Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mol Cell Neurosci ; 102: 103419, 2020 01.
Article in English | MEDLINE | ID: mdl-31770591

ABSTRACT

Friedreich's ataxia is a multisystemic genetic disorder within the family of mitochondrial diseases that is characterized by reduced levels of the essential mitochondrial protein frataxin. Based on clinical evidence, the peripheral nervous system is affected early, neuronal dysfunction progresses towards the central nervous system, and other organs (such as heart and pancreas) are affected later. However, little attention has been given to the specific aspects of mitochondria function altered by frataxin depletion in the nervous system. For years, commonly accepted views on mitochondria dysfunction in Friedreich's ataxia stemmed from studies using non-neuronal systems and may not apply to neurons, which have their own bioenergetic needs and present a unique, extensive neurite network. Moreover, the basis of the selective neuronal vulnerability, which primarily affects large sensory neurons in the dorsal root ganglia, large principal neurons in the dentate nuclei of the cerebellum, and pyramidal neurons in the cerebral cortex, remains elusive. In order to identify potential misbeliefs in the field and highlight controversies, we reviewed current knowledge on frataxin expression in different tissues, discussed the molecular function of frataxin, and the consequences of its deficiency for mitochondria structural and functional properties, with a focus on the nervous system.


Subject(s)
Friedreich Ataxia/metabolism , Mitochondria/metabolism , Neurons/metabolism , Animals , Friedreich Ataxia/genetics , Friedreich Ataxia/pathology , Humans , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Frataxin
2.
J Cell Sci ; 130(21): 3713-3727, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28864766

ABSTRACT

cAMP regulates a wide variety of physiological functions in mammals. This single second messenger can regulate multiple, seemingly disparate functions within independently regulated cell compartments. We have previously identified one such compartment inside the matrix of the mitochondria, where soluble adenylyl cyclase (sAC) regulates oxidative phosphorylation (OXPHOS). We now show that sAC knockout fibroblasts have a defect in OXPHOS activity and attempt to compensate for this defect by increasing OXPHOS proteins. Importantly, sAC knockout cells also exhibit decreased probability of endoplasmic reticulum (ER) Ca2+ release associated with diminished phosphorylation of the inositol 3-phosphate receptor. Restoring sAC expression exclusively in the mitochondrial matrix rescues OXPHOS activity and reduces mitochondrial biogenesis, indicating that these phenotypes are regulated by intramitochondrial sAC. In contrast, Ca2+ release from the ER is only rescued when sAC expression is restored throughout the cell. Thus, we show that functionally distinct, sAC-defined, intracellular cAMP signaling domains regulate metabolism and Ca2+ signaling.


Subject(s)
Adenylyl Cyclases/metabolism , Calcium Signaling , Calcium/metabolism , Cyclic AMP/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Adenylyl Cyclases/genetics , Animals , Cell Fractionation , Cell Line , Endoplasmic Reticulum/ultrastructure , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Gene Knockout Techniques , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mitochondria/ultrastructure , Oxidative Phosphorylation , Oxygen Consumption
3.
Hum Mol Genet ; 23(6): 1413-24, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24154542

ABSTRACT

Neuronal mitochondrial morphology abnormalities occur in models of familial amyotrophic lateral sclerosis (ALS) associated with SOD1 and TDP43 mutations. These abnormalities have been linked to mitochondrial axonal transport defects, but the temporal and spatial relationship between mitochondrial morphology and transport alterations in these two distinct genetic forms of ALS has not been investigated in vivo. To address this question, we crossed SOD1 (wild-type SOD1(WT) and mutant SOD1(G93A)) or TDP43 (mutant TDP43(A315T)) transgenic mice with mice expressing the fluorescent protein Dendra targeted to mitochondria in neurons (mitoDendra). At different time points during the disease course, we studied mitochondrial transport in the intact sciatic nerve of living mice and analyzed axonal mitochondrial morphology at multiple sites, spanning from the spinal cord to the motor terminals. Defects of retrograde mitochondrial transport were detected at 45 days of age, before the onset of symptoms, in SOD1(G93A) and TDP43(A315T) mice, but not in SOD1(WT). At later disease stages, also anterograde mitochondrial transport was affected in both mutant mouse lines. In SOD1(G93A) mice, mitochondrial morphological abnormalities were apparent at 15 days of age, thus preceding transport abnormalities. Conversely, in TDP43(A315T) mice, morphological abnormalities appeared after the onset of transport defects. Taken together, these findings demonstrate that neuronal mitochondrial transport and morphology abnormalities occur in vivo and that they are common denominators of different genetic forms of the ALS. At the same time, differences in the temporal and spatial manifestation of mitochondrial abnormalities between the two mouse models of familial ALS imply that different molecular mechanisms may be involved.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , DNA-Binding Proteins/genetics , Mitochondria/pathology , Neurons/pathology , Sciatic Nerve/physiopathology , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , DNA-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Neurons/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
4.
J Pharmacol Exp Ther ; 356(3): 645-55, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26718240

ABSTRACT

The extensive use of organophosphates (OPs) is an ongoing environmental health concern due to multiple reports of OP-related neurologic abnormalities. The mechanism of the acute toxicity of OPs has been attributed to inhibition of acetylcholinesterase (AChE), but there is growing evidence that this may not account for all the long-term neurotoxic effects of OPs. In previous experiments (using ex vivo and in vitro model systems) we observed that the insecticide OP chlorpyrifos impaired the movements of vesicles and mitochondria in axons. Here, using a time-lapse imaging technique, we evaluated the OP-nerve agent diisopropylfluorophosphate (DFP) across a wide range of concentrations (subnanomolar to micromolar) for effects on fast axonal transport of membrane-bound organelles (MBOs) that contain the amyloid precursor protein (APP) tagged with the fluorescent marker Dendra2 (APPDendra2). Both 1 and 24 hours of exposure to DFP and a positive control compound, colchicine, resulted in a decrease in the velocity of anterograde and retrograde movements of MBOs and an increase in the number of stationary MBOs. These effects occurred at picomolar (100 pM) to low nanomolar (0.1 nM) concentrations that were not associated with compromised cell viability or cytoskeletal damage. Moreover, the effects of DFP on axonal transport occurred at concentrations that did not inhibit AChE activity, and they were not blocked by cholinergic receptor antagonists. Given the fundamental importance of axonal transport to neuronal function, these observations may explain some of the long-term neurologic deficits that have been observed in humans who have been exposed to OPs.


Subject(s)
Axons/drug effects , Cell Membrane/drug effects , Cerebral Cortex/drug effects , Isoflurophate/toxicity , Organelles/drug effects , Animals , Axons/metabolism , Biological Transport/drug effects , Biological Transport/physiology , Cell Membrane/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cholinesterase Inhibitors/toxicity , Female , Organelles/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley
5.
J Neurosci ; 34(1): 249-59, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24381286

ABSTRACT

Familial Parkinson disease is associated with mutations in α-synuclein (α-syn), a presynaptic protein that has been localized not only to the cytosol, but also to mitochondria. We report here that wild-type α-syn from cell lines, and brain tissue from humans and mice, is present not in mitochondria but rather in mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a structurally and functionally distinct subdomain of the ER. Remarkably, we found that pathogenic point mutations in human α-syn result in its reduced association with MAM, coincident with a lower degree of apposition of ER with mitochondria, a decrease in MAM function, and an increase in mitochondrial fragmentation compared with wild-type. Although overexpression of wild-type α-syn in mutant α-syn-expressing cells reverted the fragmentation phenotype, neither overexpression of the mitochondrial fusion/MAM-tethering protein MFN2 nor inhibition/ablation of the mitochondrial fission protein DRP1 was able to do so, implying that α-syn operates downstream of the mitochondrial fusion/fission machinery. These novel results indicate that wild-type α-syn localizes to the MAM and modulates mitochondrial morphology, and that these behaviors are impaired by pathogenic mutations in α-syn. We believe that our results have far-reaching implications for both our understanding of α-syn biology and the treatment of synucleinopathies.


Subject(s)
Endoplasmic Reticulum/chemistry , Mitochondria/chemistry , alpha-Synuclein/analysis , Animals , Cells, Cultured , Female , HeLa Cells , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic
6.
J Neurochem ; 128(2): 330-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24117942

ABSTRACT

Autosomal-dominant Alzheimer's disease (ADAD) is a genetic disorder caused by mutations in Amyloid Precursor Protein (APP) or Presenilin (PSEN) genes. Studying the mechanisms underlying these mutations can provide insight into the pathways that lead to AD pathology. The majority of biochemical studies on APP mutations to-date have focused on comparing mechanisms between mutations at different codons. It has been assumed that amino acid position is a major determinant of protein dysfunction and clinical phenotype. However, the differential effect of mutations at the same codon has not been sufficiently addressed. In the present study we compared the effects of the aggressive ADAD-associated APP I716F mutation with I716V and I716T on APP processing in human neuroglioma and CHO-K1 cells. All APP I716 mutations increased the ratio of Aß42/40 and changed the product line preference of γ-secretase towards Aß38 production. In addition, the APP I716F mutation impaired the ε-cleavage and the fourth cleavage of γ-secretase and led to abnormal APP ß-CTF accumulation at the plasma membrane. Taken together, these data indicate that APP mutations at the same codon can induce diverse abnormalities in APP processing, some resembling PSEN1 mutations. These differential effects could explain the clinical differences observed among ADAD patients bearing different APP mutations at the same position. The amyloid precursor protein (APP) I716F mutation is associated with autosomal dominant Alzheimer's disease with the youngest age-at-onset for the APP locus. Here, we describe that this mutation, when compared to two other familial Alzheimer's disease mutations at the same codon (I716V and I716T), interfered distinctly with γ-secretase cleavage. While all three mutations direct γ-secretase cleavage towards the 48→38 production line, the APP I716F mutation also impaired the ε-cleavage and the fourth cleavage of γ-secretase, resembling a PSEN1 mutation. These features may contribute to the aggressiveness of this mutation.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/genetics , Adult , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cell Membrane/metabolism , Codon , Cricetinae , Cricetulus , Humans , Mutation
7.
J Neurosci ; 32(1): 229-42, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22219285

ABSTRACT

Mutations in Cu,Zn superoxide dismutase (SOD1) cause familial amyotrophic lateral sclerosis (FALS), a rapidly fatal motor neuron disease. Mutant SOD1 has pleiotropic toxic effects on motor neurons, among which mitochondrial dysfunction has been proposed as one of the contributing factors in motor neuron demise. Mitochondria are highly dynamic in neurons; they are constantly reshaped by fusion and move along neurites to localize at sites of high-energy utilization, such as synapses. The finding of abnormal mitochondria accumulation in neuromuscular junctions, where the SOD1-FALS degenerative process is though to initiate, suggests that impaired mitochondrial dynamics in motor neurons may be involved in pathogenesis. We addressed this hypothesis by live imaging microscopy of photo-switchable fluorescent mitoDendra in transgenic rat motor neurons expressing mutant or wild-type human SOD1. We demonstrate that mutant SOD1 motor neurons have impaired mitochondrial fusion in axons and cell bodies. Mitochondria also display selective impairment of retrograde axonal transport, with reduced frequency and velocity of movements. Fusion and transport defects are associated with smaller mitochondrial size, decreased mitochondrial density, and defective mitochondrial membrane potential. Furthermore, mislocalization of mitochondria at synapses among motor neurons, in vitro, correlates with abnormal synaptic number, structure, and function. Dynamics abnormalities are specific to mutant SOD1 motor neuron mitochondria, since they are absent in wild-type SOD1 motor neurons, they do not involve other organelles, and they are not found in cortical neurons. Together, these results suggest that impaired mitochondrial dynamics may contribute to the selective degeneration of motor neurons in SOD1-FALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Motor Neurons/metabolism , Superoxide Dismutase/deficiency , Synapses/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Disease Models, Animal , Energy Metabolism/genetics , Female , Humans , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/genetics , Motor Neurons/pathology , Pregnancy , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Synapses/pathology
8.
Hum Mol Genet ; 20(15): 2964-74, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21586654

ABSTRACT

Mutations in the heart and muscle isoform of adenine nucleotide translocator 1 (ANT1) are associated with autosomal-dominant progressive external opthalmoplegia (adPEO) clinically characterized by exercise intolerance, ptosis and muscle weakness. The pathogenic mechanisms underlying the mitochondrial myopathy caused by ANT1 mutations remain largely unknown. In yeast, expression of ANT1 carrying mutations corresponding to the human adPEO ones causes a wide range of mitochondrial abnormalities. However, functional studies of ANT1 mutations in mammalian cells are lacking, because they have been hindered by the fact that ANT1 expression leads to apoptotic cell death in commonly utilized replicating cell lines. Here, we successfully express functional ANT1 in differentiated mouse myotubes, which naturally contain high levels of ANT1, without causing cell death. We demonstrate, for the first time in these disease-relevant mammalian cells, that mutant human ANT1 causes dominant mitochondrial defects characterized by decreased ADP-ATP exchange function and abnormal translocator reversal potential. These abnormalities are not due to ANT1 loss of function, because knocking down Ant1 in myotubes causes functional changes different from ANT1 mutants. Under certain physiological conditions, mitochondria consume ATP to maintain membrane potential by reversing the ADP-ATP transport. The modified properties of mutant ANT1 can be responsible for disease pathogenesis in adPEO, because exchange reversal occurring at higher than normal membrane potential can cause excessive energy depletion and nucleotide imbalance in ANT1 mutant muscle cells.


Subject(s)
Adenine Nucleotide Translocator 1/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Mitochondria, Muscle/metabolism , Mutation , Adenine Nucleotide Translocator 1/genetics , Animals , Apoptosis/genetics , Blotting, Western , Cell Line , Cell Survival , Electrophoresis, Polyacrylamide Gel , Humans , Immunohistochemistry , Mice , Reverse Transcriptase Polymerase Chain Reaction
9.
Brain ; 135(Pt 9): 2865-74, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22961554

ABSTRACT

Amyotrophic lateral sclerosis is a devastating neurodegenerative disorder that is more prevalent in males than in females. A similar gender difference has been reported in some strains of transgenic mouse models of familial amyotrophic lateral sclerosis harbouring the G93A mutation in CuZn superoxide dismutase. Mitochondrial damage caused by pathological alterations in Ca(2+) accumulation is frequently involved in neurodegenerative diseases, including CuZn superoxide dismutase-related amyotrophic lateral sclerosis, but its association with gender is not firmly established. In this study, we examined the effects of genetic ablation of cyclophilin D on gender differences in mice expressing G93A mutant CuZn superoxide dismutase. Cyclophilin D is a mitochondrial protein that promotes mitochondrial damage from accumulated Ca(2+). As anticipated, we found that cyclophilin D ablation markedly increased Ca(2+) retention in brain mitochondria of both males and females. Surprisingly, cyclophilin D ablation completely abolished the phenotypic advantage of G93A females, with no effect on disease in males. We also found that the 17ß-oestradiol decreased Ca(2+) retention in brain mitochondria, and that cyclophilin D ablation abolished this effect. Furthermore, 17ß-oestradiol protected G93A cortical neurons and spinal cord motor neurons against glutamate toxicity, but the protection was lost in neurons lacking cyclophilin D. Taken together, these results identify a novel mechanism of oestrogen-mediated neuroprotection in CuZn superoxide dismutase-related amyotrophic lateral sclerosis, whereby Ca(2+) overload and mitochondrial damage are prevented in a cyclophilin D-dependent manner. Such a protective mechanism may contribute to the lower incidence and later onset of amyotrophic lateral sclerosis, and perhaps other chronic neurodegenerative diseases, in females.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Calcium/metabolism , Cyclophilins/genetics , Estradiol/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Neurons/metabolism , Neurons/pathology , Phenotype , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
10.
Proc Natl Acad Sci U S A ; 107(1): 378-83, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19966284

ABSTRACT

Phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1) and PARK2/Parkin mutations cause autosomal recessive forms of Parkinson's disease. Upon a loss of mitochondrial membrane potential (DeltaPsi(m)) in human cells, cytosolic Parkin has been reported to be recruited to mitochondria, which is followed by a stimulation of mitochondrial autophagy. Here, we show that the relocation of Parkin to mitochondria induced by a collapse of DeltaPsi(m) relies on PINK1 expression and that overexpression of WT but not of mutated PINK1 causes Parkin translocation to mitochondria, even in cells with normal DeltaPsi(m). We also show that once at the mitochondria, Parkin is in close proximity to PINK1, but we find no evidence that Parkin catalyzes PINK1 ubiquitination or that PINK1 phosphorylates Parkin. However, co-overexpression of Parkin and PINK1 collapses the normal tubular mitochondrial network into mitochondrial aggregates and/or large perinuclear clusters, many of which are surrounded by autophagic vacuoles. Our results suggest that Parkin, together with PINK1, modulates mitochondrial trafficking, especially to the perinuclear region, a subcellular area associated with autophagy. Thus by impairing this process, mutations in either Parkin or PINK1 may alter mitochondrial turnover which, in turn, may cause the accumulation of defective mitochondria and, ultimately, neurodegeneration in Parkinson's disease.


Subject(s)
Autophagy/physiology , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/metabolism , Cell Line , Humans , Ionophores/metabolism , Microtubules/metabolism , Microtubules/ultrastructure , Mitochondria/ultrastructure , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Binding , Protein Kinases/genetics , Protein Transport/physiology , Ubiquitin-Protein Ligases/genetics
11.
J Neurosci ; 31(44): 15826-37, 2011 Nov 02.
Article in English | MEDLINE | ID: mdl-22049426

ABSTRACT

Mutations in Cu,Zn superoxide dismutase (SOD1) are associated with familial amyotrophic lateral sclerosis (ALS). Mutant SOD1 causes a complex array of pathological events, through toxic gain of function mechanisms, leading to selective motor neuron degeneration. Mitochondrial dysfunction is among the well established toxic effects of mutant SOD1, but its mechanisms are just starting to be elucidated. A portion of mutant SOD1 is localized in mitochondria, where it accumulates mostly on the outer membrane and inside the intermembrane space (IMS). Evidence in cultured cells suggests that mutant SOD1 in the IMS causes mitochondrial dysfunction and compromises cell viability. Therefore, to test its pathogenic role in vivo we generated transgenic mice expressing G93A mutant or wild-type (WT) human SOD1 targeted selectively to the mitochondrial IMS (mito-SOD1). We show that mito-SOD1 is correctly localized in the IMS, where it oligomerizes and acquires enzymatic activity. Mito-G93ASOD1 mice, but not mito-WTSOD1 mice, develop a progressive disease characterized by body weight loss, muscle weakness, brain atrophy, and motor impairment, which is more severe in females. These symptoms are associated with reduced spinal motor neuron counts and impaired mitochondrial bioenergetics, characterized by decreased cytochrome oxidase activity and defective calcium handling. However, there is no evidence of muscle denervation, a cardinal pathological feature of ALS. Together, our findings indicate that mutant SOD1 in the mitochondrial IMS causes mitochondrial dysfunction and neurodegeneration, but per se it is not sufficient to cause a full-fledged ALS phenotype, which requires the participation of mutant SOD1 localized in other cellular compartments.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Brain/ultrastructure , Mitochondria , Mutation/genetics , Spinal Cord/ultrastructure , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/mortality , Analysis of Variance , Animals , Body Weight/genetics , Brain/pathology , Calcium/metabolism , Disease Models, Animal , Energy Metabolism/genetics , Heart , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Muscle, Skeletal/pathology , Myocardium/pathology , Nerve Tissue Proteins/metabolism , Spinal Cord/pathology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
12.
Eur J Immunol ; 41(8): 2323-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21538348

ABSTRACT

Adiponectin (APN), a cytokine constitutively produced in fat tissue, has been shown to exert anti-inflammatory effects in various disease models. While the influence of APN on monocytic cells has been extensively studied in vitro, little is known about its role in T cells. In this study, we show that while <10% of human peripheral blood T cells express adiponectin receptors (AdipoRs) on their surface, most T cells store AdipoRs in intracellular compartments. AdipoRs colocalized with immune regulatory molecules CTLA-4 and TIRC7 within clathrin-coated vesicles. After stimulation, the expression of adiponectin receptor 1 (AdipoR1) and AdipoR2 was upregulated on the surface of antigen-specific T cells, as determined by tetramer or CD137 staining, and AdipoR1 and AdipoR2 coexpressed with CTLA-4. Addition of APN resulted in a significant diminution of antigen-specific T-cell expansion. Mechanistically, APN enhanced apoptosis and inhibited proliferation of antigen-specific T-cell lines. Further, APN directly inhibited cytokine production in response to antigen stimulation. In line with the in vitro data, APN-deficient (knockout, KO) mice had higher frequencies of CD137(+) T cells upon Coxsackie B virus infection. Altogether, our data suggest that APN is a novel negative T-cell regulator. In contrast to the CTLA-4 ligand B7 only expressed on APCs, APN is abundant in human plasma.


Subject(s)
Adiponectin/immunology , Antigens/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Adiponectin/genetics , Adiponectin/pharmacology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , CTLA-4 Antigen , Cell Proliferation/drug effects , Cells, Cultured , Clathrin-Coated Vesicles/immunology , Clathrin-Coated Vesicles/metabolism , Coxsackievirus Infections/genetics , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Flow Cytometry , Gene Expression , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Jurkat Cells , K562 Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Receptors, Adiponectin/genetics , Receptors, Adiponectin/immunology , Receptors, Adiponectin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Vacuolar Proton-Translocating ATPases/immunology , Vacuolar Proton-Translocating ATPases/metabolism
13.
Brain ; 134(Pt 1): 196-209, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21078595

ABSTRACT

Motor axon degeneration is a critical but poorly understood event leading to weakness and muscle atrophy in motor neuron diseases. Here, we investigated oxidative stress-mediated axonal degeneration in mice lacking the antioxidant enzyme, Cu,Zn superoxide dismutase (SOD1). We demonstrate a progressive motor axonopathy in these mice and show that Sod1(-/-) primary motor neurons extend short axons in vitro with reduced mitochondrial density. Sod1(-/-) neurons also show oxidation of mitochondrial--but not cytosolic--thioredoxin, suggesting that loss of SOD1 causes preferential oxidative stress in mitochondria, a primary source of superoxide in cells. SOD1 is widely regarded as the cytosolic isoform of superoxide dismutase, but is also found in the mitochondrial intermembrane space. The functional significance of SOD1 in the intermembrane space is unknown. We used a transgenic approach to express SOD1 exclusively in the intermembrane space and found that mitochondrial SOD1 is sufficient to prevent biochemical and morphological defects in the Sod1(-/-) model, and to rescue the motor phenotype of these mice when followed to 12 months of age. These results suggest that SOD1 in the mitochondrial intermembrane space is fundamental for motor axon maintenance, and implicate oxidative damage initiated at mitochondrial sites in the pathogenesis of motor axon degeneration.


Subject(s)
Intracellular Membranes/metabolism , Mitochondria/genetics , Motor Neurons/metabolism , Superoxide Dismutase/genetics , Analysis of Variance , Animals , Blotting, Western , Cells, Cultured , Intracellular Membranes/pathology , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Motor Neurons/pathology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
14.
Hum Mol Genet ; 18(23): 4552-64, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19779023

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by motor neuron degeneration. Mutations in Cu,Zn-superoxide dismutase (SOD1) are responsible for 20% of familial ALS cases via a toxic gain of function. In mutant SOD1 transgenic mice, mitochondria of spinal motor neurons develop abnormal morphology, bioenergetic defects and degeneration, which are presumably implicated in disease pathogenesis. SOD1 is mostly a cytosolic protein, but a substantial portion is associated with organelles, including mitochondria, where it localizes predominantly in the intermembrane space (IMS). However, whether mitochondrial mutant SOD1 contributes to disease pathogenesis remains to be elucidated. We have generated NSC34 motor neuronal cell lines expressing wild-type or mutant SOD1 containing a cleavable IMS targeting signal to directly investigate the pathogenic role of mutant SOD1 in mitochondria. We show that mitochondrially-targeted SOD1 localizes to the IMS, where it is enzymatically active. We prove that mutant IMS-targeted SOD1 causes neuronal toxicity under metabolic and oxidative stress conditions. Furthermore, we demonstrate for the first time neurite mitochondrial fragmentation and impaired mitochondrial dynamics in motor neurons expressing IMS mutant SOD1. These defects are associated with impaired maintenance of neuritic processes. Our findings demonstrate that mutant SOD1 localized in the IMS is sufficient to determine mitochondrial abnormalities and neuronal toxicity, and contributes to ALS pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Mitochondria/enzymology , Motor Neurons/enzymology , Mutation , Superoxide Dismutase/genetics , Superoxide Dismutase/toxicity , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Line , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondrial Membranes/enzymology , Motor Neurons/pathology , Oxidative Stress , Protein Transport , Superoxide Dismutase-1
15.
J Neurosci Methods ; 334: 108535, 2020 Jan 20.
Article in English | MEDLINE | ID: mdl-31972184

ABSTRACT

BACKGROUND: The genetically encoded calcium (Ca2+) sensor GCaMP6 has been widely used for imaging Ca2+ transients in neuronal somata, dendrites, and synapses. NEW METHOD: Here we describe five new transgenic mouse lines expressing GCaMP6F (fast) or GCaMP6S (slow) in the central and peripheral nervous system under the control of theThy1.2 promoter. RESULTS: These transgenic lines exhibit stable and layer-specific expression of GCaMP6 in multiple brain regions. They have several unique features compared to existing Thy1.2-GCaMP6 mice, including sparse expression of GCaMP6 in layer V pyramidal neurons of the cerebral cortex, motor neurons in the spinal cord, as well as sensory neurons in dorsal root ganglia (DRG). We further demonstrate that these mouse lines allow for robust detection of Ca2+ transients in neuronal somata and apical dendrites in the cerebral cortex of both anesthetized and awake behaving mice, as well as in DRG neurons. COMPARISON WITH EXISTING METHOD(S): These transgenic lines allows Ca2+ imaging of dendrites and somas of pyramidal neurons in specific cortical layers that is difficult to achieve with existing methods. CONCLUSIONS: These GCaMP6 transgenic lines thus provide useful tools for functional analysis of neuronal circuits in both central and peripheral nervous systems.

16.
FASEB J ; 20(12): 2165-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16940437

ABSTRACT

Intracellular deposition of the beta-amyloid (Abeta) peptide is an increasingly recognized pathological hallmark associated with neurodegeneration and muscle wasting in Alzheimer's disease (AD) and inclusion body myositis (IBM), respectively. Previous reports have implicated dysregulation of beta-amyloid precursor protein (betaAPP) expression in IBM. Accumulation of full-length betaAPP, its various proteolytic derivatives including Abeta, and phospho-tau into vacuolated inclusions is an early pathogenic event. We previously reported on a statistical tendency favoring fast twitch fiber involvement in IBM, reminiscent of the tissue specific patterns of misfolded protein deposition seen in neurodegenerative diseases. To test this principle, we generated an animal model in which human wild-type (WT) betaAPP expression was limited to postnatal type II skeletal muscle. Hemizygous transgenic mice harboring increased levels of holo betaAPP751 and Abeta in skeletal muscle fibers became significantly weaker with age compared with nontransgenic littermates and exhibited typical myopathic features. A subpopulation of dissociated muscle fibers from transgenic mice exhibited a 2-fold increase in resting calcium and membrane depolarization compared with nontransgenic littermates. Taken together, these data indicate that overexpression of human betaAPP in fast twitch skeletal muscle of transgenic mice is sufficient for the development of some features characteristic of IBM, including abnormal tau histochemistry. The increase in resting calcium and depolarization are novel findings, suggesting both a mechanism for the weakness and an avenue for therapeutic intervention in IBM.


Subject(s)
Amyloid beta-Protein Precursor/pharmacology , Calcium/metabolism , Muscle Fibers, Fast-Twitch/pathology , Myositis, Inclusion Body/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Homeostasis/drug effects , Humans , Membrane Potentials/drug effects , Mice , Mice, Transgenic , Muscle Fibers, Fast-Twitch/ultrastructure , Muscle, Skeletal/metabolism , Tissue Distribution
17.
Dis Model Mech ; 10(12): 1529-1538, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29259026

ABSTRACT

Friedreich ataxia (FRDA) is an autosomal recessive neurodegenerative disorder with progressive ataxia that affects both the peripheral and central nervous system (CNS). While later CNS neuropathology involves loss of large principal neurons and glutamatergic and GABAergic synaptic terminals in the cerebellar dentate nucleus, early pathological changes in FRDA cerebellum remain largely uncharacterized. Here, we report early cerebellar VGLUT1 (SLC17A7)-specific parallel fiber (PF) synaptic deficits and dysregulated cerebellar circuit in the frataxin knock-in/knockout (KIKO) FRDA mouse model. At asymptomatic ages, VGLUT1 levels in cerebellar homogenates are significantly decreased, whereas VGLUT2 (SLC17A6) levels are significantly increased, in KIKO mice compared with age-matched controls. Additionally, GAD65 (GAD2) levels are significantly increased, while GAD67 (GAD1) levels remain unaltered. This suggests early VGLUT1-specific synaptic input deficits, and dysregulation of VGLUT2 and GAD65 synaptic inputs, in the cerebellum of asymptomatic KIKO mice. Immunohistochemistry and electron microscopy further show specific reductions of VGLUT1-containing PF presynaptic terminals in the cerebellar molecular layer, demonstrating PF synaptic input deficiency in asymptomatic and symptomatic KIKO mice. Moreover, the parvalbumin levels in cerebellar homogenates and Purkinje neurons are significantly reduced, but preserved in other interneurons of the cerebellar molecular layer, suggesting specific parvalbumin dysregulation in Purkinje neurons of these mice. Furthermore, a moderate loss of large principal neurons is observed in the dentate nucleus of asymptomatic KIKO mice, mimicking that of FRDA patients. Our findings thus identify early VGLUT1-specific PF synaptic input deficits and dysregulated cerebellar circuit as potential mediators of cerebellar dysfunction in KIKO mice, reflecting developmental features of FRDA in this mouse model.


Subject(s)
Cerebellum/pathology , Friedreich Ataxia/metabolism , Friedreich Ataxia/pathology , Synapses/metabolism , Synapses/pathology , Vesicular Glutamate Transport Protein 1/metabolism , Aging/pathology , Animals , Biomarkers/metabolism , Disease Models, Animal , Iron-Binding Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Parvalbumins/metabolism , Presynaptic Terminals/metabolism , Purkinje Cells/metabolism , Purkinje Cells/pathology , Purkinje Cells/ultrastructure , Synapses/ultrastructure , Frataxin
18.
Dis Model Mech ; 10(11): 1343-1352, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29125827

ABSTRACT

Friedreich ataxia (FRDA), the most common recessive inherited ataxia, results from deficiency of frataxin, a small mitochondrial protein crucial for iron-sulphur cluster formation and ATP production. Frataxin deficiency is associated with mitochondrial dysfunction in FRDA patients and animal models; however, early mitochondrial pathology in FRDA cerebellum remains elusive. Using frataxin knock-in/knockout (KIKO) mice and KIKO mice carrying the mitoDendra transgene, we show early cerebellar deficits in mitochondrial biogenesis and respiratory chain complexes in this FRDA model. At asymptomatic stages, the levels of PGC-1α (PPARGC1A), the mitochondrial biogenesis master regulator, are significantly decreased in cerebellar homogenates of KIKO mice compared with age-matched controls. Similarly, the levels of the PGC-1α downstream effectors, NRF1 and Tfam, are significantly decreased, suggesting early impaired cerebellar mitochondrial biogenesis pathways. Early mitochondrial deficiency is further supported by significant reduction of the mitochondrial markers GRP75 (HSPA9) and mitofusin-1 in the cerebellar cortex. Moreover, the numbers of Dendra-labeled mitochondria are significantly decreased in cerebellar cortex, confirming asymptomatic cerebellar mitochondrial biogenesis deficits. Functionally, complex I and II enzyme activities are significantly reduced in isolated mitochondria and tissue homogenates from asymptomatic KIKO cerebella. Structurally, levels of the complex I core subunit NUDFB8 and complex II subunits SDHA and SDHB are significantly lower than those in age-matched controls. These results demonstrate complex I and II deficiency in KIKO cerebellum, consistent with defects identified in FRDA patient tissues. Thus, our findings identify early cerebellar mitochondrial biogenesis deficits as a potential mediator of cerebellar dysfunction and ataxia, thereby providing a potential therapeutic target for early intervention of FRDA.


Subject(s)
Cerebellum/metabolism , Cerebellum/pathology , Friedreich Ataxia/metabolism , Friedreich Ataxia/pathology , Organelle Biogenesis , Animals , Biomarkers/metabolism , Disease Models, Animal , Electron Transport , Iron-Binding Proteins/metabolism , Mice, Knockout , Protein Subunits/metabolism , Frataxin
19.
J Neurosci ; 25(47): 10960-9, 2005 Nov 23.
Article in English | MEDLINE | ID: mdl-16306409

ABSTRACT

Early events in Alzheimer's disease (AD) pathogenesis implicate the accumulation of beta-amyloid (Abeta) peptide inside neurons in vulnerable brain regions. However, little is known about the consequences of intraneuronal Abeta on signaling mechanisms. Here, we demonstrate, using an inducible viral vector system to drive intracellular expression of Abeta42 peptide in primary neuronal cultures, that this accumulation results in the inhibition of the Akt survival signaling pathway. Induction of intraneuronal Abeta42 expression leads to a sequential decrease in levels of phospho-Akt, increase in activation of glycogen synthase kinase-3beta, and apoptosis. Downregulation of Akt also paralleled intracellular Abeta accumulation in vivo in the Tg2576 AD mouse model. Overexpression of constitutively active Akt reversed the toxic effects of Abeta through a mechanism involving the induction of heat shock proteins (Hsps). We used a small-interfering RNA approach to explore the possibility of a link between Akt activity and Hsp70 expression and concluded that neuroprotection by Akt could be mediated through downstream induction of Hsp70 expression. These results suggest that the early dysfunction associated with intraneuronal Abeta accumulation in AD involve the associated impairments of Akt signaling and suppression of the stress response.


Subject(s)
Amyloid beta-Peptides/metabolism , Down-Regulation , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stress, Physiological/physiopathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/poisoning , Animals , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Extracellular Fluid/metabolism , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/metabolism , Intracellular Membranes/metabolism , Mice , Mice, Transgenic , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , Proto-Oncogene Proteins c-akt/physiology , Rats , Rats, Sprague-Dawley , Stress, Physiological/metabolism , Tissue Distribution
20.
J Neurosci ; 24(7): 1700-6, 2004 Feb 18.
Article in English | MEDLINE | ID: mdl-14973234

ABSTRACT

Intracellular beta-amyloid 42 (Abeta42) accumulation is increasingly recognized as an early event in the pathogenesis of Alzheimer's disease (AD). We have developed a doxycycline-inducible adenoviral-based system that directs intracellular Abeta42 expression and accumulation into the endoplasmic reticulum of primary neuronal cultures in a regulated manner. Abeta42 exhibited a perinuclear distribution in cell bodies and an association with vesicular compartments. Virally expressed intracellular Abeta42 was toxic to neuronal cultures 24 hr after induction in a dose-dependent manner. Abeta42 expression prompted the rapid induction of stress-inducible Hsp70 protein in neurons, and virally mediated Hsp70 overexpression rescued neurons from the toxic effects of intracellular Abeta accumulation. Together, these results implicate the cellular stress response as a possible modulator of Abeta-induced toxicity in neuronal cultures.


Subject(s)
Amyloid beta-Peptides/biosynthesis , HSP70 Heat-Shock Proteins/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Peptide Fragments/biosynthesis , Adenoviridae/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Endoplasmic Reticulum/metabolism , Gene Expression/drug effects , Genetic Vectors/genetics , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/pharmacology , Humans , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/genetics , Peptide Fragments/toxicity , Rats , Rats, Sprague-Dawley , Stress, Physiological/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL