Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(7)2021 Mar 27.
Article in English | MEDLINE | ID: mdl-33801629

ABSTRACT

The Na/K-ATPase is the specific receptor for cardiotonic steroids (CTS) such as ouabain and digoxin. At pharmacological concentrations used in the treatment of cardiac conditions, CTS inhibit the ion-pumping function of Na/K-ATPase. At much lower concentrations, in the range of those reported for endogenous CTS in the blood, they stimulate hypertrophic growth of cultured cardiac myocytes through initiation of a Na/K-ATPase-mediated and reactive oxygen species (ROS)-dependent signaling. To examine a possible effect of endogenous concentrations of CTS on cardiac structure and function in vivo, we compared mice expressing the naturally resistant Na/K-ATPase α1 and age-matched mice genetically engineered to express a mutated Na/K-ATPase α1 with high affinity for CTS. In this model, total cardiac Na/K-ATPase activity, α1, α2, and ß1 protein content remained unchanged, and the cardiac Na/K-ATPase dose-response curve to ouabain shifted to the left as expected. In males aged 3-6 months, increased α1 sensitivity to CTS resulted in a significant increase in cardiac carbonylated protein content, suggesting that ROS production was elevated. A moderate but significant increase of about 15% of the heart-weight-to-tibia-length ratio accompanied by an increase in the myocyte cross-sectional area was detected. Echocardiographic analyses did not reveal any change in cardiac function, and there was no fibrosis or re-expression of the fetal gene program. RNA sequencing analysis indicated that pathways related to energy metabolism were upregulated, while those related to extracellular matrix organization were downregulated. Consistent with a functional role of the latter, an angiotensin-II challenge that triggered fibrosis in the α1r/rα2s/s mouse failed to do so in the α1s/sα2s/s. Taken together, these results are indicative of a link between circulating CTS, Na/K-ATPase α1, ROS, and physiological cardiac hypertrophy in mice under baseline laboratory conditions.


Subject(s)
Cardiac Glycosides/chemistry , Heart/physiology , Myocardium/enzymology , Sodium-Potassium-Exchanging ATPase/genetics , Angiotensin II/pharmacology , Animals , Cardiomegaly/pathology , Disease Models, Animal , Echocardiography , Heart/drug effects , Male , Mice , Mutation , Ouabain/pharmacology , Protein Isoforms , RNA-Seq , Reactive Oxygen Species , Signal Transduction/drug effects
2.
iScience ; 17: 334-346, 2019 Jul 26.
Article in English | MEDLINE | ID: mdl-31326700

ABSTRACT

Skeletal muscle repair and regeneration after injury requires coordinated interactions between the innate immune system and the injured muscle. Myeloid cells predominate in these interactions. This study examined the role of KLF2, a zinc-finger transcription factor that regulates immune cell activation, in specifying myeloid cell functions during muscle regeneration. Loss of KLF2 in myeloid lineage cells (myeKlf2-/- mice) dramatically enhanced the initial inflammatory response to acute muscle injury (cardiotoxin). Injured muscles showed dramatically elevated expression of inflammatory mediators and greater numbers of infiltrating, pro-inflammatory monocytes that matured earlier into activated macrophages. Notably, the inflammatory phase resolved earlier and regeneration progressed to myogenesis, marked by elevated expression of factors that promote the formation of new fibers from satellite cells. Regeneration was completed earlier, with phenotypically normal adult fibers integrated into the muscle syncytium. These findings identify myeloid KLF2 as a key regulator of myeloid cell functions in adult skeletal muscle regeneration.

3.
Int J Mol Sci ; 19(9)2018 Sep 12.
Article in English | MEDLINE | ID: mdl-30213059

ABSTRACT

The potassium affinities of Na,K-ATPase isozymes are important determinants of their physiological roles in skeletal muscle. This study measured the apparent K⁺ and Rb⁺ affinities of the Na,K-ATPase α1 and α2 isozymes in intact, dissociated myofibers obtained from WT and genetically altered mice (α1S/Sα2R/R and skα2-/-). It also validates a new method to quantify cations in intact, dissociated myofibers, using inductively coupled plasma mass spectrometry (ICP-MS). Our findings were that: (1) The extracellular substrate sites of Na,K-ATPase bind Rb⁺ and K⁺ with comparable apparent affinities; however; turnover rate is reduced when Rb⁺ is the transported ion; (2) The rate of Rb⁺ uptake by the Na,K-ATPase is not constant but declines with a half-time of approximately 1.5 min; (3) The apparent K⁺ affinity of the α2 isozymes for K⁺ is significantly lower than α1. When measured in intact fibers of WT and α1S/Sα2R/R mice in the presence of 10 µM ouabain; the K1/2,K of α1 and α2 isozymes are 1.3 and 4 mM, respectively. Collectively, these results validate the single fiber model for studies of Na,K-ATPase transport and kinetic constants, and they imply the existence of mechanisms that dynamically limit pump activity during periods of active transport.


Subject(s)
Isoenzymes/metabolism , Potassium/metabolism , Rubidium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Biological Transport , Kinetics , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Sodium/metabolism
4.
Am J Physiol Endocrinol Metab ; 314(6): E620-E629, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29438630

ABSTRACT

The distribution of Na/K-ATPase α-isoforms in skeletal muscle is unique, with α1 as the minor (15%) isoform and α2 comprising the bulk of the Na/K-ATPase pool. The acute and isoform-specific role of α2 in muscle performance and resistance to fatigue is well known, but the isoform-specific role of α1 has not been as thoroughly investigated. In vitro, we reported that α1 has a role in promoting cell growth that is not supported by α2. To assess whether α1 serves this isoform-specific trophic role in the skeletal muscle, we used Na/K-ATPase α1-haploinsufficient (α1+/-) mice. A 30% decrease of Na/K-ATPase α1 protein expression without change in α2 induced a modest yet significant decrease of 10% weight in the oxidative soleus muscle. In contrast, the mixed plantaris and glycolytic extensor digitorum longus weights were not significantly affected, likely because of their very low expression level of α1 compared with the soleus. The soleus mass reduction occurred without change in total Na/K-ATPase activity or glycogen metabolism. Serum analytes including K+, fat tissue mass, and exercise capacity were not altered in α1+/- mice. The impact of α1 content on soleus muscle mass is consistent with a Na/K-ATPase α1-specific role in skeletal muscle growth that cannot be fulfilled by α2. The preserved running capacity in α1+/- is in sharp contrast with previously reported consequences of genetic manipulation of α2. Taken together, these results lend further support to the concept of distinct isoform-specific functions of Na/K-ATPase α1 and α2 in skeletal muscle.


Subject(s)
Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Sodium-Potassium-Exchanging ATPase/physiology , Animals , Gene Expression Regulation, Enzymologic , Isoenzymes/genetics , Isoenzymes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Contraction/physiology , Muscle, Skeletal/pathology , Organ Size/genetics , Physical Conditioning, Animal , Sodium-Potassium-Exchanging ATPase/genetics
5.
mBio ; 7(3)2016 06 14.
Article in English | MEDLINE | ID: mdl-27302755

ABSTRACT

UNLABELLED: The adaptive immune response is tightly regulated by complex signals in dendritic cells (DCs). Although Th2 polarization is dictated by defined functional DC subsets, the molecular factors that govern the amplitude of these responses are not well understood. Krüppel-like factor 2 (KLF2) is a transcription factor that negatively regulates the activation of numerous immune cells in response to stimuli. Here, we demonstrate that suppression of KLF2 in conditioned DCs preferentially amplifies Th2 responses in two model systems, one of which is a prototypical intracellular pathogen and the other an allergen. This elevation in Th2 responses was dependent on contact-mediated Notch signaling in vitro and in vivo A deficiency of KLF2 increased the expression of Notch ligand Jagged2 via hypoxia-inducible factor 1α (HIF-1α), which led to Th2 amplification. Our results revealed a novel circuit in DCs for Th2 polarization that is governed by KLF2. IMPORTANCE: Dendritic cells are the key element that bridges innate and adaptive immunity. A complex and not-well-understood area in dendritic cell biology is the regulatory network that predetermines or moderates their function to shape the adaptive immune response. Our study for the first time demonstrates that KLF2, a transcription factor, conditions dendritic cells to regulate Th2 responses via a Jagged2/Notch axis. Downregulation of KLF2 expression in dendritic cells may provide a beneficial effect for treatment of diseases such as obesity or parasitic infections but may be deleterious in the case of invasion by intracellular pathogens. Strategies to tune KLF2 may be useful for future therapeutic approaches to particular diseases of mankind.


Subject(s)
Dendritic Cells/immunology , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Jagged-2 Protein/metabolism , Kruppel-Like Transcription Factors/metabolism , Receptors, Notch/metabolism , Th2 Cells/immunology , Animals , Mice, Inbred C57BL , Transcription, Genetic
6.
J Physiol ; 594(21): 6079-6103, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27350568

ABSTRACT

Reduced smooth muscle (SM)-specific α2 Na+ pump expression elevates basal blood pressure (BP) and increases BP sensitivity to angiotensin II (Ang II) and dietary NaCl, whilst SM-α2 overexpression lowers basal BP and decreases Ang II/salt sensitivity. Prolonged ouabain infusion induces hypertension in rodents, and ouabain-resistant mutation of the α2 ouabain binding site (α2R/R mice) confers resistance to several forms of hypertension. Pressure overload-induced heart hypertrophy and failure are attenuated in cardio-specific α2 knockout, cardio-specific α2 overexpression and α2R/R mice. We propose a unifying hypothesis that reconciles these apparently disparate findings: brain mechanisms, activated by Ang II and high NaCl, regulate sympathetic drive and a novel neurohumoral pathway mediated by both brain and circulating endogenous ouabain (EO). Circulating EO modulates ouabain-sensitive α2 Na+ pump activity and Ca2+ transporter expression and, via Na+ /Ca2+ exchange, Ca2+ homeostasis. This regulates sensitivity to sympathetic activity, Ca2+ signalling and arterial and cardiac contraction.


Subject(s)
Cardiovascular System/metabolism , Hypertension/metabolism , Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Angiotensins/metabolism , Animals , Binding Sites , Cardiotonic Agents/pharmacology , Cardiovascular System/drug effects , Humans , Hypertension/physiopathology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/chemistry , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiology
7.
Am J Physiol Cell Physiol ; 309(12): C813-22, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26468207

ABSTRACT

The Na(+)-K(+)-ATPase α2-isoform in skeletal muscle is rapidly stimulated during muscle use and plays a critical role in fatigue resistance. The acute mechanisms that stimulate α2-activity are not completely known. This study examines whether phosphorylation of phospholemman (PLM/FXYD1), a regulatory subunit of Na(+)-K(+)-ATPase, plays a role in the acute stimulation of α2 in working muscles. Mice lacking PLM (PLM KO) have a normal content of the α2-subunit and show normal exercise capacity, in contrast to the greatly reduced exercise capacity of mice that lack α2 in the skeletal muscles. Nerve-evoked contractions in vivo did not induce a change in total PLM or PLM phosphorylated at Ser63 or Ser68, in either WT or PLM KO. Isolated muscles of PLM KO mice maintain contraction and resist fatigue as well as wild type (WT). Rb(+) transport by the α2-Na(+)-K(+)-ATPase is stimulated to the same extent in contracting WT and contracting PLM KO muscles. Phosphorylation of sarcolemmal membranes prepared from WT but not PLM KO skeletal muscles stimulates the activity of both α1 and α2 in a PLM-dependent manner. The stimulation occurs by an increase in Na(+) affinity without significant change in Vmax and is more effective for α1 than α2. These results demonstrate that phosphorylation of PLM is capable of stimulating the activity of both isozymes in skeletal muscle; however, contractile activity alone is not sufficient to induce PLM phosphorylation. Importantly, acute stimulation of α2, sufficient to support exercise and oppose fatigue, does not require PLM or its phosphorylation.


Subject(s)
Membrane Proteins/metabolism , Muscle Fatigue/physiology , Muscle, Skeletal/metabolism , Phosphoproteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Blotting, Western , Electric Stimulation , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction/physiology , Phosphorylation , Physical Conditioning, Animal/physiology , Spectrophotometry, Atomic
8.
J Gen Physiol ; 146(4): 281-94, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26371210

ABSTRACT

The Na,K-ATPase α2 isoform is the predominant Na,K-ATPase in adult skeletal muscle and the sole Na,K-ATPase in the transverse tubules (T-tubules). In quiescent muscles, the α2 isozyme operates substantially below its maximal transport capacity. Unlike the α1 isoform, the α2 isoform is not required for maintaining resting ion gradients or the resting membrane potential, canonical roles of the Na,K-ATPase in most other cells. However, α2 activity is stimulated immediately upon the start of contraction and, in working muscles, its contribution is crucial to maintaining excitation and resisting fatigue. Here, we show that α2 activity is determined in part by the K+ concentration in the T-tubules, through its K+ substrate affinity. Apparent K+ affinity was determined from measurements of the K1/2 for K+ activation of pump current in intact, voltage-clamped mouse flexor digitorum brevis muscle fibers. Pump current generated by the α2 Na,K-ATPase, Ip, was identified as the outward current activated by K+ and inhibited by micromolar ouabain. Ip was outward at all potentials studied (-90 to -30 mV) and increased with depolarization in the subthreshold range, -90 to -50 mV. The Q10 was 2.1 over the range of 22-37°C. The K1/2,K of Ip was 4.3±0.3 mM at -90 mV and was relatively voltage independent. This K+ affinity is lower than that reported for other cell types but closely matches the dynamic range of extracellular K+ concentrations in the T-tubules. During muscle contraction, T-tubule luminal K+ increases in proportion to the frequency and duration of action potential firing. This K1/2,K predicts a low fractional occupancy of K+ substrate sites at the resting extracellular K+ concentration, with occupancy increasing in proportion to the frequency of membrane excitation. The stimulation of preexisting pumps by greater K+ site occupancy thus provides a rapid mechanism for increasing α2 activity in working muscles.


Subject(s)
Muscle Fibers, Skeletal/metabolism , Potassium/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Action Potentials , Animals , Cells, Cultured , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Contraction , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/physiology
9.
J Physiol ; 593(6): 1361-82, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25772291

ABSTRACT

This paper is the third in a series of reviews published in this issue resulting from the University of California Davis Cardiovascular Symposium 2014: Systems approach to understanding cardiac excitation-contraction coupling and arrhythmias: Na(+) channel and Na(+) transport. The goal of the symposium was to bring together experts in the field to discuss points of consensus and controversy on the topic of sodium in the heart. The present review focuses on cardiac Na(+)/Ca(2+) exchange (NCX) and Na(+)/K(+)-ATPase (NKA). While the relevance of Ca(2+) homeostasis in cardiac function has been extensively investigated, the role of Na(+) regulation in shaping heart function is often overlooked. Small changes in the cytoplasmic Na(+) content have multiple effects on the heart by influencing intracellular Ca(2+) and pH levels thereby modulating heart contractility. Therefore it is essential for heart cells to maintain Na(+) homeostasis. Among the proteins that accomplish this task are the Na(+)/Ca(2+) exchanger (NCX) and the Na(+)/K(+) pump (NKA). By transporting three Na(+) ions into the cytoplasm in exchange for one Ca(2+) moved out, NCX is one of the main Na(+) influx mechanisms in cardiomyocytes. Acting in the opposite direction, NKA moves Na(+) ions from the cytoplasm to the extracellular space against their gradient by utilizing the energy released from ATP hydrolysis. A fine balance between these two processes controls the net amount of intracellular Na(+) and aberrations in either of these two systems can have a large impact on cardiac contractility. Due to the relevant role of these two proteins in Na(+) homeostasis, the emphasis of this review is on recent developments regarding the cardiac Na(+)/Ca(2+) exchanger (NCX1) and Na(+)/K(+) pump and the controversies that still persist in the field.


Subject(s)
Action Potentials , Arrhythmias, Cardiac/metabolism , Myocytes, Cardiac/metabolism , Sodium-Calcium Exchanger/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Congresses as Topic , Humans , Myocytes, Cardiac/physiology
10.
Nat Neurosci ; 17(12): 1710-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25344630

ABSTRACT

Perturbations of astrocytes trigger neurodegeneration in several diseases, but the glial cell-intrinsic mechanisms that induce neurodegeneration remain poorly understood. We found that a protein complex of α2-Na/K ATPase and α-adducin was enriched in astrocytes expressing mutant superoxide dismutase 1 (SOD1), which causes familial amyotrophic lateral sclerosis (ALS). Knockdown of α2-Na/K ATPase or α-adducin in mutant SOD1 astrocytes protected motor neurons from degeneration, including in mutant SOD1 mice in vivo. Heterozygous disruption of the α2-Na/K ATPase gene suppressed degeneration in vivo and increased the lifespan of mutant SOD1 mice. The pharmacological agent digoxin, which inhibits Na/K ATPase activity, protected motor neurons from mutant SOD1 astrocyte-induced degeneration. Notably, α2-Na/K ATPase and α-adducin were upregulated in spinal cord of sporadic and familial ALS patients. Collectively, our findings define chronic activation of the α2-Na/K ATPase/α-adducin complex as a critical glial cell-intrinsic mechanism of non-cell autonomous neurodegeneration, with implications for potential therapies for neurodegenerative diseases.


Subject(s)
Astrocytes/metabolism , Astrocytes/pathology , Cytoskeletal Proteins/biosynthesis , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Sodium-Potassium-Exchanging ATPase/biosynthesis , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
11.
J Biol Chem ; 289(45): 31638-46, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25248747

ABSTRACT

Previous studies have shown that the myeloid-specific deficiency of the transcription factor Krüppel-like factor 2 (KLF2) accelerates atherosclerosis in hypercholesterolemic Ldlr(-/-) mice due to the enhanced adhesion of myeloid cells to activated endothelial cells in the vessel wall. This study revealed elevated basal inflammation with elevated plasma levels of Ccl2, Ccl4, Ccl5, and Ccl11 in the myeloid-specific KLF2 knock-out (myeKlf2(-/-)) mice. Peritoneal macrophages isolated from myeKlf2(-/-) mice showed increased mRNA levels of several inflammatory mediators, including Ccl2, Ccl5, Ccl7, Cox-2, Cxcl1, and IL-6. In contrast, the levels of two microRNAs, miR-124a and miR-150, were lower in Klf2(-/-) macrophages compared with Klf2(+/+) macrophages. Additional studies showed a direct inverse relationship between miR-124a levels with Ccl2 expression, with anti-miR-124a increasing Ccl2 mRNA levels in Klf2(+/+) macrophages, whereas the restoration of miR-124a levels in Klf2(-/-) macrophages significantly reduced Ccl2 mRNA expression. Likewise, the inverse relationship was observed between miR-150 levels and Cxcl1 expression in Klf2(+/+) and Klf2(-/-) mice. Moreover, miR150 likely regulates the miR124a expression and thus augments expression of inflammatory mediators in myeKlf2(-/-) macrophages. This study documented that the transcription factor KLF2 modulates inflammatory chemokine production via regulation of microRNA expression levels in immune cells.


Subject(s)
Gene Expression Regulation , Kruppel-Like Transcription Factors/genetics , Macrophages, Peritoneal/metabolism , MicroRNAs/blood , Animals , Atherosclerosis/genetics , Base Sequence , Binding Sites , Chemokines/metabolism , Female , Inflammation , Kruppel-Like Transcription Factors/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Myeloid Cells/cytology , Myeloid Cells/metabolism
12.
Cell Rep ; 4(3): 405-12, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911285

ABSTRACT

Angelman syndrome (AS) is associated with symptoms that include autism, intellectual disability, motor abnormalities, and epilepsy. We recently showed that AS model mice have increased expression of the alpha1 subunit of Na/K-ATPase (α1-NaKA) in the hippocampus, which was correlated with increased expression of axon initial segment (AIS) proteins. Our developmental analysis revealed that the increase in α1-NaKA expression preceded that of the AIS proteins. Therefore, we hypothesized that α1-NaKA overexpression drives AIS abnormalities and that by reducing its expression these and other phenotypes could be corrected in AS model mice. Herein, we report that the genetic normalization of α1-NaKA levels in AS model mice corrects multiple hippocampal phenotypes, including alterations in the AIS, aberrant intrinsic membrane properties, impaired synaptic plasticity, and memory deficits. These findings strongly suggest that increased expression of α1-NaKA plays an important role in a broad range of abnormalities in the hippocampus of AS model mice.


Subject(s)
Angelman Syndrome/genetics , Angelman Syndrome/pathology , Hippocampus/metabolism , Hippocampus/pathology , Sodium-Potassium-Exchanging ATPase/genetics , Angelman Syndrome/enzymology , Angelman Syndrome/metabolism , Animals , Ankyrins/biosynthesis , Disease Models, Animal , Female , Hippocampus/enzymology , Male , Mice , Mice, Inbred C57BL , NAV1.6 Voltage-Gated Sodium Channel/biosynthesis , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , Protein Subunits , Sodium-Potassium-Exchanging ATPase/metabolism
13.
Am J Physiol Heart Circ Physiol ; 304(8): H1147-58, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23436327

ABSTRACT

The α2-isoform of the Na,K-ATPase (α2) is the minor isoform of the Na,K-ATPase expressed in the cardiovascular system and is thought to play a critical role in the regulation of cardiovascular hemodynamics. However, the organ system/cell type expressing α2 that is required for this regulation has not been fully defined. The present study uses a heart-specific knockout of α2 to further define the tissue-specific role of α2 in the regulation of cardiovascular hemodynamics. To accomplish this, we developed a mouse model using the Cre/loxP system to generate a tissue-specific knockout of α2 in the heart using ß-myosin heavy chain Cre. We have achieved a 90% knockout of α2 expression in the heart of the knockout mice. Interestingly, the heart-specific knockout mice exhibit normal basal cardiac function and systolic blood pressure, and in addition, these mice develop ACTH-induced hypertension in response to ACTH treatment similar to control mice. Surprisingly, the heart-specific knockout mice display delayed onset of cardiac dysfunction compared with control mice in response to pressure overload induced by transverse aortic constriction; however, the heart-specific knockout mice deteriorated to control levels by 9 wk post-transverse aortic constriction. These results suggest that heart expression of α2 does not play a role in the regulation of basal cardiovascular function or blood pressure; however, heart expression of α2 plays a role in the hypertrophic response to pressure overload. This study further emphasizes that the tissue localization of α2 determines its unique roles in the regulation of cardiovascular function.


Subject(s)
Adrenocorticotropic Hormone/adverse effects , Hypertension/metabolism , Hypertrophy, Left Ventricular/metabolism , Myocytes, Cardiac/physiology , Sodium-Potassium-Exchanging ATPase/physiology , Ventricular Dysfunction, Left/metabolism , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Blood Pressure/genetics , Blood Pressure/physiology , Gene Knockout Techniques/methods , Hypertension/chemically induced , Hypertension/genetics , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/genetics , Integrases , Mice , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/analysis , Sodium-Potassium-Exchanging ATPase/genetics , Ultrasonography , Vasoconstriction , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/genetics
14.
J Biol Chem ; 288(2): 1226-37, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23192345

ABSTRACT

The Na,K-ATPase α2 isozyme is the major Na,K-ATPase of mammalian skeletal muscle. This distribution is unique compared with most other cells, which express mainly the Na,K-ATPase α1 isoform, but its functional significance is not known. We developed a gene-targeted mouse (skα2(-/-)) in which the α2 gene (Atp1a2) is knocked out in the skeletal muscles, and examined the consequences for exercise performance, membrane potentials, contractility, and muscle fatigue. Targeted knockout was confirmed by genotyping, Western blot, and immunohistochemistry. Skeletal muscle cells of skα2(-/-) mice completely lack α2 protein and have no α2 in the transverse tubules, where its expression is normally enhanced. The α1 isoform, which is normally enhanced on the outer sarcolemma, is up-regulated 2.5-fold without change in subcellular targeting. skα2(-/-) mice are apparently normal under basal conditions but show significantly reduced exercise capacity when challenged to run. Their skeletal muscles produce less force, are unable to increase force to match demand, and show significantly increased susceptibility to fatigue. The impairments affect both fast and slow muscle types. The subcellular targeting of α2 to the transverse tubules is important for this role. Increasing Na,K-ATPase α1 content cannot fully compensate for the loss of α2. The increased fatigability of skα2(-/-) muscles is reproduced in control extensor digitorum longus muscles by selectively inhibiting α2 enzyme activity with ouabain. These results demonstrate that the Na,K-ATPase α2 isoform performs an acute, isoform-specific role in skeletal muscle. Its activity is regulated by muscle use and enables working muscles to maintain contraction and resist fatigue.


Subject(s)
Isoenzymes/metabolism , Muscle, Skeletal/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Base Sequence , Blotting, Western , DNA Primers , Immunohistochemistry , Mice , Mice, Knockout , Muscle Contraction , Muscle, Skeletal/physiology , Polymerase Chain Reaction
15.
Int J Bipolar Disord ; 1(1): 33, 2013 Dec.
Article in English | MEDLINE | ID: mdl-26054600

ABSTRACT

BACKGROUND: Glial dysfunction has been purported to be important to the pathophysiology of bipolar illness. However, manic behavior has not been previously demonstrated to result as a consequence of glial pathology. The aim of the current study was to assess the behaviors of the glial-specific sodium pump alpha2 subunit (ATP1A2) knockout (KO) heterozygote mice to determine if a glial-specific abnormality can produce manic-like behavior. METHODS: Activity and behavior of hemideficient sodium pump alpha2 KO mice and wild-type (WT) littermates (C57BL6/Black Swiss background) were examined at baseline, following forced swimming stress and restraint stress and after 3 days of sleep deprivation. RESULTS AND DISCUSSION: At baseline, the 24-h total distance traveled and center time were significantly greater in KO mice, but there were no behavioral differences with sweet water preference or with inactivity time during forced swim or tail suspension tests. After restraint stress or forced swimming stress, there were no differences in activity. Three days of sleep deprivation utilizing the inverted flowerpot method induced a significant increase in the distance traveled by the KO versus WT mice in the 30-min observation period (p=0.016). Lithium pretreatment has no effect on WT animals versus their baseline but significantly reduces hyperactivity induced by sleep deprivation in KO. Knockout of the glial-specific alpha2 isoform is associated with some manic behaviors compared to WT littermates, suggesting that glial dysfunction could be associated with mania.

16.
Cardiovasc Res ; 95(4): 480-6, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22739122

ABSTRACT

AIMS: Na(+)/K(+)-ATPase (NKA) is essential in regulating [Na(+)](i), and thus cardiac myocyte Ca(2+) and contractility via Na(+)/Ca(2+) exchange. Different NKA-α subunit isoforms are present in the heart and may differ functionally, depending on specific membrane localization. In smooth muscle and astrocytes, NKA-α2 is located at the junctions with the endo(sarco)plasmic reticulum, where they could regulate local [Na(+)], and indirectly junctional cleft [Ca(2+)]. Whether this model holds for cardiac myocytes is unclear. METHODS AND RESULTS: The ouabain-resistant NKA-α1 cannot be selectively blocked to assess its effect. To overcome this, we used mice in which NKA-α1 is ouabain sensitive and NKA-α2 is ouabain resistant (SWAP mice). We measured the effect of ouabain at low concentration on [Na(+)](i), Ca(2+) transients, and the fractional sarcoplasmic reticulum (SR) Ca(2+) release in cardiac myocytes from wild-type (WT; NKA-α2 inhibition) and SWAP mice (selective NKA-α1 block). At baseline, Na(+) and Ca(2+) regulations are similar in WT and SWAP mice. For equal levels of total NKA inhibition (~25%), ouabain significantly increased Ca(2+) transients (from ΔF/F(0)= 1.5 ± 0.1 to 1.8 ± 0.1), and fractional SR Ca(2+) release (from 24 ± 3 to 29 ± 3%) in WT (NKA-α2 block) but not in SWAP myocytes (NKA-α1 block). This occurred despite a similar and modest increase in [Na(+)](i) (~2 mM) in both groups. The effect in WT mice was mediated specifically by NKA-α2 inhibition because at a similar concentration ouabain had no effect in transgenic mice where both NKA-α1 and NKA-α2 are ouabain resistant. CONCLUSION: NKA-α2 has a more prominent role (vs. NKA-α1) in modulating cardiac myocyte SR Ca(2+) release.


Subject(s)
Calcium Signaling , Myocytes, Cardiac/enzymology , Sarcoplasmic Reticulum/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Isoenzymes , Membrane Potentials , Mice , Mice, Transgenic , Mutation , Myocytes, Cardiac/drug effects , Ouabain/pharmacology , Sarcoplasmic Reticulum/drug effects , Sodium/metabolism , Sodium-Calcium Exchanger/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics
17.
Circ Res ; 110(10): 1294-302, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22474254

ABSTRACT

RATIONALE: Hemizygous deficiency of the transcription factor Krüppel-like factor 2 (KLF2) has been shown previously to augment atherosclerosis in hypercholesterolemic mice. However, the cell type responsible for the increased atherosclerosis due to KLF2 deficiency has not been identified. This study examined the consequence of myeloid cell-specific KLF2 inactivation in atherosclerosis. METHODS AND RESULTS: Cell-specific knockout mice were generated by Cre/loxP recombination. Macrophages isolated from myeloid-specific Klf2 knockout (myeKlf2(-/-)) mice were similar to myeKlf2(+/+) macrophages in response to activation, polarization, and lipid accumulation. However, in comparison to myeKlf2(+/+) macrophages, myeKlf2(-/-) macrophages adhered more robustly to endothelial cells. Neutrophils from myeKlf2(-/-) mice also adhered more robustly to endothelial cells, and fewer myeKlf2(-/-) neutrophils survived in culture over a 24-hour period in comparison with myeKlf2(+/+) neutrophils. When myeKlf2(-/-) mice were mated to Ldlr(-/-) mice and then fed a high fat and high cholesterol diet, significant increase in atherosclerosis was observed in the myeKlf2(-/-)Ldlr(-/-) mice compared with myeKlf2(+/+)Ldlr(-/-) littermates. The increased atherosclerosis in myeKlf2(-/-)Ldlr(-/-) mice was associated with elevated presence of neutrophils and macrophages, with corresponding increase of myeloperoxidase as well as chlorinated and nitrosylated tyrosine epitopes in their lesion areas compared with myeKlf2(+/+)Ldlr(-/-) mice. CONCLUSIONS: This study documents a role for myeloid KLF2 expression in modulating atherosclerosis. The increased neutrophil accumulation and atherosclerosis progression with myeloid-specific KLF2 deficiency also underscores the importance of neutrophils in promoting vascular oxidative stress and atherosclerosis. Collectively, these results suggest that elevating KLF2 expression may be a novel strategy for prevention and treatment of atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Cell Adhesion/immunology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Macrophages/immunology , Neutrophils/immunology , Animals , Atherosclerosis/pathology , Cell Death/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Female , Hypercholesterolemia/immunology , Hypercholesterolemia/pathology , Lymphocyte Count , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Vasculitis/immunology , Vasculitis/pathology
18.
J Biol Chem ; 287(20): 16390-8, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22451662

ABSTRACT

Decreases in cardiac Na/K-ATPase have been documented in patients with heart failure. Reduction of Na/K-ATPase α1 also contributes to the deficiency in cardiac contractility in animal models. Our previous studies demonstrate that reduction of cellular Na/K-ATPase causes cell growth inhibition and cell death in renal proximal tubule cells. To test whether reduction of Na/K-ATPase in combination with increased cardiotonic steroids causes cardiac myocyte death and cardiac dysfunction, we examined heart function in Na/K-ATPase α1 heterozygote knock-out mice (α1(+/-)) in comparison to wild type (WT) littermates after infusion of marinobufagenin (MBG). Adult cardiac myocytes were also isolated from both WT and α1(+/-) mice for in vitro experiments. The results demonstrated that MBG infusion increased myocyte apoptosis and induced significant left ventricle dilation in α1(+/-) mice but not in their WT littermates. Mechanistically, it was found that in WT myocytes MBG activated the Src/Akt/mTOR signaling pathway, which further increased phosphorylation of ribosome S6 kinase (S6K) and BAD (Bcl-2-associated death promoter) and protected cells from apoptosis. In α1(+/-) myocytes, the basal level of phospho-BAD is higher compared with WT myocytes, but MBG failed to induce further activation of the mTOR pathway. Reduction of Na/K-ATPase also caused the activation of caspase 9 but not caspase 8 in these cells. Using cultures of neonatal cardiac myocytes, we demonstrated that inhibition of the mTOR pathway by rapamycin also enabled MBG to activate caspase 9 and induce myocyte apoptosis.


Subject(s)
Apoptosis/drug effects , Bufanolides/adverse effects , Enzyme Inhibitors/adverse effects , Heart Diseases/enzymology , Muscle Proteins/metabolism , Myocytes, Cardiac/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/genetics , Bufanolides/pharmacology , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Heart Diseases/chemically induced , Heart Diseases/genetics , Mice , Mice, Mutant Strains , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
19.
Am J Hypertens ; 25(4): 421-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22258333

ABSTRACT

BACKGROUND: We have shown that the ouabain-sensitive α2 Na,K-ATPase is required for adrenocorticotropic hormone (ACTH)-induced hypertension and gestational blood pressure regulation. It is therefore of interest to explore whether this binding site participates in the development of other forms of hypertension, such as deoxycorticosterone acetate (DOCA)-salt using mutant mice with altered sensitivity to ouabain. METHODS: Wild-type (α1 ouabain-resistant, α2 ouabain-sensitive: α(R/R)α2(S/S)), α1-resistant, α2-resistant (α1(R/R)α2(R/R)) and α1-sensitive, α2-resistant (α1(S/S)α2(R/R)) mice were uninephrectomized and implanted with DOCA pellets. The animals were given either tap water or 1% NaCl, and blood pressure was measured before and after DOCA. RESULTS: DOCA-salt-treated α1(R/R)α2(R/R) mice developed hypertension to the same extent as α1(R/R)α2(S/S) mice (wild type), and the α1(S/S)α2(R/R) mice given DOCA-salt also showed no difference from the other two genotypes. The expression of the α1 isoform was not changed by DOCA-salt treatment in either α1(R/R)α2(S/S) or α1(R/R)α2(R/R) mice. However, the α2 subunit was expressed at substantially higher levels in the hearts of α1(R/R)α2(R/R) than α1(R/R)α2(S/S) mice, regardless of treatment. Plasma levels of ouabain did not change consistently, but those of marinobufagenin were modestly higher in DOCA-salt treated mice relatively to those without salt. CONCLUSIONS: The ouabain-binding site of either the α1 or α2 Na,K-ATPase subunit does not play an essential role in the development of DOCA-salt hypertension in this mouse model. These findings indicate that the underlying mechanisms of hypertension induced by DOCA-salt treatment are different from those of ACTH-induced hypertension.


Subject(s)
Hypertension/chemically induced , Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase/physiology , Animals , Binding Sites/physiology , Blood Pressure/drug effects , Bufanolides/blood , Desoxycorticosterone , Digoxin/immunology , Hypertension/physiopathology , Immunoglobulin Fab Fragments , Mice , Myocardial Contraction/drug effects , Sodium Chloride , Sodium-Potassium-Exchanging ATPase/biosynthesis , Sodium-Potassium-Exchanging ATPase/genetics
20.
J Biol Chem ; 287(2): 1448-57, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22110137

ABSTRACT

Although gram-positive infections account for the majority of cases of sepsis, the molecular mechanisms underlying their effects remains poorly understood. We investigated how cell wall components of gram-positive bacteria contribute to the development of sepsis. Experimental observations derived from cultured primary macrophages and the cell line indicate that gram-positive bacterial endotoxins induce hypoxia-inducible factor 1α (HIF-1α) mRNA and protein expression. Inoculation of live or heat-inactivated gram-positive bacteria with macrophages induced HIF-1 transcriptional activity in macrophages. Concordant with these results, myeloid deficiency of HIF-1α attenuated gram-positive bacterial endotoxin-induced cellular motility and proinflammatory gene expression in macrophages. Conversely, gram-positive bacteria and their endotoxins reduced expression of the myeloid anti-inflammatory transcription factor Krüppel-like transcription factor 2 (KLF2). Sustained expression of KLF2 reduced and deficiency of KLF2 enhanced gram-positive endotoxins induced HIF-1α mRNA and protein expression in macrophages. More importantly, KLF2 attenuated gram-positive endotoxins induced cellular motility and proinflammatory gene expression in myeloid cells. Consistent with these results, mice deficient in myeloid HIF-1α were protected from gram-positive endotoxin-induced sepsis mortality and clinical symptomatology. By contrast, myeloid KLF2-deficient mice were susceptible to gram-positive sepsis induced mortality and clinical symptoms. Collectively, these observations identify HIF-1α and KLF2 as critical regulators of gram-positive endotoxin-mediated sepsis.


Subject(s)
Endotoxins/toxicity , Gram-Positive Bacteria , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kruppel-Like Transcription Factors/metabolism , Macrophages/metabolism , Shock, Septic/metabolism , Animals , Cell Line , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kruppel-Like Transcription Factors/genetics , Macrophages/pathology , Mice , Mice, Transgenic , Shock, Septic/chemically induced , Shock, Septic/genetics , Shock, Septic/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...