Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Domest Anim Endocrinol ; 52: 82-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25929245

ABSTRACT

Apolipoprotein D (APOD) is a glycoprotein which is widely expressed in mammalian tissues. It is structurally and functionally similar to the lipocalins which are multiple lipid-binding proteins that transport hydrophobic ligands and other small hydrophobic molecules, including cholesterol and several steroid hormones. Although multiple functions for APOD in various tissues have been reported, its expression, biological function, and hormonal regulation in the female reproductive system are not known. Thus, in this study, we focused on correlations between APOD and estrogen during development, differentiation, regression, and regeneration of the oviduct in chickens and in the development of ovarian carcinogenesis in laying hens. Results of the present study indicated that APOD messenger RNA (mRNA) expression increased (P < 0.001) in the luminal and glandular (GE) epithelia of the chicken oviduct in response to diethylstilbestrol (a nonsteroidal synthetic estrogen). In addition, the expression of APOD mRNA and protein decreased (P < 0.001) as the oviduct regressed during induced molting, and gradually increased (P < 0.001) with abundant expression in GE of the oviduct during recrudescence after molting. Furthermore, APOD mRNA and protein were predominantly localized in GE of cancerous, but not normal ovaries from laying hens. Collectively, results of the present study suggest that APOD is a novel estrogen-stimulated gene in the chicken oviduct which likely regulates growth, differentiation, and remodeling of the oviduct during oviposition cycles. Moreover, up-regulated expression of APOD in epithelial cell-derived ovarian cancerous tissue suggests that it could be a candidate biomarker for early detection and treatment of ovarian cancer in laying hens and in women.


Subject(s)
Apolipoproteins D/genetics , Chickens , Diethylstilbestrol/pharmacology , Ovarian Neoplasms/veterinary , Oviducts/physiopathology , Poultry Diseases/physiopathology , Animals , Apolipoproteins D/analysis , Apolipoproteins D/physiology , Female , Gene Expression Regulation/drug effects , Molting/physiology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/physiopathology , Ovary/chemistry , Oviducts/chemistry , Oviducts/growth & development , Oviposition/physiology , RNA, Messenger/analysis
2.
Exp Gerontol ; 67: 19-47, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25868396

ABSTRACT

A detailed knowledge of the mechanisms underlying brain aging is fundamental to understand its functional decline and the baseline upon which brain pathologies superimpose. Endogenous protective mechanisms must contribute to the adaptability and plasticity still present in the healthy aged brain. Apolipoprotein D (ApoD) is one of the few genes with a consistent and evolutionarily conserved up-regulation in the aged brain. ApoD protecting roles upon stress or injury are well known, but a study of the effects of ApoD expression in the normal aging process is still missing. Using an ApoD-knockout mouse we analyze the effects of ApoD on factors contributing to the functional maintenance of the aged brain. We focused our cellular and molecular analyses in the cortex and hippocampus at an age representing the onset of senescence where mortality risks are below 25%, avoiding bias towards long-lived animals. Lack of ApoD causes a prematurely aged brain without altering lifespan. Age-dependent hyperkinesia and memory deficits are accompanied by differential molecular effects in the cortex and hippocampus. Transcriptome analyses reveal distinct effects of ApoD loss on the molecular age-dependent patterns of the cortex and hippocampus, with different cell-type contributions to age-regulated gene expression. Markers of glial reactivity, proteostasis, and oxidative and inflammatory damage reveal early signs of aging and enhanced brain deterioration in the ApoD-knockout brain. The lack of ApoD results in an age-enhanced significant reduction in neuronal calcium-dependent functionality markers and signs of early reduction of neuronal numbers in the cortex, thus impinging upon parameters clearly differentiating neurodegenerative conditions from healthy brain aging. Our data support the hypothesis that the physiological increased brain expression of ApoD represents a homeostatic anti-aging mechanism.


Subject(s)
Aging/metabolism , Apolipoproteins D/physiology , Cerebral Cortex/metabolism , Hippocampus/metabolism , Aging/genetics , Aging/pathology , Aging, Premature/genetics , Aging, Premature/metabolism , Aging, Premature/pathology , Animals , Apolipoproteins D/deficiency , Apolipoproteins D/genetics , Behavior, Animal , Cerebral Cortex/pathology , Cognition Disorders/genetics , Cognition Disorders/metabolism , Cognition Disorders/pathology , Female , Gene Expression Regulation/physiology , Hippocampus/pathology , Male , Mice, Knockout , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oxidative Stress/genetics , Oxidative Stress/physiology , Transcriptome
3.
Neurobiol Aging ; 35(7): 1632-42, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24612673

ABSTRACT

Apolipoprotein D (ApoD) is an ancient member of the lipocalin family with a high degree of sequence conservation from insects to mammals. It is not structurally related to other major apolipoproteins and has been known as a small, soluble carrier protein of lipophilic molecules that is mostly expressed in neurons and glial cells within the central and peripheral nervous system. Recent data indicate that ApoD not only supplies cells with lipophilic molecules, but also controls the fate of these ligands by modulating their stability and oxidation status. Of particular interest is the binding of ApoD to arachidonic acid and its derivatives, which play a central role in healthy brain function. ApoD has been shown to act as a catalyst in the reduction of peroxidized eicosanoids and to attenuate lipid peroxidation in the brain. Manipulating its expression level in fruit flies and mice has demonstrated that ApoD has a favorable effect on both stress resistance and life span. The APOD gene is the gene that is upregulated the most in the aging human brain. Furthermore, ApoD levels in the nervous system are elevated in a large number of neurologic disorders including Alzheimer's disease, schizophrenia, and stroke. There is increasing evidence for a prominent neuroprotective role of ApoD because of its antioxidant and anti-inflammatory activity. ApoD emerges as an evolutionarily conserved anti-stress protein that is induced by oxidative stress and inflammation and may prove to be an effective therapeutic agent against a variety of neuropathologies, and even against aging.


Subject(s)
Aging/genetics , Apolipoproteins D/physiology , Neurodegenerative Diseases/genetics , Animals , Anti-Inflammatory Agents , Antioxidants , Apolipoproteins D/genetics , Apolipoproteins D/pharmacology , Apolipoproteins D/therapeutic use , Brain/metabolism , Catalysis , Eicosanoids/metabolism , Humans , Lipid Peroxidation/drug effects , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents , Oxidative Stress
4.
J Neurochem ; 117(6): 949-60, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21463325

ABSTRACT

The lipocalin Apolipoprotein D (ApoD), known to protect the nervous system against oxidative stress (OS) in model organisms, is up-regulated early in the mouse brain in response to the ROS generator paraquat. However, the processes triggered by this up-regulation have not been explored. We present here a study of the effect of ApoD on the early transcriptional changes upon OS in the mouse cerebellum using microarray profiling. ApoD-KO and transgenic mice over-expressing ApoD in neurons are compared to wild-type controls. In control conditions, ApoD affects the transcriptional profile of neuron and oligodendrocyte-specific genes involved in neuronal excitability, synaptic function, and myelin homeostasis. When challenged with paraquat, the absence of ApoD modifies the response of genes mainly related to OS management and myelination. Interestingly, the over-expression of ApoD in neurons almost completely abolishes the early transcriptional response to OS. We independently evaluate the expression of protein kinase Cδ, a gene up-regulated by OS only in the ApoD-KO cerebellum, and find it over-expressed in cultured ApoD-KO primary astrocytes, which points to a role for ApoD in astrocyte-microglia signaling. Our results support the hypothesis that ApoD is necessary for a proper response of the nervous system against physiological and pathological OS.


Subject(s)
Apolipoproteins D/physiology , Cerebellum/metabolism , Oxidative Stress , Transcription, Genetic , Animals , Apolipoproteins D/biosynthesis , Apolipoproteins D/genetics , Astrocytes/metabolism , Cells, Cultured , Gene Expression , Gene Expression Profiling , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Neurons/metabolism , Oligodendroglia/metabolism , Oligonucleotide Array Sequence Analysis
5.
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
7.
Neuroscience ; 162(2): 282-91, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19414061

ABSTRACT

Co-cultures of 3T3-L1 adipocytes with neurons from the rat dorsal root ganglia (DRG) showed enhanced neuritogenesis and synaptogenesis. Microarray analysis for upregulated genes in adipocyte/DRG co-cultures currently points to apolipoproteins D and E (ApoD, ApoE) as influential proteins. We therefore tested adipocyte-secreted cholesterol and the carrier proteins ApoD and ApoE3. Cholesterol, ApoD, and ApoE3 each increased neurite outgrowth and upregulated the expression of presynaptic synaptophysin and synaptotagmin, as well as the postsynaptic density protein 95. The neurotrophic effects of ApoD and ApoE3 were associated with an increased expression of the low-density lipoprotein receptor and apolipoprotein E receptor 2. Simultaneous treatment with receptor-associated protein, an apolipoprotein receptor antagonist, inhibited the neurotrophic function of both apolipoproteins. The application of ApoD, ApoE3, and cholesterol to DRG cell cultures corresponded with increased expression of the chemokine stromal cell-derived factor 1 and its receptor CXC chemokine receptor 4 (CXCR4). Surprisingly, the inhibition of CXCR4 by the antagonistic drug AMD3100 decreased the apolipoprotein/cholesterol dependent neurotrophic effects. We thus assume that apolipoprotein-induced neuritogenesis in DRG cells interferes with CXCR4 signaling, and that adipocyte-derived apolipoproteins might be helpful in nerve repair.


Subject(s)
Apolipoprotein E3/physiology , Apolipoproteins D/physiology , Ganglia, Spinal/cytology , Neurons/physiology , Synapses/physiology , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipokines/biosynthesis , Animals , Apolipoprotein E3/pharmacology , Apolipoproteins D/pharmacology , Benzylamines , Cells, Cultured , Chemokine CXCL12/biosynthesis , Cholesterol/pharmacology , Cholesterol/physiology , Coculture Techniques , Cyclams , Disks Large Homolog 4 Protein , Heterocyclic Compounds/pharmacology , Intracellular Signaling Peptides and Proteins , LDL-Receptor Related Protein-Associated Protein/pharmacology , Membrane Proteins/biosynthesis , Neurites/physiology , Neurons/drug effects , Rats , Rats, Inbred WF , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/biosynthesis , Receptors, Lipoprotein/antagonists & inhibitors , Receptors, Lipoprotein/metabolism , Synapses/drug effects , Synaptophysin/biosynthesis , Synaptotagmins/biosynthesis , Up-Regulation
8.
J Cereb Blood Flow Metab ; 28(3): 551-62, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17851453

ABSTRACT

Injury to the brain (e.g., stroke) results in a disruption of neuronal connectivity and loss of fundamental sensori-motor functions. The subsequent recovery of certain functions involves structural rearrangements in areas adjacent to the infarct. This remodeling of the injured brain requires trafficking of macromolecular components including cholesterol and phospholipids, a transport carried out by apolipoproteins including apolipoprotein D (apoD). We investigated the changes in the levels of apoD mRNA and protein, and its cellular localization during a recovery period up to 30 days after experimental stroke in the rat brain. In the core of the brain infarct, apoD immunoreactivity but not mRNA increased in dying pyramidal neurons, indicative of cellular redistribution of lipids. During 2 to 7 days of recovery after stroke, the apoD levels increased in the peri-infarct and white matter areas in cells identified as mature oligodendrocytes. The apoD expressing cells were conspicuously located along the rim of the infarct, suggesting a role for apoD in tissue repair. Furthermore, housing animals in an enriched environment improved sensori-motor function and increased the apoD levels. Our data strongly suggest that apoD is involved in regenerative processes and scar formation in the peri-infarct area presumably by enhancing lipid trafficking.


Subject(s)
Apolipoproteins D/physiology , Oligodendroglia/metabolism , Regeneration , Stroke/pathology , Animals , Apolipoproteins D/analysis , Apolipoproteins D/genetics , Biological Transport , Lipid Metabolism , Oligodendroglia/physiology , RNA, Messenger/analysis , Rats
9.
Schizophr Res ; 89(1-3): 147-53, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17011169

ABSTRACT

Apolipoprotein D (apoD), a member of the lipocalin superfamily of lipid-binding proteins, exhibits abundant expression within the CNS of many species, including humans; however, its physiological role remains unclear. Treatment with atypical antipsychotic drugs, especially clozapine, results in elevation of apoD expression levels in rodent brain and in human plasma samples. In order to further explore the role of apoD in mechanisms of clozapine function, we have measured a panel of membrane fatty acids and membrane lipids in brain from drug-treated apoD knock-out mice. Mice received clozapine (10 mg/kg/day) in their drinking water for 28 days and forebrain samples were analyzed using high performance liquid chromatography and capillary gas chromatography. We identified significant differences in the levels of membrane fatty acids in response to clozapine treatment specifically in the brains of apoD knock-out mice, but not wild-type (wt) mice. The most striking observations were decreases in the levels of fatty acids related to metabolism of arachidonic acid (AA), which is a known binding partner for apoD. These include the precursor to arachidonic acid, linoleic acid (LA; 18:2n6c), arachidonic acid itself (20:4n6) and the elongation product of arachidonic acid, adrenic acid (22:4n6). We further report increases in LA, eicosadienoic acid and docosahexaenoic acid in apoD knock-out compared to wild-type mice. These findings implicate an important apoD/AA interaction, which may be necessary for clozapine function.


Subject(s)
Antipsychotic Agents/pharmacology , Apolipoproteins D/physiology , Arachidonic Acid/metabolism , Clozapine/pharmacology , Animals , Apolipoproteins D/genetics , Chromatography, Gas , Chromatography, High Pressure Liquid , Erucic Acids/metabolism , Fatty Acids/metabolism , Fatty Acids, Unsaturated , Linoleic Acid/metabolism , Membrane Lipids/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Prosencephalon/drug effects , Prosencephalon/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...