure. Our underlying hypothesis is that the scattering of light caused by the microparticles would increase the optical density of the irradiated volume due to the production of multiple reflections of the incident light: the nanoparticles present in the same volume would absorb more energy from the laser than without the presence of silica particles, resulting either in higher heat production or in the need for less laser power or absorbing particles for the same required temperature rise. Testing this new optical hyperthermia procedure, based on the use of a mixture of silica and metallic particles, we have measured cell mortality in vitro experiments with murine glioma (CT-2A) and mouse osteoblastic (MC3T3-E1) cell lines. We have used gold nanorods (GNRs) that absorb light with a wavelength of 808 nm, which are conventional in optical hyperthermia, and silica microparticles spheres (hereinafter referred to as SMSs) with a diameter size to scatter the light of this wavelength. The obtained results confirm our initial hypothesis, because a high mortality rate is achieved with reduced concentrations of GNR. We found a difference in mortality between CT2A cancer cells and cells considered non-cancer MC3T3, maintaining the same conditions, which gives indications that this technique possibly improves the efficiency in the cell survival. This might be related with differences in the proliferation rate. Since the experiments were carried out in the 2D dimensions of the Petri dishes, due to sedimentation of the silica particles at the bottom, whilst light scattering is a 3D phenomenon, a large amount of the energy provided by the laser escapes outside the medium. Therefore, better results might be expected when applying this methodology in tissues, which are 3D structures, where the multiple reflections of light we believe will produce higher optical density in comparison to the conventional case of no using scattering particles. Accordingly, further studies deserve to be carried out in this line of work in order to improve the optical hyperthermia technique.


Subject(s)
Glioblastoma/therapy , Hyperthermia, Induced , Metal Nanoparticles/administration & dosage , Osteoblasts/cytology , Silicon Dioxide/chemistry , Animals , Cell Survival , Cells, Cultured , Glioblastoma/pathology , Lasers , Metal Nanoparticles/chemistry , Mice
2.
Nanotechnology ; 30(35): 355101, 2019 Aug 30.
Article in English | MEDLINE | ID: mdl-31082814

ABSTRACT

Metallic nanorods are promising agents for a wide range of biomedical applications. We report an optical hyperthermia method capable of inducing slowdown tumor progression of an experimental in vivo CT-2A glioblastoma tumor. The tumor model used in this research is based on the transplantation of mouse astrocytoma CT-2A cells in the striatum of mice by intracranial stereotaxic surgery. Two weeks after cell implant, the resulting tumor is treated by irradiating intratumoral injected gold nanorods, biofunctionalized with CD133 antibody (B-GNRs), using a continuous wave laser. Nanoparticles convert the absorbed light into localized heat (reaching up to 44 °C) due to the effect of surface plasmon resonance. A significant slowdown in CT-2A tumor progression is evident, by histology and magnetic resonance imaging, at one (p = 0.03) and two weeks (p = 0.008) after irradiation treatment. A notable deceleration in tumor size (15%-75%) as compared to the control untreated groups, it is observed. Thus, laser irradiation of B-GNRs is found to be effective for the treatment of CT-2A tumor progression. Similarities between the pre-clinical CT-2A tumor model and the human astrocytoma disease, in terms of anatomy, metastatic behavior and histopathology, suggest that hyperthermic treatment by laser irradiation of B-GNRs administered into high-grade human astrocytoma might constitute a promising alternative treatment to limit the progression of this deadly disease.


Subject(s)
Astrocytoma/therapy , Brain Neoplasms/therapy , Gold/pharmacology , Hyperthermia, Induced/methods , Laser Therapy/methods , Nanotubes/chemistry , AC133 Antigen/antagonists & inhibitors , AC133 Antigen/immunology , Animals , Antibodies, Neutralizing/pharmacology , Astrocytoma/immunology , Astrocytoma/pathology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Gold/administration & dosage , Gold/chemistry , Humans , Injections, Intralesional , Lasers , Mice , Mice, Inbred C57BL , Nanotubes/ultrastructure , Neoplasm Transplantation , Stereotaxic Techniques , Surface Plasmon Resonance , Tumor Burden/radiation effects
3.
Neuropathol Appl Neurobiol ; 39(7): 722-35, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23941684

ABSTRACT

Brain ischaemia and reperfusion produce alterations in the microenvironment of the parenchyma, including ATP depletion, ionic homeostasis alterations, inflammation, release of multiple cytokines and abnormal release of neurotransmitters. As a consequence, the induction of proliferation and migration of neural stem cells is redirected towards the peri-infarct region. The success of new neurorestorative treatments for damaged brain implies the need to describe with greater accuracy the mechanisms in charge of regulating adult neurogenesis, under both physiological and pathological conditions. Recent evidence demonstrates that many neurotransmitters, glutamate in particular, control the subventricular zone (SVZ), thus being part of the complex signal network that exerts a remarkable influence on the production of new neurones. Neurotransmitters provide a link between brain activity and SVZ neurogenesis. Therefore, a deeper knowledge of the role of neurotransmitters systems, such as glutamate and its transporters, in adult neurogenesis, may prove a valuable tool to be utilized as a neurorestorative therapy in this pathology.


Subject(s)
Brain Ischemia/metabolism , Neurogenesis/physiology , Neurons/cytology , Neurotransmitter Agents/metabolism , Stroke/pathology , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Ischemia/pathology , Humans
4.
Neurologia ; 26(2): 105-10, 2011 Mar.
Article in English, Spanish | MEDLINE | ID: mdl-21163186

ABSTRACT

INTRODUCTION: The progress of effective therapies for stroke has become a challenging task for both researchers and clinicians. Some pitfalls in clinical trials might have their origins in the pre-clinical experimental ischaemic models for the evaluation of potential neuro-protective agents. METHODS: We aim to standardise the methods for the development of stroke animal models throughout Spain, to produce document with appropriate recommendations and best practice in order to improve experimental methods in the field of stroke research. RESULTS: Members of several experienced stroke research groups prepared a guide with recommendations in the application of focal cerebral ischaemic models. The main features of this guide are based on the selection of the most appropriate animal model, taking in account the objective of the study, the species, strain, age, sex of animals, as well as risk factors. The experimental design must include a sham control group and the sample size calculation. Animal randomisation and blind analysis, masked assessment of outcomes, monitoring of body temperature and cerebral blood flow, and the reporting of reasons for excluding animals from the study, as well as the mortality rate, are other main points to fulfil in the application of stroke models. CONCLUSIONS: Standardised methods are essential to increase the success of the pre-clinical findings in the stroke neuroprotection field to be able to translate to the clinical practice.


Subject(s)
Biomedical Research/standards , Disease Models, Animal , Stroke , Animals , Guidelines as Topic
5.
Neuroscience ; 171(3): 885-92, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20854881

ABSTRACT

Adrenomedullin (AM) and its binding protein, complement factor H (FH), are expressed throughout the brain. In this study we used a brain-specific conditional knockout for AM and a complete knockout for FH to investigate the effect of these molecules on the pathophysiology of stroke. Following 48 h of middle cerebral artery permanent occlusion, there was a statistically significant infarct size increase in animals lacking AM when compared to their wild type littermates. In contrast, lack of FH did not affect infarct volume. To investigate some of the mechanisms by which lack of AM may augment brain damage, markers of nitrosative stress, apoptosis, and autophagy were studied at the mRNA and protein levels. There was a significant increase of inducible nitric oxide synthase (iNOS), matrix metalloproteinase-9 (MMP9), fractin, and Beclin-1 in the peri-infarct area of AM-deficient mice when compared to their wild type counterparts and to contralateral and sham-operated controls. These data suggest that AM exerts a neuroprotective action in the brain and that this protection may be mediated by regulation of iNOS, matrix metalloproteases, and inflammatory mediators. In the future, substances that increase AM actions in the central nervous system may be used as potential neuroprotective agents in stroke.


Subject(s)
Adrenomedullin/deficiency , Adrenomedullin/genetics , Brain Infarction/metabolism , Brain Infarction/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Complement Factor H/physiology , Adrenomedullin/physiology , Animals , Brain Infarction/genetics , Brain Ischemia/genetics , Complement Factor H/deficiency , Complement Factor H/genetics , Disease Models, Animal , Disease Progression , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
6.
Curr Pharm Des ; 16(25): 2837-50, 2010.
Article in English | MEDLINE | ID: mdl-20698819

ABSTRACT

Alzheimer's disease (AD) constitutes a progressive neurodegenerative disorder and the main cause of dementia. Numerous studies have focused on the pathogenic mechanism of AD to cure or prevent this devastating disease. But, despite recent advances, our understanding on the pathophysiology of this genetically complex and heterogeneous disorder is rather limited and treatment of the disease consists of medications to control the symptoms. Acetylcholinesterase inhibitors and N-methyl-D-aspartate (NMDA) receptor antagonists are the only available treatments recommended to manage the cognitive deficits caused by the disease. Therefore, the production of new drugs that may be able to cure the underlying cause of this chronic disease, not just the symptoms, is a matter of clinical interest. There is data implicating nitric oxide (NO) in the progression of the disease. The three isoforms of the NO-synthesizing enzyme (NOS) operate as central mediators of amyloid beta-peptide (Aß) action, giving rise to elevated levels of NO that contributes to the maintenance, self-perpetuation and progression of the disease. Reducing Aß production and the cholinergic deficit is a goal in the treatment of AD. In addition, a possible way to delay the progression of the illness must include a rationale design of enzyme inhibitors, subtype selective, targeting NOS isoforms implicated in damage to brain cells in AD. We are now presenting an overview regarding approved drugs for AD treatment and substances that although are not in use for the treatment of AD, including NOS inhibitors, may represent useful tools to unravel the pathophysiologic enigma of AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Drug Delivery Systems/methods , Enzyme Inhibitors/therapeutic use , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/metabolism , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Enzyme Inhibitors/pharmacology , Humans
7.
Oncogene ; 29(15): 2165-71, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20101216

ABSTRACT

Cancer and Alzheimer's disease (AD) are commonly found among elderly patients. Chronic inflammation is the characteristic of both diseases. Amyloid-beta peptide is the main inducer of inflammation in AD. Moreover, chronic inflammation promotes cancer, suggesting that AD patients may be more prone to develop cancer than non-demented people. To test this hypothesis, we injected the carcinogen 20-methylcholanthrene in the brain of transgenic mice overexpressing the mutant forms of amyloid precursor protein (APP) and presenilin 1 (PS1), as a model of AD, and their wild-type (WT) littermates. Mutant mice developed tumors faster and with higher incidence than their WT counterparts. Expression of the inflammatory markers interleukin (IL)-1alpha, IL-1beta, IL-6, IP-10 and tumor necrosis factor-alpha (TNF-alpha) was measured in AD and WT mice of 3 and 12 months of age that had not been exposed to the carcinogen. These cytokines were elevated in older AD mice, indicating the existence of a highly inflammatory milieu in these animals. We also found elevated expression of a mutated form of p53 in older AD mice, suggesting an alternative mechanism for the predisposition of AD brains to develop brain tumors. Clinical studies reporting comorbidity of AD and brain cancer are needed to understand whether our observations hold true for humans.


Subject(s)
Alzheimer Disease/pathology , Brain/drug effects , Brain/pathology , Carcinogens/toxicity , Disease Models, Animal , Aging , Alzheimer Disease/complications , Alzheimer Disease/genetics , Animals , Brain/metabolism , Brain Neoplasms/complications , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cytokines/genetics , Disease Susceptibility , Gene Expression Regulation/drug effects , Mice , Mice, Transgenic , Mutation , Oncogenes/genetics , Time Factors , Tumor Suppressor Protein p53/genetics
8.
Brain Res Rev ; 61(2): 154-69, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19520112

ABSTRACT

Since their discovery by Cajal in 1889, the Interstitial Cells of Cajal (ICC) have generated much controversy in the scientific community. Indeed, the nervous, muscle or fibroblastic nature of the ICC has remained under debate for more than a century, as has their possible physiological function. Cajal and his colleagues considered them to be neurons, while contemporary histologists like Kölliker and Dogiel categorized these cells as fibroblasts. More recently, the role of ICC in the origin of slow-wave peristaltism has been elucidated, and several studies have shown that they participate in neurotransmission (intercalation theory). The fact that ICC assemble in the circular muscular layer and that they originate from cells which emerge from the ventral neural tube (VENT cells), a source of neurons, glia and ICC precursors other than the neural crest, suggests a neural origin for this particular subset of ICC. The discovery that ICC express the Kit protein, a type III tyrosine kinase receptor encoded by the proto-oncogene c-kit, has helped better understand their physiological role and implication in pathological conditions. Gleevec, a novel molecule designed to inhibit the mutant activated version of c-Kit receptors, is the drug of choice to treat the so-called gastrointestinal stromal tumours (GIST), the most common non-epithelial neoplasm of the gastrointestinal tract. Here we review Cajal's original contributions with the aid of unique images taken from Cajal's histological slides (preserved at the Cajal Museum, Cajal Institute, CSIC). In addition, we present a historical review of the concepts associated with this particular cell type, emphasizing current data that has advanced our understanding of the role these intriguing cells fulfil.


Subject(s)
Enteric Nervous System/cytology , Gastrointestinal Tract/cytology , Muscle, Smooth/cytology , Animals , Biological Clocks/physiology , Enteric Nervous System/physiology , Gastrointestinal Tract/physiology , Muscle, Smooth/physiology , Neurons/physiology
9.
Neuroscience ; 158(4): 1390-6, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19135137

ABSTRACT

Stroke is the second most common cause of death and major cause of disability worldwide. Actual treatment involves surgery and/or thrombolytic drugs, but there is an urgent need for new approaches. Periodic acceleration, a rocking headward to footward movement of the whole body, is a non-invasive method to induce pulsatile shear stress on the vascular endothelium eliciting an enhanced production and secretion of endothelium-derived products such as nitric oxide, prostacyclin, prostaglandin E2, tissue plasminogen activator (tPA), and adrenomedullin. All these products have been shown to protect the brain from ischemic injuries. A rat model of focal brain ischemia was treated with application of periodic acceleration for 3 h immediately after the onset of ischemia. Controls remained static for the same period of time. Brain damage was assessed by magnetic resonance imaging (MRI) and biochemical markers. A significant reduction in brain damage was observed, 7 days post-ischemia, in rocked rats when compared with the static controls, through MRI. Furthermore, rocked animals had significantly lower levels of Beclin 1 and fractin than their static counterparts, and some isoforms of nitric oxide synthase were regulated by periodic acceleration. Our results show that periodic acceleration may provide a novel, affordable, non-invasive therapeutic option for the treatment of stroke.


Subject(s)
Acceleration , Brain Injuries/etiology , Brain Injuries/therapy , Brain Ischemia/complications , Exercise Therapy/methods , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Ischemia/chemically induced , Brain Ischemia/etiology , Caspase 3/genetics , Caspase 3/metabolism , Disease Models, Animal , Endothelin-1 , Gene Expression Regulation/physiology , Magnetic Resonance Imaging/methods , Male , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Periodicity , Rats , Rats, Wistar , Time Factors
10.
Histol Histopathol ; 22(12): 1309-26, 2007 12.
Article in English | MEDLINE | ID: mdl-17701911

ABSTRACT

Animal models of glial-derived neoplasms are needed to study the biological mechanisms of glioma tumorigenesis and those that sustain the disease state. With the aim to develop and characterize a suitable in vivo experimental mouse model for infiltrating astrocytoma, with predictable and reproducible growth patterns that recapitulate human astrocytoma, this study was undertaken to analyze the long-term course of a syngeneic orthotopically implanted CT-2A mouse astrocytoma in C57BL/6J mice. Intracranial injection of CT-2A cells into caudate-putamen resulted in development of an aggressive tumor showing typical features of human glioblastoma multiforme, sharing close histological, immunohistochemical, proliferative, and metabolic profiles. To simulate metastatic disease to the brain, CT-2A cells were injected through the internal carotid artery. Tumors identical to those obtained by intracranial injection were obtained. Finally, CT-2A cells were re-isolated from experimental brain tumors and transcranially re-injected into the caudate-putamen of healthy mice. These cells generated new tumors that were indistinguishable from the initial ones, suggesting in vivo self-renewal of tumor cells. Small-animal models are essential for testing novel biological therapies directed against relevant molecular targets. In a preliminary study, experimental CT-2A tumors were chronically treated with the small molecule 77427, a gastrin-releasing peptide (GRP) blocker compound that inhibits angiogenesis. Treated animals developed significantly smaller tumors than controls, suggesting an antitumor action for 77427 in glioblastomas. We conclude that the orthotopic CT-2A tumor model, as described herein, is appropriate to explore the mechanisms of glioma development and for preclinical trials of promising drugs.


Subject(s)
Astrocytoma/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Disease Models, Animal , Angiogenesis Inhibitors/pharmacology , Animals , Astrocytoma/diagnosis , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasm Transplantation
11.
Neuroscience ; 139(3): 889-98, 2006.
Article in English | MEDLINE | ID: mdl-16533568

ABSTRACT

The nitrergic system produces nitric oxide as an atypical neurotransmitter in the nervous system. Nitric oxide is produced from l-arginine through specific enzymes known as nitric oxide synthases. Of these, the more abundant form in neurons is the constitutive neuronal nitric oxide synthase, although the inducible isoform can be expressed as well, especially following stress or other injuries. The excessive formation of nitric oxide results in protein nitration, particularly at tyrosine residues, thus the presence of nitrotyrosine can be used as a marker of nitric oxide production. In previous studies we have shown the distribution of the components of the nitrergic system in the cerebellum of rodents, where neuronal nitric oxide synthase immunoreactivity was present in stellate and basket cells, and occasionally in granule cells. Here, we present evidence that in the sheep, as a model of larger mammals, most cerebellar neurons display an intense immunostaining for neuronal nitric oxide synthase, including unipolar brush cells, and Lugaro and Golgi neurons, which are not immunoreactive in rodents. In addition, weak immunoreactivity for inducible nitric oxide synthase and nitrotyrosine was found in particular cell types, indicating a basal expression for these markers. Our results suggest a larger dependence on the nitrergic system for the cerebella of larger mammals. Since this increase happens in both activating and inhibitory neurons of the cerebellar circuitry, we propose that in these animals there is a higher steady-state regulation of the cerebellum based on nitric oxide.


Subject(s)
Cerebellum/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type I/metabolism , Tyrosine/analogs & derivatives , Animals , Immunohistochemistry , Male , Nitric Oxide/metabolism , Sheep , Tyrosine/metabolism
12.
Neuroscience ; 128(1): 73-89, 2004.
Article in English | MEDLINE | ID: mdl-15450355

ABSTRACT

Changes in the amyloid-peptide (Abeta), neuronal and inducible nitric oxide (NO)synthase (nNOS, iNOS), nitrotyrosine, glial fibrillary acidic protein, and lectin from Lycopersicon esculentum (tomato) were investigated in the cerebral cortex of transgenic mice (Tg2576) to amyloid precursor protein (APP), by immunohistochemistry (bright light, confocal, and electron microscopy). The expression of nitrergic proteins and synthesis of nitric oxide were analyzed by immunoblotting and NOS activity assays, respectively. The cerebral cortex of these transgenic mice showed an age-dependent progressive increase in intraneuronal aggregates of Abeta-peptide and extracellular formation of senile plaques surrounded by numerous microglial and reactive astrocytes. Basically, no changes to nNOS reactivity or expression were found in the cortical mantle of either wild or transgenic mice. This reactivity in wild mice corresponded to numerous large type I and small type II neurons. The transgenic mice showed swollen, twisted, and hypertrophic preterminal and terminal processes of type I neurons, and an increase of the type II neurons. The calcium-dependent NOS enzymatic activity was higher in wild than in the transgenic mice. The iNOS reactivity, expression and calcium-independent enzymatic activity increased in transgenic mice with respect to wild mice, and were related to cortical neurons and microglial cells. The progressive elevation of NO production resulted in a specific pattern of protein nitration in reactive astrocytes. The ultrastructural study carried out in the cortical mantle showed that the neurons contained intracellular aggregates of Abeta-peptide associated with the endoplasmic reticulum, mitochondria, and Golgi apparatus. The endothelial vascular cells also contained Abeta-peptide deposits. This transgenic model might contribute to understand the role of the nitrergic system in the biological changes related to neuropathological progression of Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Neurons/metabolism , Nitric Oxide/metabolism , Tyrosine/analogs & derivatives , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Astrocytes/metabolism , Astrocytes/pathology , Blotting, Western , Cerebral Cortex/ultrastructure , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Neurons/pathology , Neurons/ultrastructure , Nitric Oxide Synthase/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Tyrosine/metabolism
13.
Histol Histopathol ; 19(3): 823-44, 2004 07.
Article in English | MEDLINE | ID: mdl-15168346

ABSTRACT

Temporal cortical sections from postmortem brains of individuals without any dementing condition and with different degrees of severity of Alzheimer's disease (AD) evaluated by the Clinical Dementia Rating scale (CDR 0-CDR 3) were analyzed using immunohistochemical procedures. To demonstrate the amyloid-beta-peptide (Abeta) deposition and the neurofibrillary pathology, two monoclonal antibodies were used, a human CERAD Abeta (10D5) antibody raised against the N-terminal region of the Abeta-peptide, and an antibody raised against paired helical filaments (PHF-1). The neuron cell bodies and the glial cells were also recognized by two polyclonal antibodies raised, respectively, against the protein gene peptide (PGP 9.5) and glial fibrillary acidic protein (GFAP). Directly related to severity of AD, progressive deposits of Abeta-peptide were found within cortical pyramidal-like neurons and forming senile plaques. Ultrastructurally, Abeta-peptide deposits were related to neuronal intracytoplasmic organelles, such as the ER, the mitochondria, the Nissl bodies and lipofuscin. We have also found that the intracellular deposition of the Abeta peptide is a neuropathological finding prior to the appearance of PHF-immunoreactive structures. We suggest that the intracellular Abeta deposition in cortical pyramidal neurons is a first neurodegenerative event in AD development and that it is involved in cell dysfunction, neuronal death, and plaque formation.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal/metabolism , Severity of Illness Index , Aged , Aged, 80 and over , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/ultrastructure , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/ultrastructure , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/ultrastructure , Biomarkers , Glial Fibrillary Acidic Protein/immunology , Glial Fibrillary Acidic Protein/ultrastructure , Humans , Immunohistochemistry , Middle Aged , Neurons/pathology , Neurons/ultrastructure , Temporal Lobe/metabolism , Temporal Lobe/pathology , Temporal Lobe/ultrastructure , Ubiquitin Thiolesterase/immunology , Ubiquitin Thiolesterase/metabolism
14.
Neurobiol Dis ; 15(2): 287-305, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15006699

ABSTRACT

The expression of neuronal nitric oxide (nNOS) and inducible nitric oxide (iNOS) as isoforms of the nitric oxide synthase (NOS) as well as nitrotyrosine as an end product of protein nitration was analyzed in sections of temporal cortex taken from postmortem brains of patients with Alzheimer's disease (AD). The patients were evaluated by the Clinical Dementia Rating scale (CDR0-CDR3) and studied in the Memory and Aging Project (MAP) of the Washington University Alzheimer Disease Research Center (ADCR). With the use of immunocytochemical procedures, neurons immunoreactive to nNOS were found to show large and small multipolar and pyramidal morphologies over the entire chronic AD evolution. The iNOS and nitrotyrosine immunoreactivities were also found in pyramidal-like cortical neurons and glial cells. Here, we speculate on the interaction among all specific neurodegenerative changes in AD and nitric oxide as an additional contribution to neuronal death in AD.


Subject(s)
Alzheimer Disease/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Tyrosine/analogs & derivatives , Aged , Aged, 80 and over , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Cell Death/physiology , Humans , Immunohistochemistry , Middle Aged , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Neurons/pathology , Nitrates/metabolism , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Pyramidal Cells/enzymology , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Temporal Lobe/enzymology , Temporal Lobe/metabolism , Temporal Lobe/pathology , Tyrosine/metabolism
15.
Microsc Res Tech ; 57(2): 76-90, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11921358

ABSTRACT

Adrenomedullin (AM) is a novel vasodilator peptide first purified from human pheochromocytoma by tracing its capacity to stimulate cAMP production in platelets. AM immunoreactivity is widely distributed in the central nervous system (CNS) and in the rat has been demonstrated by immunohistochemical techniques to be present in many neurons throughout the brain and spinal cord, as well as in some vascular endothelial cells and perivascular glial cells. Electron microscopy shows that the immunoreactivity is located mainly in the neuronal cytoplasm, but also occurs in the cell nucleus in some cells of the caudate putamen and olfactory tubercle. Biochemical analyses suggest that higher molecular forms, presumably precursor forms, may predominate over fully processed AM in some brain areas. The expression of AM immunoreactivity is increased in cortical neurons, endothelial cells, and perivascular processes after a simulation of ischemia by oxygen and glucose deprivation. Immunohistochemical, electrophysiological, and pharmacological studies suggest that AM in the CNS can act as a neurotransmitter, neuromodulator, or neurohormone, or as a cytoprotective factor in ischemic/hypoxic conditions, in addition to its vasodilator role.


Subject(s)
Brain/metabolism , Peptides/physiology , Spinal Cord/metabolism , Adrenomedullin , Animals , Brain/blood supply , Humans , Hypoxia , Immunohistochemistry , Ischemia , Mice , Microscopy, Electron , Peptides/metabolism , Rats , Spinal Cord/blood supply
16.
Brain Res ; 909(1-2): 20-45, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11478918

ABSTRACT

A perfusion model of global cerebral ischemia was used for the immunohistochemical study of changes in the glutamate-nitric oxide (NO) system in the rat cerebellum and cerebellar nuclei during a 0-14 h reperfusion period after 30 min of oxygen and glucose deprivation, with and without administration of 1.5 mM N(omega)-nitro-L-arginine methyl ester (L-NAME). While immunostaining for N-methyl-D-aspartate receptor subunit 1 (NMDAR1) showed no marked changes during the reperfusion period, neuronal NO synthase (nNOS) immunostaining increased in stellate and basket cells, granule cells and neurons of the cerebellar nuclei. However, global cerebellar nNOS concentrations determined by Western blotting remained largely unchanged in comparison with actin expression. Inducible NOS (iNOS) immunostaining appeared in Purkinje cells and neurons of the cerebellar nuclei after 2-4 h of reperfusion and intensified during the 6-14 h period. This was reflected by an increase in global cerebellar iNOS expression determined by Western blotting. Immunostaining for protein nitrotyrosine was seen in Purkinje cells, stellate and basket cells, neurons of the cerebellar nuclei and glial cells in controls, and showed a progressive translocation in Purkinje cells and neurons of the cerebellar nuclei from an initial perinuclear or nuclear location towards the periphery. At the end of the reperfusion period the Purkinje cell apical dendrites were notably retracted and tortuous. Prior and concurrent L-NAME administration eliminated nitrotyrosine immunostaining in controls and blocked or reduced most of the postischemic changes observed. The results suggest that while nNOS expression may be modified in certain cells, iNOS is induced after a 2-4 h period, and that changes in protein nitration may be associated with changes in cell morphology.


Subject(s)
Cerebellum/enzymology , Glucose/deficiency , Hypoxia-Ischemia, Brain/enzymology , Neurons/enzymology , Nitrates/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Tyrosine/analogs & derivatives , Animals , Blotting, Western , Cerebellum/pathology , Cerebellum/physiopathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Immunohistochemistry , Male , NG-Nitroarginine Methyl Ester/pharmacology , Neurons/drug effects , Neurons/pathology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/isolation & purification , Purkinje Cells/drug effects , Purkinje Cells/enzymology , Purkinje Cells/pathology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Tyrosine/metabolism
18.
Brain Res ; 853(2): 245-68, 2000 Jan 24.
Article in English | MEDLINE | ID: mdl-10640622

ABSTRACT

Adrenomedullin is a peptide of marked vasodilator activity first isolated from human pheochromocytoma and subsequently demonstrated in other mammalian tissues. Using a polyclonal antiserum against human adrenomedullin-(22-52) amide and the avidin-biotin peroxidase complex technique, we have demonstrated by light and electron microscopy that adrenomedullin-like immunoreactivity is widely distributed in the rat central nervous system. Western blotting of extracts of different brain regions demonstrated the fully processed peptide as the major form in the cerebellum, whereas a 14-kDa molecular species and a small amount of the 18-kDa propeptide were present in other brain regions. Immunoreactive neurons and processes were found in multipolar neurons and pyramidal cells of layers IV-VI of the cerebral cortex and their apical processes, as well as in a large number of telencephalic, diencephalic, mesencephalic, pontine and medullary nuclei. Cerebellar Purkinje cells and mossy terminal nerve fibers as well as neurons of the cerebellar nuclei were immunostained, as were neurons in area 9 of the anterior horn of the spinal cord. Immunoreactivity was also found in some vascular endothelial cells and surrounding processes that probably originated from perivascular glial cells. Electron microscopy confirmed the light microscopy findings and showed the reaction product in relation to neurofilaments and the external membrane of small mitochondria. Immunoreactive terminal boutons were occasionally seen. The distribution of adrenomedullin-like immunoreactivity in the central nervous system suggests that it has a significant role in neuronal function as well as in the regulation of regional blood flow.


Subject(s)
Brain/metabolism , Peptide Fragments/metabolism , Spinal Cord/metabolism , Adrenomedullin , Animals , Antibodies/metabolism , Antibody Specificity , Axons/metabolism , Axons/ultrastructure , Brain/blood supply , Brain/ultrastructure , Dendrites/metabolism , Dendrites/ultrastructure , Male , Microcirculation/metabolism , Microcirculation/ultrastructure , Microtubules/metabolism , Microtubules/ultrastructure , Mitochondria/metabolism , Mitochondria/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Peptide Fragments/immunology , Purkinje Cells/metabolism , Purkinje Cells/ultrastructure , Rats , Rats, Wistar , Spinal Cord/blood supply , Spinal Cord/ultrastructure , Synapses/metabolism , Synapses/ultrastructure
19.
Brain Res Dev Brain Res ; 111(2): 205-22, 1998 Dec 07.
Article in English | MEDLINE | ID: mdl-9838118

ABSTRACT

The expression of neuronal nitric oxide synthase (nNOS) during the development of the rat cerebral cortex from embryonic day (E) 13 to postnatal day (P) 0 was analyzed by immunocytochemical procedures using a specific antibody against rat brain nNOS. Expression of nNOS was first seen on E14 in cells of Cajal-Retzius morphology located in the marginal zone. Neuronal NOS immunoreactivity persisted in this layer throughout the embryonic period and only began to decrease on E20, when neuronal migration is coming to an end. From E17 onwards, migrating neurons expressing nNOS were observed in the intermediate zone with their leading processes directed towards the cortical plate. At the same time, efferent nNOS-immunoreactive axons originating from cortical plate cells entered the intermediate zone. From E19 onwards, cells expressing nNOS and with the morphological characteristics of migrating cells were observed in and near the subventricular zone. Confocal analysis of double immunostaining for nNOS and glial fibrillary acidic protein or nestin showed no coexpression of nNOS and glial markers in these cells, suggesting that nNOS-positive cells leaving the subventricular zone were not glial cells. Commissural, callosal and fimbrial fibers were seen to express nNOS on E18 and E19. This expression decreased from E20 and was very weak on E21 and P0. The observations suggest that nitric oxide is synthesized during embryonic life in relation to maturational processes such as the organization of cerebral lamination, and is involved in controlling migrational processes and fiber ingrowth.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/enzymology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Nitric Oxide Synthase/genetics , Animals , Cerebral Cortex/cytology , Female , Fluorescent Antibody Technique , Neuroglia/enzymology , Neurons/enzymology , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type I , Pregnancy , Rabbits , Rats
20.
Microsc Res Tech ; 43(1): 75-88, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9829462

ABSTRACT

Neuronal and inducible nitric oxide synthase (nNOS and iNOS) and nitrotyrosine immunoreactivities were localized and semiquantitatively assessed in the cerebral cortex of aged rats by means of light microscopic immunocytochemistry and Western blotting, using a new series of specific polyclonal antibodies. In the aged rats the strongly nNOS-immunoreactive multipolar neurons found in layers II-VI of the cortex of young rats were seen in similar numbers, but showed varicose, vacuolated, and fragmented processes, with an irregular outline and loss of spines. A large number of more weakly nNOS-positive neurons, characterized by a ring of immunoreactive cytoplasm, and not seen in young rats, were observed in layers II-VI of aged rat cortex. While no iNOS-immunopositive neurons were found in the cortex of young rats, a large number of such neurons appeared throughout the aged rat cortex. Nitrotyrosine-positive cells outnumbered total NOS-positive neurons in the cortex of young rats, but this relation was inverted in the aged rats, although these showed a slight increase in the number and staining intensity of nitrotyrosine-positive cells. Western blots of brain extracts showed a several-fold increase in both nNOS- and iNOS-immunoreactive bands in the aged rat, but a less marked increase in nitrotyrosine-containing proteins. The results suggest that while nNOS and iNOS expression is substantially increased in the aged rat cortex, this is not necessarily accompanied by a proportionate increase in nitric oxide synthesis. The mechanisms underlying the increased expression of nNOS and iNOS, and the functional implications of this increase, require elucidation.


Subject(s)
Aging/pathology , Cerebral Cortex/chemistry , Nerve Tissue Proteins/analysis , Nitric Oxide Synthase/analysis , Tyrosine/analysis , Albinism , Animals , Blotting, Western , Cerebral Cortex/pathology , Immunohistochemistry , Male , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Rats , Rats, Wistar , Tyrosine/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL
...