Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 134(2)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-37988169

ABSTRACT

Alzheimer's disease is characterized by the accumulation of amyloid-ß plaques, aggregation of hyperphosphorylated tau (pTau), and microglia activation. Galectin-3 (Gal3) is a ß-galactoside-binding protein that has been implicated in amyloid pathology. Its role in tauopathy remains enigmatic. Here, we showed that Gal3 was upregulated in the microglia of humans and mice with tauopathy. pTau triggered the release of Gal3 from human induced pluripotent stem cell-derived microglia in both its free and extracellular vesicular-associated (EV-associated) forms. Both forms of Gal3 increased the accumulation of pathogenic tau in recipient cells. Binding of Gal3 to pTau greatly enhanced tau fibrillation. Besides Gal3, pTau was sorted into EVs for transmission. Moreover, pTau markedly enhanced the number of EVs released by iMGL in a Gal3-dependent manner, suggesting a role of Gal3 in biogenesis of EVs. Single-cell RNA-Seq analysis of the hippocampus of a mouse model of tauopathy (THY-Tau22) revealed a group of pathogenic tau-evoked, Gal3-associated microglia with altered cellular machineries implicated in neurodegeneration, including enhanced immune and inflammatory responses. Genetic removal of Gal3 in THY-Tau22 mice suppressed microglia activation, reduced the level of pTau and synaptic loss in neurons, and rescued memory impairment. Collectively, Gal3 is a potential therapeutic target for tauopathy.


Subject(s)
Galectin 3 , Tauopathies , tau Proteins , Animals , Humans , Mice , Alzheimer Disease/pathology , Disease Models, Animal , Galectin 3/genetics , Galectin 3/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice, Transgenic , Microglia/pathology , tau Proteins/genetics , tau Proteins/metabolism , Tauopathies/genetics , Tauopathies/metabolism
2.
Mov Disord ; 37(4): 767-777, 2022 04.
Article in English | MEDLINE | ID: mdl-34951052

ABSTRACT

BACKGROUND: Polyglutamine (polyQ) diseases are dominant neurodegenerative diseases caused by an expansion of the polyQ-encoding CAG repeats in the disease-causing gene. The length of the CAG repeats is the major determiner of the age at onset (AO) of polyQ diseases, including Huntington's disease (HD) and spinocerebellar ataxia type 3 (SCA3). OBJECTIVE: We set out to identify common genetic variant(s) that may affect the AO of polyQ diseases. METHODS: Three hundred thirty-seven patients with HD or SCA3 were enrolled for targeted sequencing of 583 genes implicated in proteinopathies. In total, 16 genes were identified as containing variants that are associated with late AO of polyQ diseases. For validation, we further investigate the variants of PIAS1 because PIAS1 is an E3 SUMO (small ubiquitin-like modifier) ligase for huntingtin (HTT), the protein linked to HD. RESULTS: Biochemical analyses revealed that the ability of PIAS1S510G to interact with mutant huntingtin (mHTT) was less than that of PIAS1WT , resulting in lower SUMOylation of mHTT and lower accumulation of insoluble mHTT. Genetic knock-in of PIAS1S510G in a HD mouse model (R6/2) ameliorated several HD-like deficits (including shortened life spans, poor grip strength and motor coordination) and reduced neuronal accumulation of mHTT. CONCLUSIONS: Our findings suggest that PIAS1 is a genetic modifier of polyQ diseases. The naturally occurring variant, PIAS1S510G , is associated with late AO in polyQ disease patients and milder disease severity in HD mice. Our study highlights the possibility of targeting PIAS1 or pathways governing protein homeostasis as a disease-modifying approach for treating patients with HD. © 2021 International Parkinson and Movement Disorder Society.


Subject(s)
Huntington Disease , Proteostasis , Animals , Disease Models, Animal , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/metabolism , Ligases/metabolism , Mice , Peptides , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism
3.
Transl Psychiatry ; 9(1): 209, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31455764

ABSTRACT

Sialic acids are typically added to the end of glycoconjugates by sialyltransferases. Among the six ST8 α-N-acetyl-neuraminide α-2,8-sialyltransferases (ST8SIA) existing in adult brains, ST8SIA2 is a schizophrenia-associated gene. However, the in vivo substrates and physiological functions of most sialyltransferases are currently unknown. The ST8SIA3 is enriched in the striatum. Here, we showed that ablation of St8sia3 in mice (St8sia3-KO) led to fewer disialylated and trisialylated terminal glycotopes in the striatum of St8sia3-KO mice. Moreover, the apparent sizes of several striatum-enriched G-protein-coupled receptors (GPCRs) (including the adenosine A2A receptor (A2AR) and dopamine D1/D2 receptors (D1R and D2R)) were smaller in St8sia3-KO mice than in WT mice. A sialidase treatment removed the differences in the sizes of these molecules between St8sia3-KO and WT mice, confirming the involvement of sialylation. Expression of ST8SIA3 in the striatum of St8sia3-KO mice using adeno-associated viruses normalized the sizes of these proteins, demonstrating a direct role of ST8SIA3. The lack of ST8SIA3-mediated sialylation altered the distribution of these proteins in lipid rafts and the interaction between D2R and A2AR. Locomotor activity assays revealed altered pharmacological responses of St8sia3-KO mice to drugs targeting these receptors and verified that a greater population of D2R formed heteromers with A2AR in the striatum of St8sia3-KO mice. Since the A2AR-D2R heteromer is an important drug target for several basal ganglia diseases (such as schizophrenia and Parkinson's disease), the present study not only reveals a crucial role for ST8SIA3 in striatal functions but also provides a new drug target for basal ganglia-related diseases.


Subject(s)
Corpus Striatum/metabolism , Neurons/metabolism , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Sialyltransferases/metabolism , Animals , Membrane Microdomains/metabolism , Mice , Mice, Knockout , Sialyltransferases/genetics
4.
Mol Neurobiol ; 55(12): 8936-8952, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29616397

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive impairment and synaptic dysfunction. Adenosine is an important homeostatic modulator that controls the bioenergetic network in the brain through regulating receptor-evoked signaling pathways, bioenergetic machineries, and epigenetic-mediated gene regulation. Equilibrative nucleoside transporter 1 (ENT1) is a major adenosine transporter that recycles adenosine from the extracellular space. In the present study, we report that a small adenosine analogue (designated J4) that inhibited ENT1 prevented the decline in spatial memory in an AD mouse model (APP/PS1). Electrophysiological and biochemical analyses further demonstrated that chronic treatment with J4 normalized the impaired basal synaptic transmission and long-term potentiation (LTP) at Schaffer collateral synapses as well as the aberrant expression of synaptic proteins (e.g., NR2A and NR2B), abnormal neuronal plasticity-related signaling pathways (e.g., PKA and GSK3ß), and detrimental elevation in astrocytic A2AR expression in the hippocampus and cortex of APP/PS1 mice. In conclusion, our findings suggest that modulation of adenosine homeostasis by J4 is beneficial in a mouse model of AD. Our study provides a potential therapeutic strategy to delay the progression of AD.


Subject(s)
Adenosine/therapeutic use , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Cognitive Dysfunction/drug therapy , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , Memory Disorders/drug therapy , Memory Disorders/physiopathology , Neuronal Plasticity , Presenilin-1/metabolism , Adenosine/pharmacology , Alzheimer Disease/pathology , Animals , Astrocytes/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/prevention & control , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Equilibrative Nucleoside Transporter 1/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Mice, Transgenic , Neuronal Plasticity/drug effects , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology , Receptor, Adenosine A2A/metabolism , Synaptic Transmission/drug effects
5.
Front Neurosci ; 12: 187, 2018.
Article in English | MEDLINE | ID: mdl-29615863

ABSTRACT

The A2A adenosine receptor (A2AR) and D2 dopamine receptor (D2R) are two G-protein-coupled receptors that can form dimers and negatively regulate their partners. TAR DNA-binding protein (TDP-43) is a nuclear protein that has been implicated in amyotrophic lateral sclerosis (ALS). Mislocalization of TDP-43 from the nucleus to the cytoplasm is an early step of TDP-43 proteinopathy. Our previous studies indicated that A2AR is a potential drug target for ALS because treatment with an A2AR agonist (JMF1907; a T1-11 analog) prevents reactive oxygen species (ROS)-induced TDP-43 mislocalization in a motor neuron cell line (NSC34) and delays motor impairment in a TDP-43 transgenic ALS mouse model. Here, we set out to assess whether activation of D2R interferes with the beneficial effects of an A2AR agonist on motor neurons. We first demonstrated that A2AR and D2R are both located in motor neurons of mouse and human spinal cords and human iPSC-derived motor neurons. Expression of A2AR and D2R in NSC34 cells led to dimer formation without affecting the binding affinity of A2AR toward T1-11. Importantly, activation of D2R reduced T1-11-mediated activation of cAMP/PKA signaling and subsequent inhibition of TDP-43 mislocalization in NSC34 cells. Treatment with quinpirole (a D2 agonist) blunted the rescuing effect of T1-11 on TDP-43 mislocalization and impaired grip strength in a mouse model of ALS. Our findings suggest that D2R activation may limit the beneficial responses of an A2AR agonist in motor neurons and may have an important role in ALS pathogenesis.

6.
Mol Psychiatry ; 23(12): 2375-2390, 2018 12.
Article in English | MEDLINE | ID: mdl-29298990

ABSTRACT

Translin-associated protein X (TRAX) is a scaffold protein with various functions and has been associated with mental illnesses, including schizophrenia. We have previously demonstrated that TRAX interacts with a Gsα protein-coupled receptor, the A2A adenosine receptor (A2AR), and mediates the function of this receptor in neuritogenesis. In addition, stimulation of the A2AR markedly ameliorates DNA damage evoked by elevated oxidative stress in neurons derived from induced pluripotent stem cells (iPSCs). Here, we report that glycogen synthase kinase 3 beta (GSK3ß) and disrupted-in-schizophrenia 1 (DISC1) are two novel interacting proteins of TRAX. We present evidence to suggest that the stimulation of A2AR markedly facilitated DNA repair through the TRAX/DISC1/GSK3ß complex in a rat neuronal cell line (PC12), primary mouse neurons, and human medium spiny neurons derived from iPSCs. A2AR stimulation led to the inhibition of GSK3ß, thus dissociating the TRAX/DISC1/GSK3ß complex and facilitating the non-homologous end-joining pathway (NHEJ) by enhancing the activation of a DNA-dependent protein kinase via phosphorylation at Thr2609. Similarly, pharmacological inhibition of GSK3ß by SB216763 also facilitated the TRAX-mediated repair of oxidative DNA damage. Collectively, GSK3ß binds with TRAX and negatively affects its ability to facilitate NHEJ repair. The suppression of GSK3ß by A2AR activation or a GSK3ß inhibitor releases TRAX for the repair of oxidative DNA damage. Our findings shed new light on the molecular mechanisms underlying diseases associated with DNA damage and provides a novel target (i.e., the TRAX/DISC1/GSK3ß complex) for future therapeutic development for mental disorders.


Subject(s)
DNA-Binding Proteins/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Carrier Proteins/genetics , DNA Repair , DNA-Binding Proteins/genetics , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/physiology , Hippocampus/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurites , Neurons/metabolism , PC12 Cells , Phosphorylation , Rats , Receptor, Adenosine A2A/genetics , Signal Transduction
7.
J Biomed Sci ; 24(1): 68, 2017 Sep 04.
Article in English | MEDLINE | ID: mdl-28870220

ABSTRACT

BACKGROUND: The type VI adenylyl cyclase (AC6) is a main contributor of cAMP production in the heart. The amino acid (aa) sequence of AC6 is highly homologous to that of another major cardiac adenylyl cyclase, AC5, except for its N-terminus (AC6-N, aa 1-86). Activation of AC6, rather than AC5, produces cardioprotective effects against heart failure, while the underlying mechanism remains to be unveiled. Using an AC6-null (AC6-/-) mouse and a knockin mouse with AC6-N deletion (AC6 ΔN/ΔN), we aimed to investigate the cardioprotective mechanism of AC6 in the heart. METHODS: Western blot analysis and immunofluorescence staining were performed to determine the intracellular distribution of AC6, AC6-ΔN (a truncated AC6 lacking the first 86 amino acids), and STAT3 activation. Activities of AC6 and AC6-ΔN in the heart were assessed by cAMP assay. Apoptosis of cardiomyocytes were evaluated by the TUNEL assay and a propidium iodine-based survival assay. Fibrosis was examined by collagen staining. RESULTS: Immunofluorescence staining revealed that cardiac AC6 was mainly anchored on the sarcolemmal membranes, while AC6-ΔN was redistributed to the sarcoplasmic reticulum. AC6ΔN/ΔN and AC6-/- mice had more apoptotic myocytes and cardiac remodeling than WT mice in experimental models of isoproterenol (ISO)-induced myocardial injury. Adult cardiomyocytes isolated from AC6ΔN/ΔN or AC6-/- mice survived poorly after exposure to ISO, which produced no effect on WT cardiomyocytes under the condition tested. Importantly, ISO treatment induced cardiac STAT3 phosphorylation/activation in WT mice, but not in AC6ΔN/ΔN and AC6-/- mice. Pharmacological blockage of PKA-, Src-, or STAT3- pathway markedly reduced the survival of WT myocytes in the presence of ISO, but did not affect those of AC6ΔN/ΔN and AC6-/- myocytes, suggesting an important role of AC6 in mediating cardioprotective action through the activation of PKA-Src-STAT3-signaling. CONCLUSIONS: Collectively, AC6-N controls the anchorage of cardiac AC6 on the sarcolemmal membrane, which enables the coupling of AC6 with the pro-survival PKA-STAT3 pathway. Our findings may facilitate the development of novel therapies for heart failure.


Subject(s)
Adenylyl Cyclases/genetics , Myocytes, Cardiac/metabolism , Signal Transduction , Adenylyl Cyclases/metabolism , Animals , Apoptosis/drug effects , Heart Failure/metabolism , Mice , Mice, Inbred C57BL , Protective Agents/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
8.
Hum Mol Genet ; 26(3): 467-478, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28069792

ABSTRACT

Huntington's disease (HD) is caused by an abnormal CAG expansion in the exon 1 of huntingtin gene. The treatment of HD is an unmet medical need. Given the important role of adenosine in modulating brain activity, in this study, levels of adenosine and adenine nucleotides in the cerebral spinal fluid of patients with HD and in the brain of two mouse models of HD (R6/2 and Hdh150Q) were analysed. The expression and activity of ENT1 in the striatum of mice with HD were measured. Targeting adenosine tone for treating HD was examined in R6/2 mice by genetic removal of ENT1 and by giving an ENT1 inhibitor, respectively. The results showed that the adenosine homeostasis is dysregulated in the brain of patients and mice with HD. In patients, the ratio of adenosine/ATP in the cerebral spinal fluid was negatively correlated with the disease duration, and tended to have a positive correlation with independence scale and functional capacity. In comparison to controls, mRNA level of ENT1 was higher in the striatum of R6/2 and Hdh150Q mice. Intrastriatal administration of ENT1 inhibitors increased extracellular level of adenosine in the striatum of R6/2 mice to a much higher level than controls. Chronic inhibition of ENT1 or by genetic removal of ENT1 enhanced the survival of R6/2 mice. Collectively, adenosine homeostasis and ENT1 expression are altered in HD. The inhibition of ENT1 can enhance extracellular adenosine level and be a potential therapeutic approach for treating HD.


Subject(s)
Adenosine/metabolism , Equilibrative Nucleoside Transporter 1/genetics , Huntingtin Protein/genetics , Huntington Disease/genetics , Adenine/cerebrospinal fluid , Adenine/metabolism , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Adenosine/cerebrospinal fluid , Adenosine/genetics , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , Equilibrative-Nucleoside Transporter 2/genetics , Humans , Huntington Disease/cerebrospinal fluid , Huntington Disease/drug therapy , Huntington Disease/pathology , Indoles/administration & dosage , Mice , Mice, Transgenic , Neostriatum/drug effects , Neostriatum/metabolism , Neostriatum/physiopathology , Trinucleotide Repeat Expansion/genetics
9.
Sci Rep ; 6: 22529, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26932446

ABSTRACT

The calcium-sensitive type VI adenylyl cyclase (AC6) is a membrane-bound adenylyl cyclase (AC) that converts ATP to cAMP under stimulation. It is a calcium-inhibited AC and integrates negative inputs from Ca(2+) and multiple other signals to regulate the intracellular cAMP level. In the present study, we demonstrate that AC6 functions upstream of CREB and negatively controls neuronal plasticity in the hippocampus. Genetic removal of AC6 leads to cyclase-independent and N-terminus of AC6 (AC6N)-dependent elevation of CREB expression, and enhances the expression of GluN2B-containing NMDA receptors in hippocampal neurons. Consequently, GluN2B-dependent calcium signaling and excitatory postsynaptic current, long-term depression, and spatial reversal learning are enhanced in the hippocampus of AC6(-/-) mice without altering the gross anatomy of the brain. Together, our results suggest that AC6 negatively regulates neuronal plasticity by modulating the levels of CREB and GluN2B in the hippocampus.


Subject(s)
Adenylyl Cyclases/metabolism , Learning , Long-Term Synaptic Depression/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Adenylyl Cyclases/genetics , Animals , Hippocampus/enzymology , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
FEBS Lett ; 589(4): 432-9, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25592834

ABSTRACT

Distorted mRNA metabolism contributes to amyotrophic lateral sclerosis (ALS). The human antigen R (HuR) is a major mRNA stabilizer. We report that abnormal localization of HuR was associated with enhanced AMP-activated protein kinase (AMPK) activity in the motor neurons of ALS patients. Activation of AMPK changed the location of HuR in mouse motor neurons and in a motor neuron cell line via phosphorylation of importin-α1. Stimulation of the A2A adenosine receptor normalized the AMPK-evoked redistribution of HuR. This suggests that aberrant activation of AMPK in motor neurons disrupts the normal distribution of HuR, which might imbalance RNA metabolism and contribute to ALS pathogenesis.


Subject(s)
AMP-Activated Protein Kinases/physiology , Amyotrophic Lateral Sclerosis/enzymology , ELAV Proteins/metabolism , Motor Neurons/enzymology , Adult , Aged , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Line , Enzyme Activation , Female , Humans , Male , Mice, Inbred C57BL , Middle Aged , Organ Specificity
11.
Hum Mol Genet ; 24(3): 787-801, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25256353

ABSTRACT

TAR DNA-binding protein-43 (TDP-43) is a nuclear RNA-binding protein involved in many cellular pathways. TDP-43-positive inclusions are a hallmark of amyotrophic lateral sclerosis (ALS). The major clinical presentation of ALS is muscle weakness due to the degeneration of motor neurons. Mislocalization of TDP-43 from the nucleus to the cytoplasm is an early event of ALS. In this study, we demonstrate that cytoplasmic mislocalization of TDP-43 was accompanied by increased activation of AMP-activated protein kinase (AMPK) in motor neurons of ALS patients. The activation of AMPK in a motor neuron cell line (NSC34) or mouse spinal cords induced the mislocalization of TDP-43, recapitulating this characteristic of ALS. Down-regulation of AMPK-α1 or exogenous expression of a dominant-negative AMPK-α1 mutant reduced TDP-43 mislocalization. Suppression of AMPK activity using cAMP-simulating agents rescued the mislocalization of TDP-43 in NSC34 cells and delayed disease progression in TDP-43 transgenic mice. Our findings demonstrate that activation of AMPK-α1 plays a critical role in TDP-43 mislocalization and the development of ALS; thus, AMPK-α1 may be a potential drug target for this devastating disease.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Adult , Aged , Animals , Cell Line , Cell Nucleus/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Motor Neurons/metabolism , Spinal Cord/metabolism
12.
Biochim Biophys Acta ; 1833(12): 3145-3154, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23994616

ABSTRACT

The A2A adenosine receptor (A2AR) is a G-protein-coupled receptor that contains a long cytoplasmic carboxyl terminus (A2AR-C). We report here that Gas-2 like 2 (G2L2) is a new interacting partner of A2AR-C. The interaction between A2AR and G2L2 was verified by GST pull-down, co-immunoprecipitation, immunocytochemical staining, and fluorescence resonance energy transfer. Expression of G2L2 increased the intracellular cAMP content evoked by A2AR in an A2AR-C-dependent manner. Immunoprecipitation and pull-down assays demonstrated that G2L2 selectively bound to A2AR-C and the inactive form of Gαs to facilitate the recruitment of the trimeric G protein complex to the proximal position of A2AR for efficient activation. Collectively, G2L2 is a new effector that controls the action of A2AR by modulating its ability to regulate the Gαs-mediated cAMP contents.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/metabolism , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Receptor, Adenosine A2A/metabolism , Signal Transduction , Animals , Cyclic AMP/metabolism , HEK293 Cells , Humans , Mice , Microfilament Proteins/chemistry , Microtubule-Associated Proteins/chemistry , Models, Biological , Protein Binding , Rats , Rats, Sprague-Dawley
13.
Exp Neurol ; 248: 10-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23726959

ABSTRACT

Visceral functions are regulated by a basal sympathetic nerve discharge (SND), also known as 'sympathetic tone'. We demonstrate herein that AC6 existed in tyrosine hydroxylase-positive rostral ventrolateral medulla neurons in the brainstem. Adenylyl cyclase (AC) assays showed lower basal and pituitary adenylate cyclase-activating peptide-evoked AC activities in the brainstem of AC6-null mice, indicating that AC6 is a prominent AC isozyme in the brainstem. Furthermore, two independent lines of AC6-null mice exhibited a much higher SND, recorded from splanchnic sympathetic nerves of neonatal brainstem-spinal cord preparations, than wildtype mice. An assay of urine noradrenaline confirmed this observation. Collectively, AC6 plays a critical role in the regulation of sympathetic tone.


Subject(s)
Adenylyl Cyclases/metabolism , Brain Stem/metabolism , Spinal Cord/metabolism , Splanchnic Nerves/metabolism , Sympathetic Nervous System/metabolism , Adenylyl Cyclases/genetics , Animals , Animals, Newborn , Mice , Mice, Knockout , Norepinephrine/urine
14.
Mol Cell Biol ; 33(5): 1073-84, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23275441

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disease caused by the expansion of a CAG repeat in the Huntingtin (HTT) gene. Abnormal regulation of the cyclic AMP (cAMP)/protein kinase A (PKA) pathway occurs during HD progression. Here we found that lower PKA activity was associated with proteasome impairment in the striatum for two HD mouse models (R6/2 and N171-82Q) and in mutant HTT (mHTT)-expressing striatal cells. Because PKA regulatory subunits (PKA-Rs) are proteasome substrates, the mHTT-evoked proteasome impairment caused accumulation of PKA-Rs and subsequently inhibited PKA activity. Conversely, activation of PKA enhanced the phosphorylation of Rpt6 (a component of the proteasome), rescued the impaired proteasome activity, and reduced mHTT aggregates. The dominant-negative Rpt6 mutant (Rpt6(S120A)) blocked the ability of a cAMP-elevating reagent to enhance proteasome activity, whereas the phosphomimetic Rpt6 mutant (Rpt6(S120D)) increased proteasome activity, reduced HTT aggregates, and ameliorated motor impairment. Collectively, our data demonstrated that positive feedback regulation between PKA and the proteasome is critical for HD pathogenesis.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Huntington Disease/enzymology , Huntington Disease/physiopathology , Proteasome Endopeptidase Complex/metabolism , Animals , Cell Line , Corpus Striatum/enzymology , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Enzyme Activation , Female , Huntingtin Protein , Huntington Disease/genetics , Male , Mice , Motor Activity , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Rats
15.
Mol Cell Biol ; 31(24): 4874-86, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21986494

ABSTRACT

3'-5'-Cyclic AMP (cAMP) is an important second messenger which regulates neurite outgrowth. We demonstrate here that type VI adenylyl cyclase (AC6), an enzyme which catalyzes cAMP synthesis, regulates neurite outgrowth by direct interaction with a binding protein (Snapin) of Snap25 at the N terminus of AC6 (AC6-N). We first showed that AC6 expression increased during postnatal brain development. In primary hippocampal neurons and Neuro2A cells, elevated AC6 expression suppressed neurite outgrowth, whereas the downregulation or genetic removal of AC6 promoted neurite extension. An AC6 variant (AC6-N5) that contains the N terminus of AC5 had no effect, indicating the importance of AC6-N. The downregulation of endogenous Snapin or the overexpression of a Snapin mutant (Snap(Δ33-51)) that does not bind to AC6, or another Snapin mutant (Snapin(S50A)) that does not interact with Snap25, reversed the inhibitory effect of AC6. Pulldown assays and immunoprecipitation-AC assays revealed that the complex formation of AC6, Snapin, and Snap25 is dependent on AC6-N and the phosphorylation of Snapin. The overexpression of Snap25 completely reversed the action of AC6. Collectively, in addition to cAMP production, AC6 plays a complex role in modulating neurite outgrowth by redistributing localization of the SNARE apparatus via its interaction with Snapin.


Subject(s)
Adenylyl Cyclases/metabolism , Neurites/metabolism , Synaptosomal-Associated Protein 25/metabolism , Vesicular Transport Proteins/metabolism , Adenylyl Cyclases/genetics , Animals , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation, Developmental , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Knockout , Mutation , Nerve Growth Factors/analysis , Phosphorylation , Plasmids , Rats , Rats, Sprague-Dawley , SNARE Proteins/genetics , SNARE Proteins/metabolism , Synaptosomal-Associated Protein 25/genetics , Vesicular Transport Proteins/genetics
16.
PLoS One ; 6(6): e20934, 2011.
Article in English | MEDLINE | ID: mdl-21713039

ABSTRACT

BACKGROUND: Huntington's disease (HD) is a neurodegenerative disease caused by a CAG trinucleotide expansion in the Huntingtin (Htt) gene. The expanded CAG repeats are translated into polyglutamine (polyQ), causing aberrant functions as well as aggregate formation of mutant Htt. Effective treatments for HD are yet to be developed. METHODOLOGY/PRINCIPAL FINDINGS: Here, we report a novel dual-function compound, N(6)-(4-hydroxybenzyl)adenine riboside (designated T1-11) which activates the A(2A)R and a major adenosine transporter (ENT1). T1-11 was originally isolated from a Chinese medicinal herb. Molecular modeling analyses showed that T1-11 binds to the adenosine pockets of the A(2A)R and ENT1. Introduction of T1-11 into the striatum significantly enhanced the level of striatal adenosine as determined by a microdialysis technique, demonstrating that T1-11 inhibited adenosine uptake in vivo. A single intraperitoneal injection of T1-11 in wildtype mice, but not in A(2A)R knockout mice, increased cAMP level in the brain. Thus, T1-11 enters the brain and elevates cAMP via activation of the A(2A)R in vivo. Most importantly, addition of T1-11 (0.05 mg/ml) to the drinking water of a transgenic mouse model of HD (R6/2) ameliorated the progressive deterioration in motor coordination, reduced the formation of striatal Htt aggregates, elevated proteasome activity, and increased the level of an important neurotrophic factor (brain derived neurotrophic factor) in the brain. These results demonstrate the therapeutic potential of T1-11 for treating HD. CONCLUSIONS/SIGNIFICANCE: The dual functions of T1-11 enable T1-11 to effectively activate the adenosinergic system and subsequently delay the progression of HD. This is a novel therapeutic strategy for HD. Similar dual-function drugs aimed at a particular neurotransmitter system as proposed herein may be applicable to other neurotransmitter systems (e.g., the dopamine receptor/dopamine transporter and the serotonin receptor/serotonin transporter) and may facilitate the development of new drugs for other neurodegenerative diseases.


Subject(s)
Adenosine/analogs & derivatives , Drug Design , Huntington Disease/physiopathology , Adenosine/chemistry , Adenosine/metabolism , Adenosine/pharmacology , Adenosine/therapeutic use , Animals , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , Equilibrative Nucleoside Transporter 1/chemistry , Equilibrative Nucleoside Transporter 1/genetics , Equilibrative Nucleoside Transporter 1/metabolism , Female , Humans , Huntington Disease/drug therapy , Male , Mice , Mice, Knockout , Models, Molecular , PC12 Cells/drug effects , Peptides/genetics , Peptides/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Trinucleotide Repeat Expansion
17.
FEBS Lett ; 584(13): 2883-90, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20466003

ABSTRACT

Adenylyl cyclase (AC) type VI (AC6) is a calcium-inhibitable enzyme which produces cAMP upon stimulation. Herein, we characterized the specific role of AC6 in the kidneys using two AC6-knockout mouse lines. Immunohistochemical staining revealed that AC6 exists in the tubular parts of the nephron and collecting duct. Activities of AC evoked by forskolin or a selective agonist of the V2 vasopressin receptor were lower in the kidneys of AC6-null mice compared to those of wildtype mice. Results of a metabolic cage assay and dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) showed for the first time that AC6 plays a critical role in regulating water homeostasis.


Subject(s)
Adenylyl Cyclases/metabolism , Water/metabolism , Adenylyl Cyclases/genetics , Animals , Cyclic AMP/metabolism , Immunohistochemistry , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/physiology , Magnetic Resonance Imaging , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
18.
Hum Mol Genet ; 18(16): 2929-42, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19443488

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by a CAG trinucleotide expansion in the Huntingtin (Htt) gene. The resultant mutant Htt protein (mHtt) forms aggregates in the brain and several peripheral tissues (e.g. the liver) and causes devastating neuronal degeneration. Metabolic defects resulting from Htt aggregates in peripheral tissues also contribute to HD pathogenesis. Simultaneous improvement of defects in both the CNS and peripheral tissues is thus the most effective therapeutic strategy and is highly desirable. We earlier showed that an agonist of the A(2A) adenosine receptor (A(2A) receptor), CGS21680 (CGS), attenuates neuronal symptoms of HD. We found herein that the A(2A) receptor also exists in the liver, and that CGS ameliorated the urea cycle deficiency by reducing mHtt aggregates in the liver. By suppressing aggregate formation, CGS slowed the hijacking of a crucial transcription factor (HSF1) and two protein chaperons (Hsp27 and Hsp70) into hepatic Htt aggregates. Moreover, the abnormally high levels of high-molecular-mass ubiquitin conjugates in the liver of an HD mouse model (R6/2) were also ameliorated by CGS. The protective effect of CGS against mHtt-induced aggregate formation was reproduced in two cells lines and was prevented by an antagonist of the A(2A) receptor and a protein kinase A (PKA) inhibitor. Most importantly, the mHtt-induced suppression of proteasome activity was also normalized by CGS through PKA. Our findings reveal a novel therapeutic pathway of A(2A) receptors in HD and further strengthen the concept that the A(2A) receptor can be a drug target in treating HD.


Subject(s)
Huntington Disease/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptor, Adenosine A2A/metabolism , Ubiquitin/metabolism , Urea/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Antagonists , Animals , Cell Line , Disease Models, Animal , Female , Humans , Huntington Disease/drug therapy , Liver/drug effects , Liver/metabolism , Male , Mice , Phenethylamines/pharmacology , Receptor, Adenosine A2A/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism
19.
J Neurosci ; 28(13): 3277-90, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18367595

ABSTRACT

Huntington's disease (HD) is a hereditary neurological disease caused by expended CAG repeats in the HD gene, which codes for a protein called Huntingtin (Htt). The resultant mutant Huntingtin (mHtt) forms aggregates in neurons and causes neuronal dysfunction. In astrocytes, the largest population of brain cells, mHtt also exists. We report herein that astrocyte-conditioned medium (ACM) collected from astrocytes of R6/2 mice (a mouse model of HD) caused primary cortical neurons to grow less-mature neurites, migrate more slowly, and exhibit lower calcium influx after depolarization than those maintained in wild-type (WT) ACM. Using a cytokine antibody array and ELISA assays, we demonstrated that the amount of a chemokine [chemokine (C-C motif) ligand 5 (CCL5)/regulated on activation normal T cell expressed and secreted (RANTES)] released by R6/2 astrocytes was much less than that by WT astrocytes. When cortical neurons were treated with the indicated ACM, supplementation with recombinant CCL5/RANTES ameliorated the neuronal deficiency caused by HD-ACM, whereas removing CCL5/RANTES from WT-ACM using an anti-CCL5/RANTES antibody mimicked the effects evoked by HD-ACM. Quantitative PCR and promoter analyses demonstrated that mHtt hindered the activation of the CCL5/RANTES promoter by reducing the availability of nuclear factor kappaB-p65 and, hence, reduced the transcript level of CCL5/RANTES. Moreover, ELISA assays and immunocytochemical staining revealed that mHtt retained the residual CCL5/RANTES inside R6/2 astrocytes. In line with the above findings, elevated cytosolic CCL5/RANTES levels were also observed in the brains of two mouse models of HD [R6/2 and Hdh((CAG)150)] and human HD patients. These findings suggest that mHtt hinders one major trophic function of astrocytes which might contribute to the neuronal dysfunction of HD.


Subject(s)
Astrocytes/metabolism , Chemokine CCL5/metabolism , Nerve Tissue Proteins/physiology , Neurons/metabolism , Nuclear Proteins/physiology , Trinucleotide Repeat Expansion/physiology , Aged , Aged, 80 and over , Animals , Animals, Newborn , Astrocytes/chemistry , Brain/pathology , Calcium/metabolism , Cell Movement/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Chromatin Immunoprecipitation/methods , Culture Media, Conditioned/pharmacology , Embryo, Mammalian , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/drug effects , Nuclear Proteins/genetics , Rats , Rats, Sprague-Dawley , Transfection/methods , Trinucleotide Repeat Expansion/genetics
20.
Biochem J ; 406(3): 383-8, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17593019

ABSTRACT

In the present study, we demonstrate that AC5 (type V adenylate cyclase) interacts with Ric8a through directly interacting at its N-terminus. Ric8a was shown to be a GEF (guanine nucleotide exchange factor) for several alpha subunits of heterotrimeric GTP binding proteins (Galpha proteins) in vitro. Selective Galpha targets of Ric8a have not yet been revealed in vivo. An interaction between AC5 and Ric8a was verified by pull-down assays, co-immunoprecipitation analyses, and co-localization in the brain. Expression of Ric8a selectively suppressed AC5 activity. Treating cells with pertussis toxin or expressing a dominant negative Galphai mutant abolished the suppressive effect of Ric8a, suggesting that interaction between the N-terminus of AC5 and a GEF (Ric8a) provides a novel pathway to fine-tune AC5 activity via a Galphai-mediated pathway.


Subject(s)
Adenylyl Cyclases/metabolism , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/metabolism , Isoenzymes/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/immunology , Animals , Blotting, Western , Cyclic AMP/metabolism , Electrophoresis, Polyacrylamide Gel , Genes, Dominant , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Humans , Immunoglobulin G/immunology , Immunoprecipitation , Isoenzymes/genetics , Isoenzymes/immunology , Kidney/metabolism , Pertussis Toxin/pharmacology , Protein Binding , Rabbits , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...