Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Physiol Rep ; 5(5)2017 Mar.
Article in English | MEDLINE | ID: mdl-28292879

ABSTRACT

Treatments that increase basal metabolic rate (BMR) and enhance exercise capacity may be useful therapeutic approaches for treating conditions such as type 2 diabetes, obesity, and associated circulatory problems. ß-guanidinopropionic acid (ß-GPA) supplementation decreases high-energy phosphate concentrations, such as ATP and phosphocreatine (PCr) resulting in an energetic challenge that is similar to both exercise programs and hypoxic conditions. In this study, we administered ß-GPA to mice for 2 or 6 weeks, and investigated the effect on muscle energetic status, body and muscle mass, muscle capillarity, BMR, and normoxic and hypoxic exercise tolerance (NET and HET, respectively). Relative [PCr] and PCr/ATP ratios significantly decreased during both treatment times in the ß-GPA fed mice compared to control mice. Body mass, muscle mass, and muscle fiber size significantly decreased after ß-GPA treatment, whereas muscle capillarity and BMR were significantly increased in ß-GPA fed mice. NET significantly decreased in the 2-week treatment, but was not significantly different in the 6-week treatment. HET significantly decreased in 2-week treatment, but in contrast to NET, significantly increased in the 6-week-treated mice compared to control mice. We conclude that ß-GPA induces a cellular energetic response in skeletal muscle similar to that of chronic environmental hypoxia, and this energetic perturbation leads to elevated BMR and increased hypoxic exercise capacity in the absence of hypoxic acclimation.


Subject(s)
Basal Metabolism/drug effects , Exercise Tolerance/drug effects , Guanidines/pharmacology , Physical Conditioning, Animal , Propionates/pharmacology , Animals , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism
2.
Sci Rep ; 5: 18395, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26671672

ABSTRACT

Mutations in protein kinase C substrate 80K-H (PRKCSH), which encodes for an 80 KDa protein named hepatocystin (80K-H, PRKCSH), gives rise to polycystic liver disease (PCLD). Hepatocystin functions as the noncatalytic beta subunit of Glucosidase II, an endoplasmic reticulum (ER)-resident enzyme involved in processing and quality control of newly synthesized glycoproteins. Patients harboring heterozygous germline mutations in PRKCSH are thought to develop renal cysts as a result of somatic loss of the second allele, which subsequently interferes with expression of the TRP channel polycystin-2 (PKD2). Deletion of both alleles of PRKCSH in mice results in embryonic lethality before embryonic day E11.5. Here, we investigated the function of hepatocystin during Xenopus laevis embryogenesis and identified hepatocystin as a binding partner of the TRPM7 ion channel, whose function is required for vertebrate gastrulation. We find that TRPM7 functions synergistically with hepatocystin. Although other N-glycosylated proteins are critical to early development, overexpression of TRPM7 in Xenopus laevis embryos was sufficient to fully rescue the gastrulation defect caused by loss of hepatocystin. We observed that depletion of hepatocystin in Xenopus laevis embryos decreased TRPM7 expression, indicating that the early embryonic lethality caused by loss of hepatocystin is mainly due to impairment of TRPM7 protein expression.


Subject(s)
Embryo, Nonmammalian/embryology , Embryonic Development/physiology , Gastrula/embryology , Glucosidases/metabolism , TRPM Cation Channels/metabolism , Xenopus Proteins/metabolism , Animals , Cell Line , Glucosidases/genetics , Humans , Mice , TRPM Cation Channels/genetics , Xenopus Proteins/genetics , Xenopus laevis
3.
Dev Biol ; 386(2): 428-39, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24291744

ABSTRACT

Transient receptor potential, melastatin-like 7 (Trpm7) is a combined ion channel and kinase implicated in the differentiation or function of many cell types. Early lethality in mice and frogs depleted of the corresponding gene impedes investigation of the functions of this protein particularly during later stages of development. By contrast, zebrafish trpm7 mutant larvae undergo early morphogenesis normally and thus do not have this limitation. The mutant larvae are characterized by multiple defects including melanocyte cell death, transient paralysis, and an ion imbalance that leads to the development of kidney stones. Here we report a requirement for Trpm7 in differentiation or function of dopaminergic neurons in vivo. First, trpm7 mutant larvae are hypomotile and fail to make a dopamine-dependent developmental transition in swim-bout length. Both of these deficits are partially rescued by the application of levodopa or dopamine. Second, histological analysis reveals that in trpm7 mutants a significant fraction of dopaminergic neurons lack expression of tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis. Third, trpm7 mutants are unusually sensitive to the neurotoxin 1-methyl-4-phenylpyridinium, an oxidative stressor, and their motility is partially rescued by application of the iron chelator deferoxamine, an anti-oxidant. Finally, in SH-SY5Y cells, which model aspects of human dopaminergic neurons, forced expression of a channel-dead variant of TRPM7 causes cell death. In summary, a forward genetic screen in zebrafish has revealed that both melanocytes and dopaminergic neurons depend on the ion channel Trpm7. The mechanistic underpinning of this dependence requires further investigation.


Subject(s)
Cell Differentiation/physiology , Dopaminergic Neurons/cytology , Motor Activity/genetics , Protein Serine-Threonine Kinases/genetics , TRPM Cation Channels/genetics , Zebrafish Proteins/genetics , Zebrafish/growth & development , 1-Methyl-4-phenylpyridinium/toxicity , Analysis of Variance , Animals , Cell Line , DNA Primers/genetics , Deferoxamine/pharmacology , Electroretinography , Larva/growth & development , Melanocytes/metabolism , Motor Activity/drug effects , Motor Activity/physiology , Mutation/genetics , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Tyrosine 3-Monooxygenase/metabolism , Zebrafish/genetics
4.
Biochem J ; 445(3): 441-8, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22587440

ABSTRACT

The TRPM7 (transient receptor potential melastatin 7) channel has been shown to play a pivotal role in cell survival during brain ischaemia as well as in the survival of other cell types challenged with apoptotic stimuli. Ca(2+) is thought to be central to the channel's ability to regulate ROS (reactive oxygen species) production. However, channel-mediated entry of Mg(2+) and Zn(2+) have also been implicated in cell death. In the present study, we show that depletion of TRPM7 by RNA interference in fibroblasts increases cell resistance to apoptotic stimuli by decreasing ROS levels in an Mg(2+)-dependent manner. Depletion of TRPM7 lowered cellular Mg(2+), decreased the concentration of ROS and lessened p38 MAPK (mitogen-activated protein kinase) and JNK (c-Jun N-terminal kinase) activation as well as decreased caspase 3 activation and PARP [poly(ADP-ribose) polymerase] cleavage in response to apoptotic stimuli. Re-expression of TRPM7 or of a kinase-inactive mutant of TRPM7 in TRPM7-knockdown cells increased cellular Mg(2+) and ROS levels, as did expression of the Mg(2+) transporter SLC41A2 (solute carrier family 41 member 2). In addition, expression of SLC41A2 increased the sensitivity of TRPM7-knockdown cells to apoptotic stimuli and boosted ROS generation in response to cell stress. Taken together, these data uncover an essential role for Mg(2+) in TRPM7's control of cell survival and in the regulation of cellular ROS levels.


Subject(s)
Magnesium/metabolism , TRPM Cation Channels/metabolism , Animals , Apoptosis , Cation Transport Proteins/metabolism , Cell Survival , Gene Knockdown Techniques , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Oxidative Stress , RNA Interference , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Swiss 3T3 Cells , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Sci Rep ; 1: 146, 2011.
Article in English | MEDLINE | ID: mdl-22180838

ABSTRACT

TRPM6 is crucial for human Mg2+ homeostasis as patients carrying TRPM6 mutations develop hypomagnesemia and secondary hypocalcemia (HSH). However, the activation mechanism of TRPM6 has remained unknown. Here we demonstrate that phosphatidylinositol-4,5-bisphophate (PIP2) controls TRPM6 activation and Mg2+ influx. Stimulation of PLC-coupled M1-receptors to deplete PIP2 potently inactivates TRPM6. Translocation of over-expressed 5-phosphatase to cell membrane to specifically hydrolyze PIP2 also completely inhibits TRPM6. Moreover, depolarization-induced-activation of the voltage-sensitive-phosphatase (Ci-VSP) simultaneously depletes PIP2 and inhibits TRPM6. PLC-activation induced PIP2-depletion not only inhibits TRPM6, but also abolishes TRPM6-mediated Mg2+ influx.Furthermore, neutralization of basic residues in the TRP domain leads to nonfunctional or dysfunctional mutants with reduced activity by PIP2, suggesting that they are likely to participate in interactions with PIP2.Our data indicate that PIP2 is required for TRPM6 channel function; hydrolysis of PIP2 by PLC-coupled hormones/agonists may constitute an important pathway for TRPM6 gating, and perhaps Mg2+ homeostasis.


Subject(s)
Magnesium/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , TRPM Cation Channels/metabolism , Amino Acid Sequence , Binding Sites , Cell Line , HEK293 Cells , Homeostasis , Humans , Ion Channel Gating , Kinetics , Molecular Sequence Data , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics , Type C Phospholipases/metabolism
6.
J Mol Biol ; 396(4): 858-69, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20070945

ABSTRACT

TRPM7 is a Ca(2)(+)-permeant and Mg(2)(+)-permeant ion channel in possession of its own kinase domain. In a previous study, we showed that overexpression of the channel-kinase in HEK-293 cells produced cell rounding and loss of adhesion, which was dependent on the Ca(2+)-dependent protease m-calpain. The TRPM7-elicited change in cell morphology was channel-dependent and occurred without any significant increase in cytosolic Ca(2+). Here we demonstrate that overexpression of TRPM7 increased levels of cellular reactive oxygen species (ROS) and nitric oxide, causing the activation of p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK). Application of inhibitors of p38 MAPK and JNK blocked TRPM7-induced cell rounding and activation of m-calpain, without affecting the phosphorylation state of the protease. Overexpression of TRPM7 increased intracellular Mg(2+); however, when the concentration of either external Ca(2+) or Mg(2+) was increased to favor the permeation of one divalent cation over the other, a similar increase in cell rounding and calpain activity was detected, indicating that TRPM7-mediated activation of m-calpain is not dependent on the nature of the divalent conducted by the channel. Application of inhibitors of nitric oxide synthase and mitochondrial-derived ROS reduced TRPM7-induced increases in nitric oxide and ROS production, blocked the change in cell morphology, and reduced cellular calpain activity. Collectively, our data reveal that excessive TRPM7 channel activity causes oxidative and nitrosative stresses, producing cell rounding mediated by p38 MAPK/JNK-dependent activation of m-calpain.


Subject(s)
Calpain/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , TRPM Cation Channels/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Calcium/metabolism , Cell Adhesion , Cell Line , Cell Shape , Feedback, Physiological , Gene Expression , Humans , MAP Kinase Signaling System , Mice , Nitric Oxide/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , TRPM Cation Channels/genetics
7.
Article in English | MEDLINE | ID: mdl-19100334

ABSTRACT

The effect of chronic hypobaric hypoxia (1/2 atmospheric pressure) on high energy phosphate (HEP) compounds was investigated in slow (soleus; SOL) and fast twitch (extensor digitorum longus; EDL) muscle from 3 strains of mice with large differences in hypoxic exercise tolerance (HET). Phosphocreatine concentration ([PCr]) decreased 16-29% following hypoxia in EDL and SOL in all strains, while [ADP] and [AMP] increased. In the EDL, HET was negatively correlated with the PCr/ATP ratio and positively correlated with the ATP/P(i) ratio. The free energy of ATP hydrolysis (DeltaG(obs)) remained constant despite the substantial changes that occurred in HEP profiles. The alteration of HEP set points and preservation of DeltaG(obs) are consistent with the notion that (1) maximal rates of steady-state ATP turnover are reduced under hypoxia, and (2) HEP perturbations during rest to work transitions are reduced in skeletal muscle from hypoxia acclimated animals. We therefore expected a lower phosphorylation ratio of AMP-activated protein kinase (AMPK-P/AMPK) during stimulation in hypoxic acclimated animals. However, neither the resting nor stimulated AMPK-P/AMPK was influenced by hypoxia, although there were significant differences among strains.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Energy Metabolism , Hypoxia/metabolism , Muscle, Skeletal/metabolism , Phosphates/metabolism , Physical Exertion/physiology , Animals , Body Mass Index , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...