Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Aging Neurosci ; 15: 1055433, 2023.
Article in English | MEDLINE | ID: mdl-36819730

ABSTRACT

With the recent legalization of inhaled cannabis for medicinal and recreational use, the elderly represents one of the newest, rapidly growing cohorts of cannabis users. To understand the neurobiological effects of cannabis on the aging brain, 19-20 months old mice were divided into three groups exposed to vaporized cannabis containing ~10% Δ9-THC, ~10% CBD, or placebo for 30 min each day. Voxel based morphometry, diffusion weighted imaging, and resting state functional connectivity data were gathered after 28 days of exposure and following a two-week washout period. Tail-flick, open field, and novel object preference tests were conducted to explore analgesic, anxiolytic, and cognitive effects of cannabis, respectively. Vaporized cannabis high in Δ9-THC and CBD achieved blood levels reported in human users. Mice showed antinociceptive effects to chronic Δ9-THC without tolerance while the anxiolytic and cognitive effects of Δ9-THC waned with treatment. CBD had no effect on any of the behavioral measures. Voxel based morphometry showed a decrease in midbrain dopaminergic volume to chronic Δ9-THC followed but an increase after a two-week washout. Fractional anisotropy values were reduced in the same area by chronic Δ9-THC, suggesting a reduction in gray matter volume. Cannabis high in CBD but not THC increased network strength and efficiency, an effect that persisted after washout. These data would indicate chronic use of inhaled cannabis high in Δ9-THC can be an effective analgesic but not for treatment of anxiety or cognitive decline. The dopaminergic midbrain system was sensitive to chronic Δ9-THC but not CBD showing robust plasticity in volume and water diffusivity prior to and following drug cessation an effect possibly related to the abuse liability of Δ9-THC. Chronic inhaled CBD resulted in enhanced global network connectivity that persisted after drug cessation. The behavioral consequences of this sustained change in brain connectivity remain to be determined.

2.
Neurosci Lett ; 758: 136010, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34090937

ABSTRACT

Aging is a major risk factor for Alzheimer's disease (AD), the most common cause of dementia worldwide. TDP-43 proteinopathy is reported to be associated with AD pathology is almost 50% of cases. Our exploratory study examined near end-stage (28 months old) mice selectively driving expression of human TDP-43 in the hippocampus and cortex in an APP/PSEN1 background. We hypothesized that hippocampal neuropathology caused by ß-amyloidosis with TDP-43 proteinopathy induced in this model, resembling the pathology seen in AD cases, manifest with changes in resting state functional connectivity. In vivo magnetic resonance imaging and post-mortem histology were performed on four genotypes: wild type, APP/PSEN1, Camk2a/TDP-43, and Camk2a/TDP-43/APP/PSEN1. Our results revealed loss of functional coupling in hippocampus and amygdala that was associated with severe neuronal loss in dentate gyrus of Camk2a/TDP-43/APP/PSEN1 mice compared to APP/PSEN1 and wild type mice. The loss of cells was accompanied by high background of ß-amyloid plaques with sparse phosphorylated TDP-43 pathology. The survival rate was also reduced in Camk2a/TDP-43/APP/PSEN1 mice compared to other groups. This end-of-life study provides exploratory data to reach a better understanding of the role of TDP-43 hippocampal neuropathology in diseases with co-pathologies of TDP-43 proteinopathy and ß-amyloidosis such as AD and limbic predominant age-related TDP-43 encephalopathy (LATE).


Subject(s)
Aging/pathology , Alzheimer Disease/physiopathology , Hippocampus/pathology , TDP-43 Proteinopathies/physiopathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amygdala/diagnostic imaging , Amygdala/pathology , Amygdala/physiology , Amyloid beta-Protein Precursor/genetics , Animals , Brain Mapping , DNA-Binding Proteins/genetics , Disease Models, Animal , Female , Hippocampus/diagnostic imaging , Hippocampus/physiopathology , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Presenilin-1/genetics , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/pathology
3.
Neurosci Lett ; 678: 8-15, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29715546

ABSTRACT

Transactive response DNA-binding protein of 43 kDa (TDP-43) functions as a heterogeneous nuclear ribonucleoprotein and is the major pathological protein in frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis/motor neuron disease (ALS/MND). TDP-43 pathology may also be present as a comorbidity in approximately 20-50% of sporadic Alzheimer's disease cases. In a mouse model of MND, full-length TDP-43 increases association with the mitochondria and blocking the TDP-43/mitochondria interaction ameliorates motor dysfunction. Utilizing a proteomics screen, several mitochondrial TDP-43-interacting partners were identified, including voltage-gated anion channel 1 (VDAC1) and prohibitin 2 (PHB2), a crucial mitophagy receptor. Overexpression of TDP-43 led to an increase in PHB2 whereas TDP-43 knockdown reduced PHB2 expression in cells treated with carbonyl cyanide m-chlorophenylhydrazone (CCCP), an inducer of mitophagy. These results suggest that TDP-43 expression contributes to metabolism and mitochondrial function however we show no change in bioenergetics when TDP-43 is overexpressed and knocked down in HEK293T cells. Furthermore, the fusion protein mitofusin 2 (MFN2) interacts in complex with TDP-43 and selective expression of human TDP-43 in the hippocampus and cortex induced an age-dependent change in Mfn2 expression. Mitochondria morphology is altered in 9-month-old mice selectively expressing TDP-43 in an APP/PS1 background compared with APP/PS1 littermates. We further confirmed TDP-43 localization to the mitochondria using immunogold labeled TDP-43 transmission electron microscopy (TEM) and mitochondrial isolation methods There was no increase in full-length TDP-43 localized to the mitochondria in APP/PS1 mice compared to wild-type (littermates); however, using C- and N-terminal-specific TDP-43 antibodies, the N-terminal (27 kDa, N27) and C-terminal (30 kDa, C30) fragments of TDP-43 are greatly enriched in mitochondrial fractions. In addition, when the mitochondrial peptidase (PMPCA) is overexpressed there is an increase in the N-terminal fragment (N27). These results suggest that TDP-43 processing may contribute to metabolism and mitochondrial function.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Mitophagy , Animals , Cerebral Cortex/metabolism , Disease Models, Animal , GTP Phosphohydrolases/metabolism , Hippocampus/metabolism , Hippocampus/ultrastructure , Mice , Mitochondria/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Prohibitins , Repressor Proteins/metabolism
4.
Neurobiol Dis ; 103: 154-162, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28416393

ABSTRACT

Although the main focus in Alzheimer's disease (AD) has been an investigation of mechanisms causing Aß plaque deposition and tau tangle formation, recent studies have shown that phosphorylated TDP-43 pathology is present in up to 50% of sporadic cases. Furthermore, elevated phosphorylated TDP-43 has been associated with more severe AD pathology. Therefore, we hypothesized that TDP-43 may regulate amyloid-beta precursor protein (APP) trafficking and tau phosphorylation/aggregation. In order to examine the role of TDP-43 in AD, we developed a transgenic mouse that overexpresses hippocampal and cortical neuronal TDP-43 in a mouse expressing familial mutations (K595N and M596L) in APP and presenilin 1 (PSEN1ΔE9). In our model, increased TDP-43 was related to increased tau aggregation as evidenced by thioflavin S-positive phosphorylated tau, which may implicate TDP-43 expression in pre-tangle formation. In addition, there was increased endosomal/lysosomal localization of APP and reduced Aß plaque formation with increased TDP-43. Furthermore, there was decreased calcineurin with elevated TDP-43 expression. Since calcineurin is a phosphatase for TDP-43, the decreased calcineurin expression may be one mechanism leading to an increase in accumulation of diffuse phosphorylated TDP-43 in the hippocampus and cortex. We further show that when TDP-43 is knocked down there is an increase in calcineurin. In our model of selective TDP-43 overexpression in an APP/PSEN1 background, we show that TDP-43 decreases Aß plaque deposition while increasing abnormal tau aggregation. These observations indicate that TDP-43 may play a role in regulating APP trafficking and tau aggregation. Our data suggest that TDP-43 could be a putative target for therapeutic intervention in AD affecting both Aß plaque formation and tauopathy.


Subject(s)
DNA-Binding Proteins/biosynthesis , Plaque, Amyloid/metabolism , Protein Aggregation, Pathological/metabolism , tau Proteins/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Presenilin-1/genetics , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , tau Proteins/genetics
5.
J Med Primatol ; 44(2): 66-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25639184

ABSTRACT

BACKGROUND: Most amyotrophic lateral sclerosis (ALS) research has focused on mice, but there are distinct differences in the functional neuroanatomy of the corticospinal pathway in primates vs. rodents. A non-human primate model may be more sensitive and more predictive for therapeutic efficacy. METHODS: Rhesus macaques received recombinant adeno-associated virus (AAV9) encoding either the ALS-related pathological protein TDP-43 or a green fluorescent protein (GFP) control by intravenous administration. Motor function and electromyography were assessed over a nine-month expression interval followed by post-mortem analyses. RESULTS: Recombinant TDP-43 or GFP was stably expressed long term. Although the TDP-43 subjects did not manifest severe paralysis and atrophy, there were trends of a partial disease state in the TDP-43 subjects relative to the control. CONCLUSIONS: These data indicate that a higher gene vector dose will likely be necessary for more robust effects, yet augur that a relevant primate model is feasible.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , DNA-Binding Proteins/genetics , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors/genetics , Macaca mulatta , Administration, Intravenous , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , DNA-Binding Proteins/administration & dosage , Dependovirus/genetics , Electromyography , Green Fluorescent Proteins/administration & dosage , Humans , Motor Activity , Recombinant Proteins/genetics
6.
Mol Ther ; 21(7): 1324-34, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23689600

ABSTRACT

Pathological inclusions containing transactive response DNA-binding protein 43 kDa (TDP-43) are common in several neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). TDP-43 normally localizes predominantly to the nucleus, but during disease progression, it mislocalizes to the cytoplasm. We expressed TDP-43 in rats by an adeno-associated virus (AAV9) gene transfer method that transduces neurons throughout the central nervous system (CNS). To mimic the aberrant cytoplasmic TDP-43 found in disease, we expressed a form of TDP-43 with mutations in the nuclear localization signal sequence (TDP-NLS). The TDP-NLS was detected in both the cytoplasm and the nucleus of transduced neurons. Unlike wild-type TDP-43, expression of TDP-NLS did not induce mortality. However, the TDP-NLS induced disease-relevant motor impairments over 24 weeks. We compared the TDP-NLS to a 25 kDa C-terminal proaggregatory fragment of TDP-43 (TDP-25). The clinical phenotype of forelimb impairment was pronounced with the TDP-25 form, supporting a role of this C-terminal fragment in pathogenesis. The results advance previous rodent models by inducing cytoplasmic expression of TDP-43 in the spinal cord, and the non-lethal phenotype enabled long-term study. Approaching a more relevant disease state in an animal model that more closely mimics underlying mechanisms in human disease could unlock our ability to develop therapeutics.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/metabolism , Forelimb/metabolism , Forelimb/pathology , Animals , Blotting, Western , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Dependovirus/genetics , Female , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/metabolism , Spinal Cord/pathology
7.
Eur J Neurosci ; 34(8): 1179-88, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21777407

ABSTRACT

The neuropathological hallmark of the majority of amyotrophic lateral sclerosis (ALS) and a class of frontotemporal lobar degeneration is ubiquitinated cytoplasmic aggregates composed of transactive response DNA binding protein 43 kDa (TDP-43). Genetic manipulation of TDP-43 in animal models has been used to study the protein's role in pathogenesis. Transgenic rodents for TDP-43 have recapitulated key aspects of ALS such as paralysis, loss of spinal motor neurons and muscle atrophy. Viral vectors are an alternate approach to express pathological proteins in animals. Use of the recombinant adeno-associated virus vector serotype 9 has permitted widespread transgene expression throughout the central nervous system after intravenous administration. Expressing TDP-43 in rats with this method produced a phenotype that was consistent with and similar to TDP-43 transgenic lines. Increased levels of TDP-43 in the nucleus are toxic to neurons and sufficient to produce ALS-like symptoms. Animal models based on TDP-43 will address the relationships between TDP-43 expression levels, pathology, neuronal loss, muscle atrophy, motor function and causative mechanisms of disease. New targets that modify TDP-43 function, or targets from previous ALS models and other models of spinal cord diseases, could be tested for efficacy in the recent rodent models of ALS based on TDP-43. The vector approach could be an important therapeutic channel because the entire spinal cord can be affected from a one-time peripheral administration.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , DNA-Binding Proteins/genetics , Disease Models, Animal , Genetic Therapy/methods , Rodentia , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Animals, Genetically Modified , Gene Transfer Techniques , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Germ Cells/physiology , Humans , Spinal Cord/pathology , Spinal Cord/physiology , Spinal Cord/physiopathology
8.
J Mol Neurosci ; 45(3): 409-21, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21603978

ABSTRACT

Fused in sarcoma (FUS)-immunoreactive neuronal and glial inclusions define a novel molecular pathology called FUS proteinopathy. FUS has been shown to be a component of inclusions of familial amyotrophic lateral sclerosis with FUS mutation and three frontotemporal lobar degeneration entities, including neuronal intermediate filament inclusion disease (NIFID). The pathogenic role of FUS is unknown. In addition to FUS, many neuronal cytoplasmic inclusions (NCI) of NIFID contain aggregates of α-internexin and neurofilament proteins. Herein, we have shown that: (1) FUS becomes relatively insoluble in NIFID and there are no apparent posttranslational modifications, (2) there are no pathogenic abnormalities in the FUS gene in NIFID, and (3) immunoelectron microscopy demonstrates the fine structural localization of FUS in NIFID which has not previously been described. FUS localized to euchromatin, and strongly with paraspeckles, in nuclei, consistent with its RNA/DNA-binding functions. NCI of varying morphologies were observed. Most frequent were the "loosely aggregated cytoplasmic inclusions," 81% of which had moderate or high levels of FUS immunoreactivity. Much rarer "compact cytoplasmic inclusions" and "tangled twine ball inclusions" were FUS-immunoreactive at their granular peripheries, or heavily FUS-positive throughout, respectively. Thus, FUS may aggregate in the cytoplasm and then admix with neuronal intermediate filament accumulations.


Subject(s)
Frontotemporal Lobar Degeneration/pathology , Immunohistochemistry , Inclusion Bodies/pathology , Intermediate Filaments/pathology , Microscopy, Electron, Transmission/methods , Microscopy, Immunoelectron/methods , Neurons/pathology , RNA-Binding Protein FUS/metabolism , Adult , Aged , Brain/pathology , Female , Frontotemporal Lobar Degeneration/metabolism , Humans , Intermediate Filaments/metabolism , Male , Middle Aged , Neurofilament Proteins/metabolism
9.
J Neurochem ; 112(5): 1305-15, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20028451

ABSTRACT

Frontotemporal dementia with ubiquitin-positive inclusions (FTLD-U) can be caused by mutations in the progranulin gene (GRN). Progranulin (PGRN) is a cysteine-rich growth factor, which is proteolytically cleaved by elastase to produce several granulins (GRNs). All FTLD-U mutations in GRN characterized to date result in reduced secreted PGRN protein. We recently reported a Spanish family with progressive non-fluent aphasia and dementia in which a novel C521Y mutation segregates with disease. A second cysteine mutation (C139R) has also been reported to be disease specific. Allele-specific mRNA expression assays in brain reveal that the C521Y mutant allele is expressed at similar levels to the wild-type allele. Furthermore, plasma PGRN levels in C521Y carriers are comparable with non-carrier family relatives, suggesting that the mutation does not affect PGRN protein expression and secretion in vivo. Despite normal PGRN levels C521Y and C139R mutant GRNs show reduced neurite growth-stimulating activity in vitro. Further study revealed that these mutations also cause impaired cleavage of PGRN by elastase. Our data suggest that these mutations affect the function of full-length PGRN as well as elastase cleavage of PGRN into GRNs, leading to neurodegeneration.


Subject(s)
Cysteine/genetics , Frontotemporal Dementia/genetics , Genetic Predisposition to Disease , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Mutation/genetics , Animals , Arginine/genetics , Axons/physiology , Brain/cytology , Brain/pathology , Cell Enlargement , Cell Line, Transformed , Dithiothreitol/pharmacology , Embryo, Mammalian , Enzyme-Linked Immunosorbent Assay/methods , Family Health , Frontotemporal Dementia/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Humans , Motor Neurons/cytology , Motor Neurons/drug effects , Motor Neurons/metabolism , Neurites , Pancreatic Elastase/pharmacology , Progranulins , RNA, Messenger/metabolism , Rats , Rats, Wistar , Transfection , Tyrosine/genetics
10.
Acta Neuropathol ; 118(5): 633-45, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19618195

ABSTRACT

Pathogenic mutations in the gene encoding TDP-43, TARDBP, have been reported in familial amyotrophic lateral sclerosis (FALS) and, more recently, in families with a heterogeneous clinical phenotype including both ALS and frontotemporal lobar degeneration (FTLD). In our previous study, sequencing analyses identified one variant in the 3'-untranslated region (3'-UTR) of the TARDBP gene in two affected members of one family with bvFTD and ALS and in one unrelated clinically assessed case of FALS. Since that study, brain tissue has become available and provides autopsy confirmation of FTLD-TDP in the proband and ALS in the brother of the bvFTD-ALS family and the neuropathology of those two cases is reported here. The 3'-UTR variant was not found in 982 control subjects (1,964 alleles). To determine the functional significance of this variant, we undertook quantitative gene expression analysis. Allele-specific amplification showed a significant increase of 22% (P < 0.05) in disease-specific allele expression with a twofold increase in total TARDBP mRNA. The segregation of this variant in a family with clinical bvFTD and ALS adds to the spectrum of clinical phenotypes previously associated with TARDBP variants. In summary, TARDBP variants may result in clinically and neuropathologically heterogeneous phenotypes linked by a common molecular pathology called TDP-43 proteinopathy.


Subject(s)
3' Untranslated Regions/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration , Genetic Predisposition to Disease , Genetic Variation/genetics , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Autopsy/methods , Brain/metabolism , Brain/pathology , Chi-Square Distribution , DNA Mutational Analysis/methods , Female , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Humans , Male , Middle Aged , RNA, Messenger/metabolism
11.
J Biol Chem ; 284(18): 12384-98, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19237541

ABSTRACT

Frontotemporal lobar degeneration (FTLD) with inclusion body myopathy and Paget disease of bone is a rare, autosomal dominant disorder caused by mutations in the VCP (valosin-containing protein) gene. The disease is characterized neuropathologically by frontal and temporal lobar atrophy, neuron loss and gliosis, and ubiquitin-positive inclusions (FTLD-U), which are distinct from those seen in other sporadic and familial FTLD-U entities. The major component of the ubiquitinated inclusions of FTLD with VCP mutation is TDP-43 (TAR DNA-binding protein of 43 kDa). TDP-43 proteinopathy links sporadic amyotrophic lateral sclerosis, sporadic FTLD-U, and most familial forms of FTLD-U. Understanding the relationship between individual gene defects and pathologic TDP-43 will facilitate the characterization of the mechanisms leading to neurodegeneration. Using cell culture models, we have investigated the role of mutant VCP in intracellular trafficking, proteasomal function, and cell death and demonstrate that mutations in the VCP gene 1) alter localization of TDP-43 between the nucleus and cytosol, 2) decrease proteasome activity, 3) induce endoplasmic reticulum stress, 4) increase markers of apoptosis, and 5) impair cell viability. These results suggest that VCP mutation-induced neurodegeneration is mediated by several mechanisms.


Subject(s)
Adenosine Triphosphatases/metabolism , Apoptosis , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Dementia/metabolism , Models, Biological , Mutation , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Survival/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , Cytoplasm/pathology , DNA-Binding Proteins/genetics , Dementia/genetics , Dementia/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Female , Humans , Male , Middle Aged , Protein Transport/genetics , Ubiquitination/genetics , Valosin Containing Protein
12.
Ann Neurol ; 63(4): 535-8, 2008 04.
Article in English | MEDLINE | ID: mdl-18288693

ABSTRACT

To identify novel causes of familial neurodegenerative diseases, we extended our previous studies of TAR DNA-binding protein 43 (TDP-43) proteinopathies to investigate TDP-43 as a candidate gene in familial cases of motor neuron disease. Sequencing of the TDP-43 gene led to the identification of a novel missense mutation, Ala-315-Thr, which segregates with all affected members of an autosomal dominant motor neuron disease family. The mutation was not found in 1,505 healthy control subjects. The discovery of a missense mutation in TDP-43 in a family with dominantly inherited motor neuron disease provides evidence of a direct link between altered TDP-43 function and neurodegeneration.


Subject(s)
Amino Acid Substitution/genetics , DNA-Binding Proteins/genetics , Motor Neuron Disease/genetics , Mutation, Missense/genetics , Aged , Aged, 80 and over , Alanine/genetics , Amino Acid Sequence , DNA Mutational Analysis , Female , Genes, Dominant , Humans , Male , Middle Aged , Molecular Sequence Data , Motor Neuron Disease/metabolism , Motor Neuron Disease/pathology , Pedigree , Threonine/genetics
13.
Am J Physiol Cell Physiol ; 292(1): C564-72, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16870829

ABSTRACT

Data from the use of activators and inhibitors of the AMP-activated protein kinase (AMPK) suggest that AMPK increases sensitivity of glucose transport to stimulation by insulin in muscle cells. We assayed insulin action after adenoviral (Ad) transduction of constitutively active (CA; a truncated form of AMPKalpha(1)) and dominant-negative (DN; which depletes endogenous AMPKalpha) forms of AMPKalpha (Ad-AMPKalpha-CA and Ad-AMPKalpha-DN, respectively) into C(2)C(12) myotubes. Compared with control (Ad-green fluorescent protein), Ad-AMPK-CA increased the ability of insulin to stimulate glucose transport. The increased insulin action in cells expressing AMPK-CA was suppressed by compound C (an AMPK inhibitor). Exposure of cells to 5-aminoimidazole-4-carboxamide-1beta-D-ribofuranoside (an AMPK activator) increased insulin action in uninfected myotubes and myotubes transduced with green fluorescent protein but not in Ad-AMPK-DN-infected myotubes. In Ad-AMPK-CA-transduced cells, serine phosphorylation of insulin receptor substrate 1 was decreased at a mammalian target of rapamycin (or p70 S6 kinase) target site that has been reported to be associated with insulin resistance. These data suggest that, in myotubes, activated AMPKalpha(1) is sufficient to increase insulin action and that the presence of functional AMPKalpha is required for 5-aminoimidazole-4-carboxamide-1beta,D-ribofuranoside-related increases in insulin action.


Subject(s)
Glucose/metabolism , Hypoglycemic Agents/metabolism , Insulin/metabolism , Multienzyme Complexes/pharmacology , Protein Serine-Threonine Kinases/pharmacology , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Biological Transport/drug effects , Cells, Cultured , Drug Synergism , Enzyme Activators/pharmacology , Genes, Dominant , Green Fluorescent Proteins , Insulin Receptor Substrate Proteins , Isoenzymes/pharmacology , Luminescent Agents , Mice , Multienzyme Complexes/genetics , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Ribonucleotides/pharmacology , Signal Transduction/physiology , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...