Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38673761

ABSTRACT

Multiple sclerosis (MS) is an autoimmune, inflammatory, and neurodegenerative disease of the central nervous system for which there is no cure, making it necessary to search for new treatments. The endocannabinoid system (ECS) plays a very important neuromodulatory role in the CNS. In recent years, the formation of heteromers containing cannabinoid receptors and their up/downregulation in some neurodegenerative diseases have been demonstrated. Despite the beneficial effects shown by some phytocannabinoids in MS, the role of the ECS in its pathophysiology is unknown. The main objective of this work was to identify heteromers of cell surface proteins receptive to cannabinoids, namely GPR55, CB1 and CB2 receptors, in brain samples from control subjects and MS patients, as well as determining their cellular localization, using In Situ Proximity Ligation Assays and immunohistochemical techniques. For the first time, CB1R-GPR55 and CB2R-GPR55 heteromers are identified in the prefrontal cortex of the human brain, more in the grey than in the white matter. Remarkably, the number of CB1R-GPR55 and CB2R-GPR55 complexes was found to be increased in MS patient samples. The results obtained open a promising avenue of research on the use of these receptor complexes as potential therapeutic targets for the disease.


Subject(s)
Multiple Sclerosis , Prefrontal Cortex , Receptor, Cannabinoid, CB1 , Receptor, Cannabinoid, CB2 , Receptors, Cannabinoid , Humans , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Prefrontal Cortex/metabolism , Receptors, Cannabinoid/metabolism , Receptor, Cannabinoid, CB2/metabolism , Receptor, Cannabinoid, CB1/metabolism , Male , Adult , Female , Receptors, G-Protein-Coupled/metabolism , Middle Aged , Up-Regulation , Protein Multimerization
2.
Cell Death Dis ; 14(11): 761, 2023 11 23.
Article in English | MEDLINE | ID: mdl-37996408

ABSTRACT

Pancreatic cancer cells with mutant KRAS require strong basal autophagy for viability and growth. Here, we observed that some processes that allow the maintenance of basal autophagy in pancreatic cancer cells are controlled by protein methylation. Thus, by maintaining the methylation status of proteins such as PP2A and MRAS, these cells can sustain their autophagic activity. Protein methylation disruption by a hypomethylating treatment (HMT), which depletes cellular S-adenosylmethionine levels while inducing S-adenosylhomocysteine accumulation, resulted in autophagy inhibition and endoplasmic reticulum stress-induced apoptosis in pancreatic cancer cells. We observed that by reducing the membrane localization of MRAS, hypomethylation conditions produced an imbalance in KRAS signaling, resulting in the partial inactivation of ERK and hyperactivation of the PI3K/AKT-mTORC1 pathway. Interestingly, HMT impeded CRAF activation by disrupting the ternary SHOC2 complex (SHOC2/MRAS/PP1), which functions as a CRAF-S259 holophosphatase. The demethylation events that resulted in PP2A inactivation also favored autophagy inhibition by preventing ULK1 activation while restoring the cytoplasmic retention of the MiT/TFE transcription factors. Since autophagy provides pancreatic cancer cells with metabolic plasticity to cope with various metabolic stress conditions, while at the same time promoting their pathogenesis and resistance to KRAS pathway inhibitors, this hypomethylating treatment could represent a therapeutic opportunity for pancreatic adenocarcinomas.


Subject(s)
Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Phosphatidylinositol 3-Kinases , Methylation , Cell Line, Tumor , Pancreatic Neoplasms/genetics , Autophagy/genetics , Intracellular Signaling Peptides and Proteins
3.
Brain Sci ; 11(2)2021 Feb 22.
Article in English | MEDLINE | ID: mdl-33671675

ABSTRACT

Suitable in vivo and in vitro models are instrumental for the development of new drugs aimed at improving symptoms or progression of multiple sclerosis (MS). The cuprizone (CPZ)-induced murine model has gained momentum in recent decades, aiming to address the demyelination component of the disease. This work aims at assessing the differential cytotoxicity of CPZ in cells of different types and from different species: human oligodendroglial (HOG), human neuroblastoma (SH-SY5Y), human glioblastoma (T-98), and mouse microglial (N-9) cell lines. Moreover, the effect of CPZ was investigated in primary rat brain cells. Cell viability was assayed by oxygen rate consumption and by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide-based (MTT) method. Our results demonstrated that CPZ did not cause death in any of the assayed cell models but affected mitochondrial function and aerobic cell respiration, thus compromising cell metabolism in neural cells and neuron-glia co-cultures. In this sense, we found differential vulnerability between glial cells and neurons as is the case of the CPZ-induced mouse model of MS. In addition, our findings demonstrated that reduced viability was spontaneous reverted in a time-dependent manner by treatment discontinuation. This reversible cell-based model may help to further investigate the role of mitochondria in the disease, and study the molecular intricacies underlying the pathophysiology of the MS and other demyelinating diseases.

4.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33514021

ABSTRACT

Apolipoprotein D (Apo D) overexpression is a general finding across neurodegenerative conditions so the role of this apolipoprotein in various neuropathologies such as multiple sclerosis (MS) has aroused a great interest in last years. However, its mode of action, as a promising compound for the development of neuroprotective drugs, is unknown. The aim of this work was to address the potential of Apo D to prevent the action of cuprizone (CPZ), a toxin widely used for developing MS models, in oligodendroglial and neuroblastoma cell lines. On one hand, immunocytochemical quantifications and gene expression measures showed that CPZ compromised neural mitochondrial metabolism but did not induce the expression of Apo D, except in extremely high doses in neurons. On the other hand, assays of neuroprotection demonstrated that antipsychotic drug, clozapine, induced an increase in Apo D synthesis only in the presence of CPZ, at the same time that prevented the loss of viability caused by the toxin. The effect of the exogenous addition of human Apo D, once internalized, was also able to directly revert the loss of cell viability caused by treatment with CPZ by a reactive oxygen species (ROS)-independent mechanism of action. Taken together, our results suggest that increasing Apo D levels, in an endo- or exogenous way, moderately prevents the neurotoxic effect of CPZ in a cell model that seems to replicate some features of MS which would open new avenues in the development of interventions to afford MS-related neuroprotection.


Subject(s)
Apolipoproteins D/genetics , Demyelinating Diseases/genetics , Multiple Sclerosis/genetics , Oligodendroglia/metabolism , Animals , Cell Line , Cell Survival/drug effects , Cuprizone/toxicity , Demyelinating Diseases/pathology , Demyelinating Diseases/therapy , Disease Models, Animal , Humans , Mice , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Neuroprotective Agents/therapeutic use , Oligodendroglia/drug effects , Oligodendroglia/pathology , Reactive Oxygen Species/metabolism
5.
Front Aging Neurosci ; 10: 254, 2018.
Article in English | MEDLINE | ID: mdl-30186153

ABSTRACT

Apolipoprotein D (Apo D) is a key molecule in the lipid transport during homeostasis and repair processes in normal and pathological conditions of the nervous system with a putative neuroprotective effect. In the last decades, huge experimental efforts have been made to know the exact mechanism of action of Apo D, even though, it remains an open question. In this regard, studies in mammals and flies have suggested that Apo D seems to act through a variety of cellular mechanisms related with its ability to selectively bind different lipid ligands. For instance, this apolipoprotein is required to myelin compaction, it participates in axon regeneration/remyelination, and it can control the magnitude and timing of the inflammatory response after injury, promoting myelin clearance, and regulating the number of immune cells recruited to the damaged area. These, among others, are some of the reasons to study Apo D in multiple sclerosis (MS) pathology, where it could be particularly important since the autoimmune reaction against oligodendrocytes (OLGs) and myelin is generally assumed as the most plausible cause of this pathology. The aim of this work was to investigate the Apo D expression pattern in MS lesions, including active and inactive demyelinating plaques, and also remyelinating ones. Human brain tissues with inflammatory demyelination consistent with MS were used to quantify Apo D immunosignal in different lesions. Our results show a clear decrease of Apo D expression in all sclerosis plaques, being lower in the inactive than in active areas but recovers in the remyelination ones. Apo D is mainly produced by the matured OLGs of white matter and is located in cell processes surrounding the myelin sheath. All these data seem to indicate an important role of Apo D in myelination/remyelination processes as a molecule with a neuroprotective potential, and may serve as a good starting point for its study in MS.

6.
Front Aging Neurosci ; 8: 213, 2016.
Article in English | MEDLINE | ID: mdl-27679571

ABSTRACT

BACKGROUND: Learning processes or language development are only some of the cognitive functions that differ qualitatively between men and women. Gender differences in the brain structure seem to be behind these variations. Indeed, this sexual dimorphism at neuroanatomical level is accompanied unequivocally by differences in the way that aging and neurodegenerative diseases affect men and women brains. OBJECTIVE: The aim of this study is the analysis of neuronal density in four areas of the hippocampus, and entorhinal and frontal cortices to analyze the possible gender influence during normal aging and in Alzheimer's disease (AD). METHODS: Human brain tissues of different age and from both sexes, without neurological pathology and with different Braak's stages of AD, were studied. Neuronal density was quantified using the optical dissector. RESULTS: Our results showed the absence of a significant neuronal loss during aging in non-pathological brains in both sexes. However, we have demonstrated specific punctual significant variations in neuronal density related with the age and gender in some regions of these brains. In fact, we observed a higher neuronal density in CA3 and CA4 hippocampal areas of non-pathological brains of young men compared to women. During AD, we observed a negative correlation between Braak's stages and neuronal density in hippocampus, specifically in CA1 for women and CA3 for men, and in frontal cortex for both, men and women. CONCLUSION: Our data demonstrated a sexual dimorphism in the neuronal vulnerability to degeneration suggesting the need to consider the gender of the individuals in future studies, regarding neuronal loss in aging and AD, in order to avoid problems in interpreting data.

7.
J Alzheimers Dis ; 53(2): 639-50, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27197790

ABSTRACT

Apolipoprotein D (Apo D) and Apolipoprotein J (Apo J) are among the only nine apolipoproteins synthesized in the nervous system. Apart from development, these apolipoproteins are implicated in the normal aging process as well as in different neuropathologies as Alzheimer's disease (AD), where a neuroprotective role has been postulated. Different authors have proposed that Apo D and Apo J could be biomarkers for AD but as far as we know, there are no studies about the relationship between them as well as their expression pattern along the progression of the disease. In this paper, using double immunohistochemistry techniques, we have demonstrated that Apo D is mainly located in glial cells while Apo J expression preferentially occurs in neurons; both proteins are also present in AD diffuse and mature senile plaques but without signal overlap. In addition, we have observed that Apo J and Apo D immunostaining shows a positive correlation with the progression of the disease and the Braak's stages. These results suggest complementary and cell-dependent neuroprotective roles for each apolipoprotein during AD progress.


Subject(s)
Alzheimer Disease/pathology , Apolipoproteins D/metabolism , Brain/metabolism , Clusterin/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Brain/pathology , Disease Progression , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Middle Aged , Neurofibrillary Tangles/pathology , Neurons/pathology , Plaque, Amyloid/pathology , Psychiatric Status Rating Scales , Statistics, Nonparametric
8.
Invest Ophthalmol Vis Sci ; 56(8): 4835-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26218912

ABSTRACT

PURPOSE: Mutations in the OPA1 gene cause autosomal dominant optic atrophy (ADOA), a visual disorder associated with degeneration of retinal ganglion cells (RGCs). Here, we characterized the disease progression in a homologous mouse model B6;C3-Opa1 329-355del and asked whether the pronounced cell death affects certain RGC types more than others. METHODS: The influence of the Opa1 mutation was assessed by morphologic (retina and optic nerve histology) and functional (multielectrode array) methods. RESULTS: The RGC loss of approximately 50% within 18 months was significantly more pronounced in RGCs with small-caliber axons. Small-caliber axon RGCs comprise a variety of functional RGC types. Accordingly, electrophysiological analyses of RGCs did not show a dropout of distinct functional RGC subgroups. However, the response properties of RGCs were affected significantly by the mutation. Surprisingly, these functional changes were different under different luminance conditions (scotopic, mesopic, and photopic). Finally, melanopsin cells are known to be less susceptible to retinal insults. We found that these cells are also spared in the Opa1 mouse model, and demonstrated for the first time that this resistance persisted even when the melanopsin gene had been knocked-out. CONCLUSIONS: Small-caliber axons show a higher vulnerability to the Opa1 mutation in our mouse model for ADOA. Luminance-dependent functional changes suggest an influence of the Opa1 mutation on the retinal circuitry upstream of RGCs. Photoresponsive RGCs are protected against cell death due to the Opa1 mutation, but not by melanopsin expression itself.


Subject(s)
DNA/genetics , GTP Phosphohydrolases/genetics , Mutation , Optic Atrophy, Autosomal Dominant/genetics , Retinal Ganglion Cells/pathology , Animals , DNA Mutational Analysis , Disease Models, Animal , Female , GTP Phosphohydrolases/metabolism , Immunohistochemistry , Male , Mice , Mice, Mutant Strains , Optic Atrophy, Autosomal Dominant/metabolism , Optic Atrophy, Autosomal Dominant/pathology , Retinal Ganglion Cells/metabolism
9.
Acta Histochem ; 117(6): 536-44, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25953740

ABSTRACT

Apolipoprotein D (Apo D) is a secreted glycoprotein, member of the lipocalin superfamily, with a related beneficial role in metabolism and lipid transport due to the presence of a binding pocket that allows its interaction with several lipids. Nowadays, it has been clearly demonstrated that Apo D expression is induced and its subcellular location undergoes modifications in stressful and pathological conditions that characterize aging processes and neurodegenerative diseases. The aim of the present work was to study in detail the effect of H2O2 on the subcellular location of Apo D, in the hippocampal cell line HT22, by structural, ultrastructural, immunocytochemical, and molecular techniques in order to characterize the Apo D distribution pattern in neurons during oxidative stress. Our results indicate that Apo D is located in the cytoplasm under physiological conditions but treatment with H2O2 induces apoptosis and causes a displacement of Apo D location to the nucleus, coinciding with DNA fragmentation. In addition, we demonstrated that Apo D tends to accumulate around the nuclear envelope in neurons and glial cells of different brain areas in some neurodegenerative diseases and during human aging, but never inside the nucleus. These data suggest that the presence of Apo D in the nucleus, which some authors related with a specific transport, is a consequence of structural and functional alterations during oxidative stress and not the result of a specific role in the regulation of nuclear processes.


Subject(s)
Apolipoproteins D/metabolism , Neurons/metabolism , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , DNA Fragmentation/drug effects , Humans , Hydrogen Peroxide/pharmacology , Immunohistochemistry , In Vitro Techniques , Microscopy, Electron, Transmission , Neurons/drug effects , Oxidative Stress/drug effects
10.
PLoS One ; 8(10): e77852, 2013.
Article in English | MEDLINE | ID: mdl-24167586

ABSTRACT

The lipocalin apolipoprotein D (Apo D) is upregulated in peripheral nerves following injury and in regions of the central nervous system, such as the cerebral cortex, hippocampus, and cerebellum, during aging and progression of certain neurological diseases. In contrast, few studies have examined Apo D expression in the brainstem, a region necessary for survival and generally less prone to age-related degeneration. We measured Apo D expression in whole human brainstem lysates by slot-blot and at higher spatial resolution by quantitative immunohistochemistry in eleven brainstem nuclei (the 4 nuclei of the vestibular nuclear complex, inferior olive, hypoglossal nucleus, oculomotor nucleus, facial motor nucleus, nucleus of the solitary tract, dorsal motor nucleus of the vagus nerve, and Roller`s nucleus). In contrast to cortex, hippocampus, and cerebellum, apolipoprotein D was highly expressed in brainstem tissue from subjects (N = 26, 32-96 years of age) with no history of neurological disease, and expression showed little variation with age. Expression was significantly stronger in somatomotor nuclei (hypoglossal, oculomotor, facial) than visceromotor or sensory nuclei. Both neurons and glia expressed Apo D, particularly neurons with larger somata and glia in the periphery of these brainstem centers. Immunostaining was strongest in the neuronal perinuclear region and absent in the nucleus. We propose that strong brainstem expression of Apo D throughout adult life contributes to resistance against neurodegenerative disease and age-related degeneration, possibly by preventing oxidative stress and ensuing lipid peroxidation.


Subject(s)
Aging/metabolism , Apolipoproteins D/biosynthesis , Brain Stem/metabolism , Gene Expression Regulation , Nerve Tissue Proteins/biosynthesis , Neurodegenerative Diseases/metabolism , Adult , Aged , Aged, 80 and over , Aging/pathology , Brain Stem/pathology , Female , Humans , Lipid Peroxidation , Male , Middle Aged , Neurodegenerative Diseases/pathology , Retrospective Studies
11.
J Alzheimers Dis ; 36(1): 129-44, 2013.
Article in English | MEDLINE | ID: mdl-23568103

ABSTRACT

Apolipoprotein D (Apo D) is a lipid binding protein whose expression is strongly induced in the mammalian brain during aging and age-dependent neurodegenerative diseases such as Alzheimer's disease (AD), where it can play an important function as a neuroprotective and antioxidant protein. Increasing evidence suggests that the gradual increase in free radicals and oxidative stress with age is the primary determinant to aging brain. The aim of this work is to study the effect of hydrogen peroxide (H2O2) in Apo D expression, in hippocampal cells, in order to investigate the relationship between oxidative stress and elevated levels of Apo D found in hippocampus during aging and AD and also elucidate the possible pathways that lead to this increase. In this study, we demonstrated that Apo D expression in hippocampal neurons of aged and AD brains directly correlates with age-related increase in oxidative stress. More importantly, our results in the HT22 cell line indicate that Apo D protein level increases in a concentration-dependent manner specifically at those H2O2 concentrations that caused oxidative damage and apoptotic cell death. These data support the idea that oxidative stress-induced apoptosis during aging and AD may be associated with the increment in the expression of Apo D in these situations.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Apolipoproteins D/biosynthesis , Hippocampus/metabolism , Oxidative Stress/physiology , Adult , Aged , Aged, 80 and over , Aging/physiology , Alzheimer Disease/physiopathology , Apoptosis , Blotting, Western , Case-Control Studies , Cells, Cultured , Flow Cytometry , Hippocampus/physiopathology , Humans , Microscopy, Electron , Middle Aged
12.
J Alzheimers Dis ; 35(3): 589-97, 2013.
Article in English | MEDLINE | ID: mdl-23455992

ABSTRACT

A highly selective, rapid, inexpensive, simple, and immunocytochemical compatible fluorescence staining method for Alzheimer's disease hallmark lesions applicable to sections of human specimens embedded in paraffin is described. Human necropsy material was fixed in buffered formalin, sectioned at 10 µm, mounted on slides, deparaffinized, and partially hydrated (70% ethanol). After partial hydration, sections were stained for 10 min in a solution of 0.2% Congo red in 70% isopropanol. After washing in 70% isopropanol and rehydration, auto-fluorescence of sections were quenched (optional) and processed for immunocytochemistry (optional). Finally, sections were mounted in an adequate mounting medium. Amyloid deposits appear pink at light microscopy and all Alzheimer's disease hallmark lesions appear orange or red under fluorescence microscopy using blue or green exciting light, respectively. The present method can be used in combination with all pre- or post-immunocytochemical techniques.


Subject(s)
2-Propanol , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Amyloidosis/diagnosis , Amyloidosis/pathology , Brain/pathology , Congo Red , Aged , Blood Vessels/pathology , Brain/blood supply , Cerebral Amyloid Angiopathy/pathology , Humans , Microscopy, Fluorescence , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Solutions
13.
Age (Dordr) ; 35(3): 821-37, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22648398

ABSTRACT

Estrogens are not only critical for sexual differentiation it is well-known for the role of 17ß-estradiol (E2) in the adult brain modulating memory, learning, mood and acts as a neuroprotector. E2 exerts its actions through two classical receptors: estrogen receptor alpha (ERα) and estrogen receptor beta (ERß). The distribution of both receptors changes from one brain area to another, E2 being able to modulate their expression. Among the classical features of aging in humans, we find cognitive impairment, dementia, memory loss, etc. As estrogen levels change with age, especially in females, it is important to know the effects of low E2 levels on ERα distribution; results from previous studies are controversial regarding this issue. In the present work, we have studied the effects of long-term E2 depletion as well as the ones of E2 treatment on ERα brain distribution of ovariectomized rats along aging in the diencephalon and in the telencephalon. We have found that ovariectomy causes downregulation and affects subcellular localization of ERα expression during aging, meanwhile prolonged estrogen treatment produces upregulation and overexpression of the receptor levels. Our results support the idea of the region-specific neuroprotection mechanisms mediated by estradiol.


Subject(s)
Aging/metabolism , Brain Chemistry/physiology , Brain/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Hormone Replacement Therapy , Intracellular Space/metabolism , Aging/drug effects , Animals , Brain/cytology , Brain/drug effects , Cerebral Cortex/chemistry , Cerebral Cortex/cytology , Densitometry , Diencephalon/chemistry , Diencephalon/cytology , Diencephalon/drug effects , Estrogens/pharmacology , Female , Immunohistochemistry , Intracellular Space/drug effects , Rats , Rats, Wistar , Telencephalon/chemistry , Telencephalon/cytology , Telencephalon/drug effects
14.
J Alzheimers Dis ; 30(2): 233-44, 2012.
Article in English | MEDLINE | ID: mdl-22398376

ABSTRACT

Apolipoprotein D (ApoD) is a secreted glycoprotein that is markedly induced in several pathological and stressful conditions in the nervous system. In the central nervous system, ApoD expression is upregulated during aging, after traumatic brain injury, and in several human neuropathologies such as Alzheimer's disease (AD), where it is found associated with amyloid-ß (Aß) plaques. Recent studies have indicated that ApoD has an important function as a neuroprotective and antioxidant protein. The aim of this work is to study the effect of the peptide fragment Aß25-35, which is believed to play a major role in the neurodegenerative process of AD, in ApoD expression in a mouse hippocampal cell line. In addition, we studied whether direct addition of exogenous human recombinant ApoD protein has neuroprotective effect against Aß25-35 treatment on neuronal cells. Our results demonstrate that Aß25-35 induces ApoD expression in hippocampal cells in response to stress-induced growth arrest. This observed relationship between Aß and ApoD expression could explain the elevated levels of ApoD found in AD brain, where it may be a neuroprotective molecule in the course of AD, probably related to its lipid transport function or a direct antioxidant property. However, the addition of exogenous human recombinant ApoD does not exert any protective effect, most likely due to its major structural modifications.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoproteins D/metabolism , Hippocampus/cytology , Neurons/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Apolipoproteins D/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/physiology , Humans , Mice , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/cytology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peptide Fragments/toxicity , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
15.
Neurobiol Aging ; 33(2): 433.e11-20, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21429623

ABSTRACT

Apolipoprotein D (Apo D) is a lipocalin expressed in a wide variety of mammalian tissues. Different studies have shown that this protein is upregulated in the central nervous system (CNS) in several neuropathological conditions, after traumatic brain injury and in aging. The Apo D promoter shows 3 estrogen response elements and it has been shown that its expression is influenced by estrogens in breast cyst fluid. The aim of this work is to study the possible relationship between gender and Apo D expression in human hippocampus and in the entorhinal and frontal cortices during aging and Alzheimer's disease (AD). We visualized Apo D immunohistochemically and then performed a quantification of the chromogen signal strength. Our findings show that Apo D expression is influenced by age, Braak stage, and sex. In most of the studied areas, Apo D expression is increased with age in women but not in men, and in AD progression in both genders. Apo D is always expressed by neurons with no signs of degeneration or death.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Adult , Aged , Aged, 80 and over , Apolipoproteins D , Female , Humans , Male , Middle Aged , Sex Factors , Tissue Distribution
16.
Age (Dordr) ; 34(4): 895-904, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21761133

ABSTRACT

Aging is associated with a reduction in metabolic functions, increased incidence of neurodegenerative diseases, and memory or cognitive dysfunction. With aging, a decrease in plasma estrogen levels, related to loss of gonadal function, occurs in females. Estrogens have neuroprotective effects and estradiol treatment improves some aspects of neuronal homeostasis affected by aging. In other way, recent studies show that apo D can play a neuroprotective role in some neuropathologies and during aging. The possible relation between estradiol treatment and the expression of apo D, during aging in the CNS, was investigated in female rats. Our results confirm an expression of apo D zone-dependent, in relation with aging, and an overexpression of apo D related to ovariectomy and estradiol treatment. This overexpression strengthens the idea that apo D plays a neuroprotective role in the CNS.


Subject(s)
Aging/drug effects , Apolipoproteins D/metabolism , Central Nervous System/drug effects , Estradiol/pharmacology , Aging/physiology , Analysis of Variance , Animals , Apolipoproteins D/genetics , Central Nervous System/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Estradiol/blood , Female , Gene Expression Regulation , Neuroprotective Agents , Ovariectomy/methods , Random Allocation , Rats , Rats, Wistar , Reference Values , Sensitivity and Specificity
17.
Glia ; 58(11): 1320-34, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20607718

ABSTRACT

Glial cells are a key element to the process of axonal regeneration, either promoting or inhibiting axonal growth. The study of glial derived factors induced by injury is important to understand the processes that allow or preclude regeneration, and can explain why the PNS has a remarkable ability to regenerate, while the CNS does not. In this work we focus on Apolipoprotein D (ApoD), a Lipocalin expressed by glial cells in the PNS and CNS. ApoD expression is strongly induced upon PNS injury, but its role has not been elucidated. Here we show that ApoD is required for: (1) the maintenance of peripheral nerve function and tissue homeostasis with age, and (2) an adequate and timely response to injury. We study crushed sciatic nerves at two ages using ApoD knock-out and transgenic mice over-expressing human ApoD. The lack of ApoD decreases motor nerve conduction velocity and the thickness of myelin sheath in intact nerves. Following injury, we analyze the functional recovery, the cellular processes, and the protein and mRNA expression profiles of a group of injury-induced genes. ApoD helps to recover locomotor function after injury, promoting myelin clearance, and regulating the extent of angiogenesis and the number of macrophages recruited to the injury site. Axon regeneration and remyelination are delayed without ApoD and stimulated by excess ApoD. The mRNA and protein expression profiles reveal that ApoD is functionally connected in an age-dependent manner to specific molecular programs triggered by injury.


Subject(s)
Apolipoproteins D/physiology , Cellular Senescence/physiology , Nerve Regeneration/physiology , Neuroglia/metabolism , Neuroglia/pathology , Peripheral Nerves/metabolism , Peripheral Nerves/pathology , Recovery of Function/physiology , Animals , Apolipoproteins D/biosynthesis , Apolipoproteins D/deficiency , Cellular Senescence/genetics , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Crush , Nerve Regeneration/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/physiology , Peripheral Nerves/physiopathology , RNA, Messenger/biosynthesis , Reaction Time/genetics , Reaction Time/physiology , Recovery of Function/genetics , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Sciatic Neuropathy/physiopathology
18.
Age (Dordr) ; 32(1): 85-96, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19936966

ABSTRACT

Apolipoprotein D (apo D) is a lipocalin present in the nervous system that may be related to processes of reinnervation, regeneration and neuronal cell protection. On the other hand, apo D expression has been correlated, in some brain regions, with normal ageing and neurodegenerative diseases. To elucidate the regional and cellular expression of apo Din normal human brain during ageing, we performed a detailed and extensive study in samples of post-mortem human cerebral cortices. To achieve this study, slot-blot techniques, for protein and mRNA,as well as immunohistochemistry and hybridohistochemistry methods, were used. A positive correlation for apo D expression with ageing was found;furthermore, mRNA levels, as well as the protein ones, were higher in the white than in the grey matter. Immunohistochemistry and non-isotopic in situ hybridization showed that apo D is synthesised in both neurons and glial cells. Apo D expression is notorious in oligodendrocytes, but with ageing, the number of neurons that synthesise apo D is increased.Our results indicate that apo D could play a fundamental role in central nervous system ageing and in the reduction of products derived from lipid peroxidation. The increment in the expression of apo D with ageing can be included in a global mechanism of cellular protection to prevent the deleterious effects caused by ageing.


Subject(s)
Aging/metabolism , Apolipoproteins D/biosynthesis , Frontal Lobe/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Humans , Immunoblotting/methods , Immunoenzyme Techniques , In Situ Hybridization , In Vitro Techniques , Linear Models , Lipid Peroxidation , Middle Aged , RNA, Messenger/metabolism , Statistics, Nonparametric
19.
Histol Histopathol ; 23(8): 995-1001, 2008 08.
Article in English | MEDLINE | ID: mdl-18498075

ABSTRACT

Apolipoprotein D (apo D), a lipocalin transporter of small hydrophobic molecules could play an important role in several neurodegenerative diseases. However, its role in those diseases remains unclear. There has been reported increments of apo D in relation with different neuropathologic diseases. Recently, we reported the absence of apo D in neurons of substantia nigra which can contribute to the lability of neurons to oxidative damage. In order to determine the relationship between apo D expression and neuronal death, we studied the expression of apo D in various regions of human brains from patients without any neurological or psychological disorders, in relation with the neuronal damage revealed by Fluoro-Jade B staining. The absence of expression for apo D in injured neurons and the negative staining for Fluoro-Jade B of neurons that express apo D was observed in all sections studied. These findings are in accordance with the role possibly played by apo D in the neuroprotection of the nervous system.


Subject(s)
Apolipoproteins D/metabolism , Central Nervous System/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Central Nervous System/pathology , Fluoresceins , Fluorescent Dyes , Humans , Immunoenzyme Techniques , Middle Aged , Nerve Degeneration/pathology , Neurons/pathology , Organic Chemicals
20.
Endocrinology ; 149(1): 57-72, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17901235

ABSTRACT

Aging is associated with a reduction in metabolic function, insulin resistance, increased incidence of neurodegenerative diseases, and memory or cognitive dysfunction. In aging females, loss of gonadal function determines the beginning of the period of reduced metabolic function. Estrogens have neuroprotective effects, but the mechanisms by which they exert these effects remain unclear. The effects of estradiol treatment on the activation of the insulin receptor substrate (IRS)-1 signaling pathway, the interactions between estrogen receptor (ER)-alpha and IRS-1 and the p85alpha subunit of phosphatidylinositol-3 kinase, together with the possible effects of estradiol treatment on glucose transporter-3 and -4 levels, were investigated in female rats. The level of expression of each glucose transporter was greater in control and estradiol-treated groups than in the ovariectomized group. Interactions of ERalpha46-IRS-1, ERalpha46-p85alpha, and p85alpha-IRS-1, as well as IRS-1 phosphorylation, appeared to increase with estradiol treatment. The results indicate that estradiol treatment improves some aspects of neuronal homeostasis that are affected by aging; this may indicate that estradiol has neuroprotective effects in female rats. Additional animal studies are required to clarify the neuroprotective role of estradiol in relation to other important molecules involved in the IRS-1-phosphatidylinositol-3 kinase signaling pathway.


Subject(s)
Aging/physiology , Brain/drug effects , Brain/physiology , Estradiol/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Aging/drug effects , Animals , Brain/metabolism , Estradiol/blood , Estrogen Receptor alpha/metabolism , Female , Glucose Transporter Type 3/metabolism , Glucose Transporter Type 4/metabolism , Homeostasis/drug effects , Insulin Receptor Substrate Proteins , Neuroprotective Agents/blood , Neuroprotective Agents/pharmacology , Ovariectomy , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Rats , Rats, Wistar , Signal Transduction/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...