Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Cell Neurosci ; 107: 103536, 2020 09.
Article in English | MEDLINE | ID: mdl-32777345

ABSTRACT

Peroxisomes exist in nearly every cell, oxidizing fats, synthesizing lipids and maintaining redox balance. As the brain ages, multiple pathways are negatively affected, but it is currently unknown if peroxisomal proteins are affected by aging in the brain. While recent studies have investigated a PEX5 homolog in aging C. elegans models and found that it is reduced in aging, it is unclear if PEX5, a mammalian peroxisomal protein that plays a role in peroxisomal homeostasis and degradation, is affected in the aging brain. To answer this question, we first determined the amount of PEX5, in brain homogenates from young (3 months) and aged (26 through 32+ months of age) wild-type mice of both sexes. PEX5 protein was decreased in aged male brains, but this reduction was not significant in female brains. RNAScope and real-time qPCR analyses showed that Pex5 mRNA was also reduced in aged male brain cortices, but not in females. Immunohistochemistry assays of cortical neurons in young and aged male brains showed that the amount of neuronal PEX5 was reduced in aged male brains. Cortical neurons in aged female mice also had reduced PEX5 levels in comparison to younger female mice. In conclusion, total PEX5 levels and Pex5 gene expression both decrease with age in male brains, and neuronal PEX5 levels lower in an age-dependent manner in the cortices of animals of both sexes.


Subject(s)
Aging/physiology , Brain/metabolism , Neurons/metabolism , Peroxisome-Targeting Signal 1 Receptor/metabolism , Animals , Cytosol/metabolism , Female , Male , Mice , Peroxisomes/genetics , Protein Transport/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Ubiquitination
2.
Front Cell Neurosci ; 14: 44, 2020.
Article in English | MEDLINE | ID: mdl-32210766

ABSTRACT

Peroxisomes exist in most cells, where they participate in lipid metabolism, as well as scavenging the reactive oxygen species (ROS) that are produced as by-products of their metabolic functions. In certain tissues such as the liver and kidneys, peroxisomes have more specific roles, such as bile acid synthesis in the liver and steroidogenesis in the adrenal glands. In the brain, peroxisomes are critically involved in creating and maintaining the lipid content of cell membranes and the myelin sheath, highlighting their importance in the central nervous system (CNS). This review summarizes the peroxisomal lifecycle, then examines the literature that establishes a link between peroxisomal dysfunction, cellular aging, and age-related disorders that affect the CNS. This review also discusses the gap of knowledge in research on peroxisomes in the CNS.

3.
Acta Neuropathol Commun ; 8(1): 36, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32197663

ABSTRACT

Neurodegenerative disorders, including chemotherapy-induced cognitive impairment, are associated with neuronal mitochondrial dysfunction. Cisplatin, a commonly used chemotherapeutic, induces neuronal mitochondrial dysfunction in vivo and in vitro. Astrocytes are key players in supporting neuronal development, synaptogenesis, axonal growth, metabolism and, potentially mitochondrial health. We tested the hypothesis that astrocytes transfer healthy mitochondria to neurons after cisplatin treatment to restore neuronal health.We used an in vitro system in which astrocytes containing mito-mCherry-labeled mitochondria were co-cultured with primary cortical neurons damaged by cisplatin. Culture of primary cortical neurons with cisplatin reduced neuronal survival and depolarized neuronal mitochondrial membrane potential. Cisplatin induced abnormalities in neuronal calcium dynamics that were characterized by increased resting calcium levels, reduced calcium responses to stimulation with KCl, and slower calcium clearance. The same dose of cisplatin that caused neuronal damage did not affect astrocyte survival or astrocytic mitochondrial respiration. Co-culture of cisplatin-treated neurons with astrocytes increased neuronal survival, restored neuronal mitochondrial membrane potential, and normalized neuronal calcium dynamics especially in neurons that had received mitochondria from astrocytes which underlines the importance of mitochondrial transfer. These beneficial effects of astrocytes were associated with transfer of mitochondria from astrocytes to cisplatin-treated neurons. We show that siRNA-mediated knockdown of the Rho-GTPase Miro-1 in astrocytes reduced mitochondrial transfer from astrocytes to neurons and prevented the normalization of neuronal calcium dynamics.In conclusion, we showed that transfer of mitochondria from astrocytes to neurons rescues neurons from the damage induced by cisplatin treatment. Astrocytes are far more resistant to cisplatin than cortical neurons. We propose that transfer of functional mitochondria from astrocytes to neurons is an important repair mechanism to protect the vulnerable cortical neurons against the toxic effects of cisplatin.


Subject(s)
Antineoplastic Agents/toxicity , Astrocytes/drug effects , Calcium/metabolism , Cisplatin/toxicity , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Neurons/drug effects , Animals , Astrocytes/metabolism , Astrocytes/physiology , Calcium Signaling , Cell Respiration/drug effects , Chemotherapy-Related Cognitive Impairment/etiology , Chemotherapy-Related Cognitive Impairment/metabolism , Coculture Techniques , Gene Knockdown Techniques , In Vitro Techniques , Luminescent Agents , Luminescent Proteins , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Turnover/drug effects , Neurons/metabolism , Neurotoxicity Syndromes , Optical Imaging , Oxygen Consumption/drug effects , Primary Cell Culture , Rats , rho GTP-Binding Proteins/genetics , Red Fluorescent Protein
4.
Cell Death Dis ; 9(5): 521, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29743513

ABSTRACT

Autophagy is a degradative pathway for removing aggregated proteins, damaged organelles, and parasites. Evidence indicates that autophagic pathways differ between cell types. In neurons, autophagy plays a homeostatic role, compared to a survival mechanism employed by starving non-neuronal cells. We investigated if sphingosine kinase 1 (SK1)-associated autophagy differs between two symbiotic brain cell types-neurons and astrocytes. SK1 synthesizes sphingosine-1-phosphate, which regulates autophagy in non-neuronal cells and in neurons. We found that benzoxazine autophagy inducers upregulate SK1 and neuroprotective autophagy in neurons, but not in astrocytes. Starvation enhances SK1-associated autophagy in astrocytes, but not in neurons. In astrocytes, SK1 is cytoprotective and promotes the degradation of an autophagy substrate, mutant huntingtin, the protein that causes Huntington's disease. Overexpressed SK1 is unexpectedly toxic to neurons, and its toxicity localizes to the neuronal soma, demonstrating an intricate relationship between the localization of SK1's activity and neurotoxicity. Our results underscore the importance of cell type-specific autophagic differences in any efforts to target autophagy therapeutically.


Subject(s)
Astrocytes/enzymology , Autophagy , Neurons/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Astrocytes/cytology , Neurons/cytology , Organ Specificity , Phosphotransferases (Alcohol Group Acceptor)/genetics , Rats
5.
Mol Cell Neurosci ; 86: 65-71, 2018 01.
Article in English | MEDLINE | ID: mdl-29180229

ABSTRACT

Doxorubicin, a commonly used anti-neoplastic agent, causes severe neurotoxicity. Doxorubicin promotes thinning of the brain cortex and accelerates brain aging, leading to cognitive impairment. Oxidative stress induced by doxorubicin contributes to cellular damage. In addition to mitochondria, peroxisomes also generate reactive oxygen species (ROS) and promote cell senescence. Here, we investigated if doxorubicin affects peroxisomal homeostasis in neurons. We demonstrate that the number of peroxisomes is increased in doxorubicin-treated neurons and in the brains of mice which underwent doxorubicin-based chemotherapy. Pexophagy, the specific autophagy of peroxisomes, is downregulated in neurons, and peroxisomes produce more ROS. 2-hydroxypropyl-ß-cyclodextrin (HPßCD), an activator of the transcription factor TFEB, which regulates expression of genes involved in autophagy and lysosome function, mitigates damage of pexophagy and decreases ROS production induced by doxorubicin. We conclude that peroxisome-associated oxidative stress induced by doxorubicin may contribute to neurotoxicity, cognitive dysfunction, and accelerated brain aging in cancer patients and survivors. Peroxisomes might be a valuable new target for mitigating neuronal damage caused by chemotherapy drugs and for slowing down brain aging in general.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Neurons/drug effects , Oxidative Stress/drug effects , Peroxisomes/drug effects , Animals , Cells, Cultured , Female , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Frontal Lobe/ultrastructure , Mice , Neurons/metabolism , Neurons/ultrastructure , Oxidative Stress/physiology , Peroxisomes/metabolism , Peroxisomes/ultrastructure , Rats , Reactive Oxygen Species/metabolism
6.
Hum Mol Genet ; 26(7): 1305-1317, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28175299

ABSTRACT

Huntington disease (HD) is the most common inherited neurodegenerative disorder. It has no cure. The protein huntingtin causes HD, and mutations to it confer toxic functions to the protein that lead to neurodegeneration. Thus, identifying modifiers of mutant huntingtin-mediated neurotoxicity might be a therapeutic strategy for HD. Sphingosine kinases 1 (SK1) and 2 (SK2) synthesize sphingosine-1-phosphate (S1P), a bioactive lipid messenger critically involved in many vital cellular processes, such as cell survival. In the nucleus, SK2 binds to and inhibits histone deacetylases 1 and 2 (HDAC1/2). Inhibiting both HDACs has been suggested as a potential therapy in HD. Here, we found that SK2 is nuclear in primary neurons and, unexpectedly, overexpressed SK2 is neurotoxic in a dose-dependent manner. SK2 promotes DNA double-strand breaks in cultured primary neurons. We also found that SK2 is hyperphosphorylated in the brain samples from a model of HD, the BACHD mice. These data suggest that the SK2 pathway may be a part of a pathogenic pathway in HD. ABC294640, an inhibitor of SK2, reduces DNA damage in neurons and increases survival in two neuron models of HD. Our results identify a novel regulator of mutant huntingtin-mediated neurotoxicity and provide a new target for developing therapies for HD.


Subject(s)
Cell Nucleus/metabolism , Huntingtin Protein/genetics , Huntington Disease/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Cell Nucleus/genetics , Disease Models, Animal , Gene Expression Regulation , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , Humans , Huntington Disease/metabolism , Huntington Disease/pathology , Lysophospholipids/metabolism , Mice , Neurons/metabolism , Neurons/pathology , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Sphingosine/analogs & derivatives , Sphingosine/metabolism
7.
Aging (Albany NY) ; 8(12): 3507-3519, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27992857

ABSTRACT

Doxorubicin, a commonly used chemotherapy agent, induces severe cardio- and neurotoxicity. Molecular mechanisms of cardiotoxicity have been extensively studied, but mechanisms by which doxorubicin exhibits its neurotoxic properties remain unclear. Here, we show that doxorubicin impairs neuronal autophagy, leading to the accumulation of an autophagy substrate p62. Neurons treated with doxorubicin contained autophagosomes, damaged mitochondria, and lipid droplets. The brains from mice treated with pegylated liposomal doxorubicin exhibited autophagosomes, often with mitochondria, lipofuscin, and lipid droplets. Interestingly, lysosomes were less acidic in doxorubicin-treated neurons. Overexpression of the transcription factor EB (TFEB), which controls the autophagy-lysosome axis, increased survival of doxorubicin-treated neurons. 2-Hydroxypropyl-ß-cyclodextrin (HPßCD), an activator of TFEB, also promoted neuronal survival, decreased the levels of p62, and lowered the pH in lysosomes. Taken together, substantial changes induced by doxorubicin contribute to neurotoxicity, cognitive disturbances in cancer patients and survivors, and accelerated brain aging. The TFEB pathway might be a new approach for mitigating damage of neuronal autophagy caused by doxorubicin.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Doxorubicin/pharmacology , Lysosomes/physiology , Neurons/physiology , Animals , Autophagosomes/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Lipid Droplets/physiology , Mice , Mice, Nude , Rats , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Topoisomerase II Inhibitors/pharmacology
8.
Autophagy ; 12(8): 1418-24, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27467777

ABSTRACT

Although implicated in neurodegeneration, autophagy has been characterized mostly in yeast and mammalian non-neuronal cells. In a recent study, we sought to determine if SPHK1 (sphingosine kinase 1), implicated previously in macroautophagy/autophagy in cancer cells, regulates autophagy in neurons. SPHK1 synthesizes sphingosine-1-phosphate (S1P), a bioactive lipid involved in cell survival. In our study, we discovered that, when neuronal autophagy is pharmacologically stimulated, SPHK1 relocalizes to the endocytic and autophagic organelles. Interestingly, in non-neuronal cells stimulated with growth factors, SPHK1 translocates to the plasma membrane, where it phosphorylates sphingosine to produce S1P. Whether SPHK1 also binds to the endocytic and autophagic organelles in non-neuronal cells upon induction of autophagy has not been demonstrated. Here, we determined if the effect in neurons is operant in the SH-SY5Y neuroblastoma cell line. In both non-differentiated and differentiated SH-SY5Y cells, a short incubation of cells in amino acid-free medium stimulated the formation of SPHK1-positive puncta, as in neurons. We also found that, unlike neurons in which these puncta represent endosomes, autophagosomes, and amphisomes, in SH-SY5Y cells SPHK1 is bound only to the endosomes. In addition, a dominant negative form of SPHK1 was very toxic to SH-SY5Y cells, but cultured primary cortical neurons tolerated it significantly better. These results suggest that autophagy in neurons is regulated by mechanisms that differ, at least in part, from those in SH-SY5Y cells.


Subject(s)
Autophagy , Neuroblastoma/metabolism , Neurons/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Apoptosis/physiology , Autophagosomes/metabolism , Cell Differentiation , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival , Cells, Cultured , Endocytosis , Endosomes/metabolism , Green Fluorescent Proteins/metabolism , Humans , Light , Lipids/chemistry , Lysophospholipids/metabolism , Lysosomes/metabolism , Phagosomes/metabolism , Phosphorylation , Rats , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism
9.
Sci Rep ; 6: 25705, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27168474

ABSTRACT

Neurotoxicity may occur in cancer patients and survivors during or after chemotherapy. Cognitive deficits associated with neurotoxicity can be subtle or disabling and frequently include disturbances in memory, attention, executive function and processing speed. Searching for pathways altered by anti-cancer treatments in cultured primary neurons, we discovered that doxorubicin, a commonly used anti-neoplastic drug, significantly decreased neuronal survival. The drug promoted the formation of DNA double-strand breaks in primary neurons and reduced synaptic and neurite density. Pretreatment of neurons with levetiracetam, an FDA-approved anti-epileptic drug, enhanced survival of chemotherapy drug-treated neurons, reduced doxorubicin-induced formation of DNA double-strand breaks, and mitigated synaptic and neurite loss. Thus, levetiracetam might be part of a valuable new approach for mitigating synaptic damage and, perhaps, for treating cognitive disturbances in cancer patients and survivors.


Subject(s)
DNA Damage , Doxorubicin/adverse effects , Neurons/pathology , Piracetam/analogs & derivatives , Synapses/pathology , Animals , BRCA1 Protein/metabolism , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/pathology , DNA Breaks, Double-Stranded , Down-Regulation/drug effects , Levetiracetam , Neurites/drug effects , Neurites/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Piracetam/pharmacology , Rats , Synapses/drug effects , Synapses/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
10.
Sci Rep ; 5: 15213, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26477494

ABSTRACT

Autophagy is an important homeostatic mechanism that eliminates long-lived proteins, protein aggregates and damaged organelles. Its dysregulation is involved in many neurodegenerative disorders. Autophagy is therefore a promising target for blunting neurodegeneration. We searched for novel autophagic pathways in primary neurons and identified the cytosolic sphingosine-1-phosphate (S1P) pathway as a regulator of neuronal autophagy. S1P, a bioactive lipid generated by sphingosine kinase 1 (SK1) in the cytoplasm, is implicated in cell survival. We found that SK1 enhances flux through autophagy and that S1P-metabolizing enzymes decrease this flux. When autophagy is stimulated, SK1 relocalizes to endosomes/autophagosomes in neurons. Expression of a dominant-negative form of SK1 inhibits autophagosome synthesis. In a neuron model of Huntington's disease, pharmacologically inhibiting S1P-lyase protected neurons from mutant huntingtin-induced neurotoxicity. These results identify the S1P pathway as a novel regulator of neuronal autophagy and provide a new target for developing therapies for neurodegenerative disorders.


Subject(s)
Autophagy , Lysophospholipids/metabolism , Neurons/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Aldehyde-Lyases/antagonists & inhibitors , Aldehyde-Lyases/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Biomarkers , Cell Survival/drug effects , Cytoplasm , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression , Phagosomes/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Protein Transport , Rats , Sphingosine/metabolism
11.
Horm Behav ; 60(2): 226-32, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21621542

ABSTRACT

When ovariectomized Fischer female rats are hormonally primed with 10 µg estradiol benzoate, a 5 min restraint experience rapidly inhibits lordosis behavior. Addition of progesterone to the hormonal priming prevents this restraint-induced inhibition. In prior work, we reported evidence that progesterone receptors (PR) may contribute to this protective effect of progesterone. In the current manuscript, we provide evidence that progesterone metabolites may also contribute to progesterone's ability to reduce the effects of restraint. Ovariectomized female rats were hormonally primed with 10 µg estradiol benzoate followed 2 days later with 4.0 mg/kg of the progesterone metabolite, allopregnanolone. Allopregnanolone, administered either 4 h or 2 h before the restraint experience, was as effective as progesterone in reducing the lordosis-inhibitory effects of restraint. In the second experiment, progesterone metabolism was blocked with 50 mg/kg of the 5α-reductase inhibitor, finasteride. Surprisingly, finasteride did not prevent progesterone from reducing the effects of restraint. In a third experiment, we tested the possibility that allopregnanolone acted through metabolism to dihydroprogesterone. Rats were treated with allopregnanolone or with allopregnanolone plus the 3α-hydroxysteroid dehydrogenase inhibitor, indomethacin. Indomethacin did not prevent allopregnanolone from reducing the effects of restraint. Mechanisms are discussed whereby cross-talk between PR-mediated and metabolite-mediated events may converge in producing progesterone's attenuation of the effect of restraint.


Subject(s)
Posture/physiology , Pregnanolone/pharmacology , Progestins/pharmacology , Receptors, Progesterone/metabolism , Sexual Behavior, Animal/physiology , 5-alpha Reductase Inhibitors/pharmacology , Animals , Cyclooxygenase Inhibitors/pharmacology , Female , Finasteride/pharmacology , Indomethacin/pharmacology , Ovariectomy , Rats , Rats, Inbred F344 , Restraint, Physical , Sexual Behavior, Animal/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...