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










Publication year range
1.
J Pineal Res ; 75(4): e12909, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37721126

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative brain disorder associated with uncontrolled body movements, cognitive decline, and reduced circulating melatonin levels. Melatonin is a potent antioxidant and exogenous melatonin treatment is neuroprotective in experimental HD models. In neurons, melatonin is exclusively synthesized in the mitochondrial matrix. Thus, we investigated the integrity of melatonin biosynthesis pathways in pineal and extrapineal brain areas in human HD brain samples, in the R6/2 mouse model of HD and in full-length mutant huntingtin knock-in cells. Aralkylamine N-acetyltransferase (AANAT) is the rate-limiting step enzyme in the melatonin biosynthetic pathway. We found that AANAT expression is significantly decreased in the pineal gland and the striatum of HD patients compared to normal controls. In the R6/2 mouse forebrain, AANAT protein expression was decreased in synaptosomal, but not nonsynaptosomal, mitochondria and was associated with decreased synaptosomal melatonin levels compared to wild type mice. We also demonstrate sequestration of AANAT in mutant-huntingtin protein aggregates likely resulting in decreased AANAT bioavailability. Paradoxically, AANAT mRNA expression is increased in tissues where AANAT protein expression is decreased, suggesting a potential feedback loop that is, ultimately unsuccessful. In conclusion, we demonstrate that pineal, extrapineal, and synaptosomal melatonin levels are compromised in the brains of HD patients and R6/2 mice due, at least in part, to protein aggregation.


Subject(s)
Huntington Disease , Melatonin , Pineal Gland , Humans , Mice , Animals , Melatonin/metabolism , Pineal Gland/metabolism
2.
Proc Natl Acad Sci U S A ; 116(33): 16593-16602, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31346086

ABSTRACT

Mutant huntingtin (mHTT), the causative protein in Huntington's disease (HD), associates with the translocase of mitochondrial inner membrane 23 (TIM23) complex, resulting in inhibition of synaptic mitochondrial protein import first detected in presymptomatic HD mice. The early timing of this event suggests that it is a relevant and direct pathophysiologic consequence of mHTT expression. We show that, of the 4 TIM23 complex proteins, mHTT specifically binds to the TIM23 subunit and that full-length wild-type huntingtin (wtHTT) and mHTT reside in the mitochondrial intermembrane space. We investigated differences in mitochondrial proteome between wtHTT and mHTT cells and found numerous proteomic disparities between mHTT and wtHTT mitochondria. We validated these data by quantitative immunoblotting in striatal cell lines and human HD brain tissue. The level of soluble matrix mitochondrial proteins imported through the TIM23 complex is lower in mHTT-expressing cell lines and brain tissues of HD patients compared with controls. In mHTT-expressing cell lines, membrane-bound TIM23-imported proteins have lower intramitochondrial levels, whereas inner membrane multispan proteins that are imported via the TIM22 pathway and proteins integrated into the outer membrane generally remain unchanged. In summary, we show that, in mitochondria, huntingtin is located in the intermembrane space, that mHTT binds with high-affinity to TIM23, and that mitochondria from mHTT-expressing cells and brain tissues of HD patients have reduced levels of nuclearly encoded proteins imported through TIM23. These data demonstrate the mechanism and biological significance of mHTT-mediated inhibition of mitochondrial protein import, a mechanism likely broadly relevant to other neurodegenerative diseases.


Subject(s)
Huntingtin Protein/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mutant Proteins/metabolism , Proteostasis , Cell Line , Cell Nucleus/metabolism , Cerebral Cortex/pathology , Corpus Striatum/pathology , Humans , Huntington Disease , Mitochondrial Membranes/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/metabolism , Protein Binding , Proteome/metabolism
3.
Proc Natl Acad Sci U S A ; 116(2): 650-659, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30584104

ABSTRACT

Neuritic retraction in the absence of overt neuronal death is a shared feature of normal aging and neurodegenerative disorders, but the intracellular mechanisms modulating this process are not understood. We propose that cumulative distal mitochondrial protein damage results in impaired protein import, leading to mitochondrial dysfunction and focal activation of the canonical apoptosis pathway in neurites. This is a controlled process that may not lead to neuronal death and, thus, we term this phenomenon "neuritosis." Consistent with our hypothesis, we show that in primary cerebrocortical neurons, mitochondrial distance from the soma correlates with increased mitochondrial protein damage, PINK1 accumulation, reactive oxygen species production, and decreased mitochondrial membrane potential and depolarization threshold. Furthermore, we demonstrate that the distance-dependent mitochondrial membrane potential gradient exists in vivo in mice. We demonstrate that impaired distal mitochondria have a lower threshold for focal/nonlethal neuritic caspase-3 activation in normal neurons that is exacerbated in aging, stress, and neurodegenerative conditions, thus delineating a fundamental mechanistic underpinning for synaptic vulnerability.


Subject(s)
Apoptosis , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Neurites/metabolism , Neurodegenerative Diseases/metabolism , Animals , Caspase 3/genetics , Caspase 3/metabolism , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , Neurites/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Protein Kinases/genetics , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism
4.
Proc Natl Acad Sci U S A ; 114(38): E7997-E8006, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28874589

ABSTRACT

G protein-coupled receptors (GPCRs) are classically characterized as cell-surface receptors transmitting extracellular signals into cells. Here we show that central components of a GPCR signaling system comprised of the melatonin type 1 receptor (MT1), its associated G protein, and ß-arrestins are on and within neuronal mitochondria. We discovered that the ligand melatonin is exclusively synthesized in the mitochondrial matrix and released by the organelle activating the mitochondrial MT1 signal-transduction pathway inhibiting stress-mediated cytochrome c release and caspase activation. These findings coupled with our observation that mitochondrial MT1 overexpression reduces ischemic brain injury in mice delineate a mitochondrial GPCR mechanism contributing to the neuroprotective action of melatonin. We propose a new term, "automitocrine," analogous to "autocrine" when a similar phenomenon occurs at the cellular level, to describe this unexpected intracellular organelle ligand-receptor pathway that opens a new research avenue investigating mitochondrial GPCR biology.


Subject(s)
Brain Injuries/metabolism , Brain Ischemia/metabolism , Melatonin/biosynthesis , Mitochondria/metabolism , Receptor, Melatonin, MT1/metabolism , Signal Transduction , Animals , Brain Injuries/genetics , Brain Ischemia/genetics , Cytochromes c/genetics , Cytochromes c/metabolism , Male , Melatonin/genetics , Mice , Mitochondria/genetics , Receptor, Melatonin, MT1/genetics
5.
J Neurosci Methods ; 263: 1-6, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26808294

ABSTRACT

BACKGROUND: Functional and structural properties of mitochondria are highly tissue and cell dependent, but isolation of highly purified human neuronal mitochondria is not currently available. NEW METHOD: We developed and validated a procedure to isolate purified neuronal mitochondria from brain tissue. The method combines Percoll gradient centrifugation to obtain synaptosomal fraction with nitrogen cavitation mediated synaptosome disruption and extraction of mitochondria using anti mitochondrial outer membrane protein antibodies conjugated to magnetic beads. The final products of isolation are non-synaptosomal mitochondria, which are a mixture of mitochondria isolated from different brain cells (i.e. neurons, astrocytes, oligodendrocytes, microglia) and synaptic mitochondria, which are of neuronal origin. This method is well suited for preparing functional mitochondria from human cortex tissue that is surgically extracted. RESULTS: The procedure produces mitochondria with minimal cytoplasmic contaminations that are functionally active based on measurements of mitochondrial respiration as well as mitochondrial protein import. The procedure requires approximately four hours for the isolation of human neuronal mitochondria and can also be used to isolate mitochondria from mouse/rat/monkey brains. COMPARISON WITH EXISTING METHODS AND CONCLUSIONS: This method will allow researchers to study highly enriched neuronal mitochondria without the confounding effect of cellular and organelle contaminants.


Subject(s)
Cerebral Cortex/cytology , Mitochondria/physiology , Neurons/ultrastructure , Antibodies/metabolism , Cell Fractionation , HLA Antigens/metabolism , Humans , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/immunology , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/metabolism , Synaptosomes/metabolism , Synaptosomes/ultrastructure
6.
Nat Neurosci ; 17(6): 822-31, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24836077

ABSTRACT

Mitochondrial dysfunction is associated with neuronal loss in Huntington's disease (HD), a neurodegenerative disease caused by an abnormal polyglutamine expansion in huntingtin (Htt). However, the mechanisms linking mutant Htt and mitochondrial dysfunction in HD remain unknown. We identify an interaction between mutant Htt and the TIM23 mitochondrial protein import complex. Remarkably, recombinant mutant Htt directly inhibited mitochondrial protein import in vitro. Furthermore, mitochondria from brain synaptosomes of presymptomatic HD model mice and from mutant Htt-expressing primary neurons exhibited a protein import defect, suggesting that deficient protein import is an early event in HD. The mutant Htt-induced mitochondrial import defect and subsequent neuronal death were attenuated by overexpression of TIM23 complex subunits, demonstrating that deficient mitochondrial protein import causes mutant Htt-induced neuronal death. Collectively, these findings provide evidence for a direct link between mutant Htt, mitochondrial dysfunction and neuronal pathology, with implications for mitochondrial protein import-based therapies in HD.


Subject(s)
Huntington Disease/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Nerve Tissue Proteins/genetics , Aged , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Huntingtin Protein , Huntington Disease/pathology , Huntington Disease/therapy , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Middle Aged , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mutation , Nerve Tissue Proteins/physiology , Protein Transport/genetics
7.
Database (Oxford) ; 2010: baq013, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20624719

ABSTRACT

The human oral microbiome is the most studied human microflora, but 53% of the species have not yet been validly named and 35% remain uncultivated. The uncultivated taxa are known primarily from 16S rRNA sequence information. Sequence information tied solely to obscure isolate or clone numbers, and usually lacking accurate phylogenetic placement, is a major impediment to working with human oral microbiome data. The goal of creating the Human Oral Microbiome Database (HOMD) is to provide the scientific community with a body site-specific comprehensive database for the more than 600 prokaryote species that are present in the human oral cavity based on a curated 16S rRNA gene-based provisional naming scheme. Currently, two primary types of information are provided in HOMD--taxonomic and genomic. Named oral species and taxa identified from 16S rRNA gene sequence analysis of oral isolates and cloning studies were placed into defined 16S rRNA phylotypes and each given unique Human Oral Taxon (HOT) number. The HOT interlinks phenotypic, phylogenetic, genomic, clinical and bibliographic information for each taxon. A BLAST search tool is provided to match user 16S rRNA gene sequences to a curated, full length, 16S rRNA gene reference data set. For genomic analysis, HOMD provides comprehensive set of analysis tools and maintains frequently updated annotations for all the human oral microbial genomes that have been sequenced and publicly released. Oral bacterial genome sequences, determined as part of the Human Microbiome Project, are being added to the HOMD as they become available. We provide HOMD as a conceptual model for the presentation of microbiome data for other human body sites. Database URL: http://www.homd.org.


Subject(s)
Databases, Genetic , Metagenome/genetics , Mouth/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Computers , Genome, Bacterial , Humans , Internet , Metagenomics , Phenotype , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Sequence Alignment , Software
8.
J Neurochem ; 102(3): 991-1000, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17630988

ABSTRACT

One of the hypotheses for the development of familial amyotrophic lateral sclerosis (ALS) is that mutations in the superoxide dismutase 1 enzyme lead to aberrant properties of the copper within the active site of the enzyme which then causes increased oxidative damage. The lipophilic metal chelators DP-109 and DP-460 which chelate calcium, copper, and zinc were tested in the G93A-transgenic ALS mouse model. Both compounds significantly extended survival, DP-109 (5 mg/kg/day) by 10%, DP-460 (10 mg/kg/day) by 9%. While the effect on survival was relatively small, chelator treatment also improved motor performance, dramatically reduced cell loss in the lumbar spinal cord and decreased reactive astrocytosis and microgliosis. Markers of oxidative damage, tumor necrosis factor (TNF)-alpha and alpha-synuclein were reduced in the lumbar spinal cord of G93A mice treated with DP-109 or DP-460 as compared with vehicle-treated animals. Furthermore, the treatment induced protein expression of the transcription factor hypoxia inducible factor-1alpha and mRNA levels of vascular endothelial growth factor as a corresponding target gene. In line with previous studies using metal chelators in the G93A animal model, our results suggest that these compounds have neuroprotective capacities in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Central Nervous System/drug effects , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Metals/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Biomarkers/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Central Nervous System/metabolism , Central Nervous System/physiopathology , Chelating Agents/therapeutic use , Disease Models, Animal , Egtazic Acid/pharmacology , Egtazic Acid/therapeutic use , Female , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Membrane Lipids/metabolism , Metals/metabolism , Mice , Mice, Transgenic , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Neuroprotective Agents/therapeutic use , Oxidative Stress/physiology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Treatment Outcome , Vascular Endothelial Growth Factor A/genetics
9.
J Neurosci ; 27(23): 6320-32, 2007 Jun 06.
Article in English | MEDLINE | ID: mdl-17554006

ABSTRACT

In the present study, we show a biphasic activation of hypoxia inducible factor 1alpha (HIF-1) after stroke that lasts for up to 10 d, suggesting the involvement of the HIF pathway in several aspects of the pathophysiology of cerebral ischemia. We provide evidence that HIF-1-mediated responses have an overall beneficial role in the ischemic brain as indicated by increased tissue damage and reduced survival rate of mice with neuron-specific knockdown of HIF-1alpha that were subjected to transient focal cerebral ischemia. In addition, we demonstrated that drugs known to activate HIF-1 in cultured cells as well as in vivo had neuroprotective properties in this model of cerebral ischemia. This protective effect was significantly attenuated but not completely abolished in neuron-specific HIF-1alpha-deficient mice, suggesting that alternative mechanisms of neuroprotection are also implicated. Last, our study showed that hypoxia-induced tolerance to ischemia was preserved in neuron-specific HIF-1alpha-deficient mice, indicating that the neuroprotective effects of hypoxic preconditioning do not depend on neuronal HIF-1 activation.


Subject(s)
Brain Injuries/metabolism , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Ischemic Attack, Transient/metabolism , Neurons/metabolism , Animals , Brain Injuries/genetics , Cerebrovascular Circulation/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ischemic Attack, Transient/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains
10.
Neurobiol Dis ; 26(3): 671-80, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17481908

ABSTRACT

Recent advances in cancer cell biology have focused on histone deacetylase inhibitors (HDACi's) because they target pathways critical to the development and progression of disease. In particular, HDACi's can induce expression of epigenetically silenced genes that promote growth arrest, differentiation and cell death. In glioma cells, one such repressed gene is the tetraspanin CD81, which regulates cytostasis in various cell lines and in astrocytes, the major cellular component of gliomas. Our studies show that HDACi's, trichostatin and sodium butyrate, promote growth arrest and differentiation with negligible cell death in glioma cells and induce expression of CD81 and cyclin-dependent kinase inhibitor 1A (p21(CIP/WAF-1)), another regulator of cytostasis in astrocytes. Interference RNA knock-down of CD81 abrogates cytostasis promoted by HDAC inhibition indicating that HDACi-induced CD81 is responsible for growth arrest. Induction of CD81 expression through HDAC inhibition is a novel strategy to promote growth arrest in glioma cells.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/enzymology , Enzyme Inhibitors/pharmacology , Glioma/enzymology , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Animals , Antigens, CD/genetics , Brain Neoplasms/genetics , Brain Neoplasms/physiopathology , Butyrates/pharmacology , Butyrates/therapeutic use , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Down-Regulation/drug effects , Down-Regulation/physiology , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing/drug effects , Gene Silencing/physiology , Genes, cdc/drug effects , Glioma/genetics , Glioma/physiopathology , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , RNA Interference , Rats , Rats, Inbred F344 , Rats, Wistar , Tetraspanin 28
11.
J Neurosci ; 26(37): 9471-81, 2006 Sep 13.
Article in English | MEDLINE | ID: mdl-16971531

ABSTRACT

In the ischemic or hypoxic brain, astrocytes appear to be one of the main sources of erythropoietin (EPO). In this study, we investigated the differential contribution of hypoxia inducible factor (HIF) isoforms to the regulation of hypoxic EPO expression in cultured astrocytes. In addition, using an in vitro model of oxygen-glucose deprivation (OGD), we studied the role of HIF-1alpha and HIF-2alpha in the generation of paracrine protective signals by astrocytes that modulate the survival of neurons exposed to OGD. Expression of HIF-1alpha or HIF-2alpha was abrogated by infecting astrocytes with lentiviral particles encoding small interference RNA specific for HIF-1alpha or HIF-2alpha (siHIF-1alpha or siHIF-2alpha). Astrocytes infected with siHIF-1alpha showed abrogated hypoxic induction of vascular endothelial growth factor (VEGF) and lactate dehydrogenase (LDH) but normal EPO induction. In contrast, reduction of HIF-2alpha expression by siHIF-2alpha led to a drastic decrease of EPO hypoxic expression, but it did not affect LDH or VEGF upregulation. To further test whether HIF-2 is sufficient to drive EPO upregulation, we expressed oxygen-insensitive mutant forms of HIF-1alpha (mtHIF-1alpha) (P402A/P577A) and HIF-2alpha (mtHIF-2alpha) (P405A/P530A). Expression of mtHIF-2alpha but not mtHIF-1alpha in normoxic astrocytes resulted in a significant upregulation of EPO mRNA and protein. Accordingly, HIF-2alpha but not HIF-1alpha was found to be associated with the EPO hypoxia-response element by a chromatin immunoprecipitation assay. Interestingly, conditioned medium from astrocytes challenged by sublethal OGD improved neuronal survival to OGD; however, this effect was abolished during the downregulation of astrocytic HIF-2alpha using siHIF-2alpha. These results indicate that HIF-2alpha mediates the transcriptional activation of EPO expression in astrocytes, and this pathway may promote astrocytic paracrine-dependent neuronal survival during ischemia.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Erythropoietin/metabolism , Hypoxia-Ischemia, Brain/metabolism , Oxygen/metabolism , Transcription Factors/metabolism , Animals , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/physiopathology , Down-Regulation/physiology , Erythropoietin/genetics , Genetic Vectors/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Ischemia, Brain/physiopathology , L-Lactate Dehydrogenase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Paracrine Communication/physiology , Response Elements/genetics , Transcription Factors/genetics , Transcriptional Activation/physiology , Transfection , Up-Regulation/physiology , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...