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










Publication year range
1.
Ann Am Thorac Soc ; 14(Supplement_6): S415-S420, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29048937

ABSTRACT

Sarcoidosis is a systemic granulomatous disease that primarily affects the lung; it is associated with significant disparities, more commonly impacting those in the prime of their lives (age 20-50 yr, with a second peak after age 60 yr), black individuals, and women. However, the burden of disease, the ability to diagnose and prognose organ involvement and course, as well as specific treatment options, management options, and disease pathogenesis remain poorly understood. As a result, the National Heart, Lung, and Blood Institute undertook a sarcoidosis workshop, "Leveraging Current Scientific Advancements to Understand Sarcoidosis Variability and Improve Outcomes," to help address these issues by defining the scientific and clinical priorities to improve sarcoidosis care. The overarching recommendations from this workshop are outlined in the following summary and detailed in the accompanying articles. The recommendations included establishing collaborations and networks to conduct research based on consensus definitions of disease phenotypes and standards of care, and to provide clinical outreach to areas with a burden of disease to improve care. These collaborative networks would also serve as the hub to conduct clinical trials of devastating phenotypes (e.g., cardiac, neurologic, and fibrotic disease) not only for treatment but to enhance our understanding of the burden of disease. In addition, the networks would be used to leverage state-of-the-art "omics" and systems biology research, as well as other studies to advance understanding of disease pathogenesis, and development of biomarkers and therapeutic targets, with a goal to translate this information to improve care of individuals with sarcoidosis.


Subject(s)
Biomedical Research/trends , Sarcoidosis/diagnosis , Sarcoidosis/therapy , Humans , National Heart, Lung, and Blood Institute (U.S.) , United States
2.
JCI Insight ; 2(9)2017 May 04.
Article in English | MEDLINE | ID: mdl-28469087

ABSTRACT

Lung transplantation, a cure for a number of end-stage lung diseases, continues to have the worst long-term outcomes when compared with other solid organ transplants. Preclinical modeling of the most common and serious lung transplantation complications are essential to better understand and mitigate the pathophysiological processes that lead to these complications. Various animal and in vitro models of lung transplant complications now exist and each of these models has unique strengths. However, significant issues, such as the required technical expertise as well as the robustness and clinical usefulness of these models, remain to be overcome or clarified. The National Heart, Lung, and Blood Institute (NHLBI) convened a workshop in March 2016 to review the state of preclinical science addressing the three most important complications of lung transplantation: primary graft dysfunction (PGD), acute rejection (AR), and chronic lung allograft dysfunction (CLAD). In addition, the participants of the workshop were tasked to make consensus recommendations on the best use of these complimentary models to close our knowledge gaps in PGD, AR, and CLAD. Their reviews and recommendations are summarized in this report. Furthermore, the participants outlined opportunities to collaborate and directions to accelerate research using these preclinical models.

4.
Am J Respir Crit Care Med ; 189(2): 214-22, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24160862

ABSTRACT

The median survival of patients with idiopathic pulmonary fibrosis (IPF) continues to be approximately 3 years from the time of diagnosis, underscoring the lack of effective medical therapies for this disease. In the United States alone, approximately 40,000 patients die of this disease annually. In November 2012, the NHLBI held a workshop aimed at coordinating research efforts and accelerating the development of IPF therapies. Basic, translational, and clinical researchers gathered with representatives from the NHLBI, patient advocacy groups, pharmaceutical companies, and the U.S. Food and Drug Administration to review the current state of IPF research and identify priority areas, opportunities for collaborations, and directions for future research. The workshop was organized into groups that were tasked with assessing and making recommendations to promote progress in one of the following six critical areas of research: (1) biology of alveolar epithelial injury and aberrant repair; (2) role of extracellular matrix; (3) preclinical modeling; (4) role of inflammation and immunity; (5) genetic, epigenetic, and environmental determinants; (6) translation of discoveries into diagnostics and therapeutics. The workshop recommendations provide a basis for directing future research and strategic planning by scientific, professional, and patient communities and the NHLBI.


Subject(s)
Idiopathic Pulmonary Fibrosis , Animals , Biomedical Research/trends , Disease Models, Animal , Extracellular Matrix/pathology , Genetic Predisposition to Disease , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/physiopathology , Idiopathic Pulmonary Fibrosis/therapy , Inflammation/immunology , Mice , Pulmonary Alveoli/pathology , Respiratory Mucosa/pathology
5.
J Biol Chem ; 287(39): 32755-69, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22851173

ABSTRACT

GRP78, a molecular chaperone with critical endoplasmic reticulum functions, is aberrantly expressed on the surface of cancer cells, including prostate and melanoma. Here it functions as a pro-proliferative and anti-apoptotic signaling receptor via NH(2)-terminal domain ligation. Auto-antibodies to this domain may appear in cancer patient serum where they are a poor prognostic indicator. Conversely, GRP78 COOH-terminal domain ligation is pro-apoptotic and anti-proliferative. There is no method to disrupt cell-surface GRP78 without compromising the total GRP78 pool, making it difficult to study cell-surface GRP78 function. We studied six cell lines representing three cancer types. One cell line per group expresses high levels of cell-surface GRP78, and the other expresses low levels (human hepatoma: Hep3B and HepG2; human prostate cancer: PC3 and 1-LN; murine melanoma: B16F0 and B16F1). We investigated the effect of Escherichia coli subtilase cytoxin catalytic subunit (SubA) on GRP78. We report that SubA specifically cleaves cell-surface GRP78 on HepG2, 1-LN, and B16F1 cells without affecting intracellular GRP78. B16F0 cells (GRP78(low)) have lower amounts of cleaved cell-surface GRP78. SubA has no effect on Hep3B and PC3 cells. The predicted 28-kDa GRP78 COOH-terminal fragment is released into the culture medium by SubA treatment, and COOH-terminal domain signal transduction is abrogated, whereas pro-proliferative signaling mediated through NH(2)-terminal domain ligation is unaffected. These experiments clarify cell-surface GRP78 topology and demonstrate that the COOH-terminal domain is necessary for pro-apoptotic signal transduction occurring upon COOH-terminal antibody ligation. SubA is a powerful tool to specifically probe the functions of cell-surface GRP78.


Subject(s)
Escherichia coli Proteins/pharmacology , Escherichia coli/enzymology , Heat-Shock Proteins/metabolism , Melanoma/metabolism , Prostatic Neoplasms/metabolism , Proteolysis/drug effects , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Subtilisins/pharmacology , Animals , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/metabolism , Autoantibodies/genetics , Autoantibodies/metabolism , Catalytic Domain , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Hep G2 Cells , Humans , Male , Melanoma/genetics , Mice , Prostatic Neoplasms/genetics , Receptors, G-Protein-Coupled/genetics
6.
Methods Mol Biol ; 798: 373-82, 2012.
Article in English | MEDLINE | ID: mdl-22130848

ABSTRACT

The ryanodine receptor ion channels (RyRs) release Ca(2+) from the endo/sarcoplasmic reticulum in a variety of nonvertebrate and vertebrate species including flies, crustaceans, birds, fish, and amphibians. They are most abundant in skeletal and cardiac muscle, where in response to an action potential, the release of Ca(2+) ions from the sarcoplasmic reticulum through the RyRs into the cytoplasm leads to muscle contraction (i.e., excitation-contraction coupling). Here, we describe how to determine their cellular location using isoform-specific antibodies, their protein levels using an in vitro ((3)H)ryanodine-binding assay, and their cellular release of Ca(2+) using RyR-specific channel agonists and inhibitors.


Subject(s)
Calcium/metabolism , Ryanodine Receptor Calcium Release Channel/analysis , Animals , HEK293 Cells , Humans , Immunohistochemistry , Mice , Muscles/metabolism , Protein Binding/physiology , Protein Isoforms/analysis , Protein Isoforms/immunology , Protein Transport , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/immunology , Ryanodine Receptor Calcium Release Channel/metabolism
7.
Proc Natl Acad Sci U S A ; 108(38): 16098-103, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21896730

ABSTRACT

Physiological sensing of O(2) tension (partial O(2) pressure, pO(2)) plays an important role in some mammalian cellular systems, but striated muscle generally is not considered to be among them. Here we describe a molecular mechanism in skeletal muscle that acutely couples changes in pO(2) to altered calcium release through the ryanodine receptor-Ca(2+)-release channel (RyR1). Reactive oxygen species are generated in proportion to pO(2) by NADPH oxidase 4 (Nox4) in the sarcoplasmic reticulum, and the consequent oxidation of a small set of RyR1 cysteine thiols results in increased RyR1 activity and Ca(2+) release in isolated sarcoplasmic reticulum and in cultured myofibers and enhanced contractility of intact muscle. Thus, Nox4 is an O(2) sensor in skeletal muscle, and O(2)-coupled hydrogen peroxide production by Nox4 governs the redox state of regulatory RyR1 thiols and thereby governs muscle performance. These findings reveal a molecular mechanism for O(2)-based signaling by an NADPH oxidase and demonstrate a physiological role for oxidative modification of RyR1.


Subject(s)
Muscle, Skeletal/metabolism , NADPH Oxidases/metabolism , Oxygen/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cell Line , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Hydrogen Peroxide/metabolism , Mice , Muscle Contraction/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Myoblasts/cytology , Myoblasts/metabolism , NADP/pharmacology , NADPH Oxidase 4 , NADPH Oxidases/genetics , Oxidation-Reduction , RNA Interference , Rabbits , Rats , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sulfhydryl Compounds/metabolism
8.
Crit Care Med ; 39(11): 2478-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21765360

ABSTRACT

OBJECTIVE: Transfusion of red blood cells has been linked to disappointing clinical outcomes in the critically ill, but specific mechanisms of organ dysfunction after transfusion remain poorly understood. We tested the hypothesis that red blood cell storage impairs the ability of red blood cells to release adenosine-5'-triphosphate and that impaired adenosine-5'-triphosphate release was injurious in vivo, in part through increased red blood cell adhesion. DESIGN: Prospective, controlled, mechanistic study. SETTING: University research laboratory. SUBJECTS: Human and mouse blood donors; nude mouse transfusion recipients. INTERVENTIONS: Manipulation of adenosine-5'-triphosphate release, supplemental adenosine-5'-triphosphate, and antibodies to red blood cell and endothelial adhesion receptors were used in vitro and in vivo to probe the roles of released adenosine-5'-triphosphate and adhesion in responses to (transfused) red blood cells. MEASUREMENTS AND MAIN RESULTS: The ability of stored red blood cells to release adenosine-5'-triphosphate declined markedly within 14 days after collection despite relatively stable levels of adenosine-5'-triphosphate within the red blood cells. Inhibiting adenosine-5'-triphosphate release promoted the adhesion of stored red blood cells to endothelial cells in vitro and red blood cell sequestration in the lungs of transfused mice in vivo. Unlike transfusion of fresh human red blood cells, stored red blood cell transfusion in mice decreased blood oxygenation and increased extravasation of red blood cells into the lung's alveolar air spaces. Similar findings were seen with transfusion of fresh red blood cells treated with the adenosine-5'-triphosphate release inhibitors glibenclamide and carbenoxolone. These findings were prevented by either coinfusion of an adenosine-5'-triphosphate analog or pretransfusion incubation of the red blood cells with an antibody against the erythrocyte adhesion receptor Landsteiner-Wiener (intercellular adhesion molecule-4). CONCLUSIONS: The normal flow of red blood cells in pulmonary microvessels depends in part on the release of antiadhesive adenosine-5'-triphosphate from red blood cells, and storage-induced deficiency in adenosine-5'-triphosphate release from transfused red blood cells may promote or exacerbate microvascular pathophysiology in the lung, in part through increased red blood cell adhesion.


Subject(s)
Adenosine Triphosphate/biosynthesis , Blood Transfusion , Endothelial Cells/physiology , Erythrocytes/physiology , Animals , Apyrase/pharmacology , Blood Preservation , Carbenoxolone/pharmacology , Cell Adhesion/drug effects , Cell Hypoxia , Endothelial Cells/metabolism , Erythrocytes/metabolism , Extravasation of Diagnostic and Therapeutic Materials , Glyburide/pharmacology , Humans , Lung/blood supply , Lung/cytology , Mice , Mice, Nude , Time Factors
9.
Environ Health Perspect ; 119(10): 1403-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21684833

ABSTRACT

BACKGROUND: Our previous work demonstrated that the extracellular matrix protein mindin contributes to allergic airways disease. However, the role of mindin in nonallergic airways disease has not previously been explored. OBJECTIVES: We hypothesized that mindin would contribute to airways disease after inhalation of either lipopolysaccharide (LPS) or ozone. METHODS: We exposed C57BL/6J and mindin-deficient (-/-) mice to aerosolized LPS (0.9 µg/m3 for 2.5 hr), saline, ozone (1 ppm for 3 hr), or filtered air (FA). All mice were evaluated 4 hr after LPS/saline exposure or 24 hr after ozone/FA exposure. We characterized the physiological and biological responses by analysis of airway hyperresponsiveness (AHR) with a computer-controlled small-animal ventilator (FlexiVent), inflammatory cellular recruitment, total protein in bronchoalveolar lavage fluid (BALF), proinflammatory cytokine profiling, and ex vivo bronchial ring studies. RESULTS: After inhalation of LPS, mindin-/- mice demonstrated significantly reduced total cell and neutrophil recruitment into the airspace compared with their wild-type counterparts. Mindin-/- mice also exhibited reduced proinflammatory cytokine production and lower AHR to methacholine challenge by FlexiVent. After inhalation of ozone, mice had no detectible differences in cellular inflammation or total BALF protein dependent on mindin. However, mindin-/- mice were protected from increased proinflammatory cytokine production and AHR compared with their C57BL/6J counterparts. After ozone exposure, bronchial rings derived from mindin-/- mice demonstrated reduced constriction in response to carbachol. CONCLUSIONS: These data demonstrate that the extracellular matrix protein mindin modifies the airway response to both LPS and ozone. Our data support a conserved role of mindin in production of proinflammatory cytokines and the development of AHR in two divergent models of reactive airways disease, as well as a role of mindin in airway smooth muscle contractility after exposure to ozone.


Subject(s)
Extracellular Matrix Proteins/metabolism , Animals , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/immunology , Extracellular Matrix Proteins/genetics , Immunity, Innate , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neutrophil Infiltration/drug effects , Ozone/toxicity , Toll-Like Receptor 4/metabolism
10.
Am J Physiol Cell Physiol ; 300(5): C998-C1012, 2011 May.
Article in English | MEDLINE | ID: mdl-21289290

ABSTRACT

In vitro, calmodulin (CaM) and S100A1 activate the skeletal muscle ryanodine receptor ion channel (RyR1) at submicromolar Ca(2+) concentrations, whereas at micromolar Ca(2+) concentrations, CaM inhibits RyR1. One amino acid substitution (RyR1-L3625D) has previously been demonstrated to impair CaM binding and regulation of RyR1. Here we show that the RyR1-L3625D substitution also abolishes S100A1 binding. To determine the physiological relevance of these findings, mutant mice were generated with the RyR1-L3625D substitution in exon 74, which encodes the CaM and S100A1 binding domain of RyR1. Homozygous mutant mice (Ryr1(D/D)) were viable and appeared normal. However, single RyR1 channel recordings from Ryr1(D/D) mice exhibited impaired activation by CaM and S100A1 and impaired CaCaM inhibition. Isolated flexor digitorum brevis muscle fibers from Ryr1(D/D) mice had depressed Ca(2+) transients when stimulated by a single action potential. However, during repetitive stimulation, the mutant fibers demonstrated greater relative summation of the Ca(2+) transients. Consistently, in vivo stimulation of tibialis anterior muscles in Ryr1(D/D) mice demonstrated reduced twitch force in response to a single action potential, but greater summation of force during high-frequency stimulation. During repetitive stimulation, Ryr1(D/D) fibers exhibited slowed inactivation of sarcoplasmic reticulum Ca(2+) release flux, consistent with increased summation of the Ca(2+) transient and contractile force. Peak Ca(2+) release flux was suppressed at all voltages in voltage-clamped Ryr1(D/D) fibers. The results suggest that the RyR1-L3625D mutation removes both an early activating effect of S100A1 and CaM and delayed suppressing effect of CaCaM on RyR1 Ca(2+) release, providing new insights into CaM and S100A1 regulation of skeletal muscle excitation-contraction coupling.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , S100 Proteins/metabolism , Sarcoplasmic Reticulum/metabolism , Action Potentials/physiology , Animals , Calcium/physiology , Calmodulin/physiology , Female , Male , Mice , Muscle Contraction/physiology , Muscle Strength/physiology , Muscle, Skeletal/physiology , Protein Binding , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/physiology , S100 Proteins/physiology , Sarcoplasmic Reticulum/physiology
11.
Am J Respir Cell Mol Biol ; 44(2): 175-84, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20348208

ABSTRACT

Surfactant protein A (SP-A) mediates innate immune cell responses to LPS, a cell wall component of gram-negative bacteria that is found ubiquitously in the environment and is associated with adverse health effects. Inhaled LPS induces lung inflammation and increases airway responsiveness (AR). However, the role of SP-A in mediating LPS-induced AR is not well-defined. Nitric oxide (NO) is described as a potent bronchodilator, and previous studies showed that SP-A modulates the LPS-induced production of NO. Hence, we tested the hypothesis that increased AR, observed in response to aerosolized LPS exposure, would be significantly reduced in an SP-A-deficient condition. Wild-type (WT) and SP-A null (SP-A(-/-)) mice were challenged with aerosolized LPS. Results indicate that despite similar inflammatory indices, LPS-treated SP-A(-/-) mice had attenuated AR after methacholine challenge, compared with WT mice. The attenuated AR could not be attributed to inherent differences in SP-D concentrations or airway smooth muscle contractile and relaxation properties, because these measures were similar between WT and SP-A(-/-) mice. LPS-treated SP-A(-/-) mice, however, had elevated nitrite concentrations, inducible nitric oxide synthase (iNOS) expression, and NOS activity in their lungs. Moreover, the administration of the iNOS-specific inhibitor 1400W completely abrogated the attenuated AR. Thus, when exposed to aerosolized LPS, SP-A(-/-) mice demonstrate a relative airway hyporesponsiveness that appears to be mediated at least partly via an iNOS-dependent mechanism. These findings may have clinical significance, because recent studies reported associations between surfactant protein polymorphisms and a variety of lung diseases.


Subject(s)
Lipopolysaccharides/pharmacology , Lung/immunology , Lung/physiopathology , Nitric Oxide/physiology , Pulmonary Surfactant-Associated Protein A/deficiency , Animals , Immunity, Innate , Lung/drug effects , Methacholine Chloride/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/immunology , Pulmonary Surfactant-Associated Protein A/physiology , Pulmonary Surfactant-Associated Protein D/metabolism
13.
Cell Calcium ; 45(1): 29-37, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18620751

ABSTRACT

Triadin and junctin are integral sarcoplasmic reticulum membrane proteins that form a macromolecular complex with the skeletal muscle ryanodine receptor (RyR1) but their roles in skeletal muscle calcium homeostasis remain incompletely understood. Here we report that delivery of siRNAs specific for triadin or junctin into C2C12 skeletal myoblasts reduced the expression of triadin and junctin in 8-day-old myotubes by 80 and 100%, respectively. Knocking down either triadin or junctin in these cells reduced Ca2+ release induced by depolarization (10mM KCl) by 20-25%. Unlike triadin knockdown myotubes, junctin knockdown and junctin/triadin double knockdown myotubes also had reduced Ca2+ release induced by 400 microM 4-chloro-m-cresol, 10mM caffeine, 400 microM UTP, or 1 microM thapsigargin. Thus, knocking down junctin compromised the Ca2+ stores in the sarcoplasmic reticulum of these cells. Our subsequent studies showed that in junctin knockdown myotubes at least two sarcoplasmic reticulum proteins (RyR1 and skeletal muscle calsequestrin) were down-regulated while these proteins' mRNA expression was not affected. The results suggest that triadin has a role in facilitating KCl depolarization-induced Ca2+ release in contrast to junctin which has a role in maintaining sarcoplasmic reticulum Ca2+ store size in C2C12 myotubes.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Mixed Function Oxygenases/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Myoblasts, Skeletal/metabolism , Animals , Calcium Signaling/drug effects , Calcium Signaling/genetics , Calcium-Binding Proteins/genetics , Calsequestrin/metabolism , Carrier Proteins/genetics , Cell Line , Feedback, Physiological , Membrane Proteins/genetics , Mice , Mixed Function Oxygenases/genetics , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/genetics , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Neuromuscular Depolarizing Agents/pharmacology , RNA, Small Interfering/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Transduction, Genetic
14.
Biochemistry ; 47(52): 13985-90, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19053230

ABSTRACT

The cardiac and skeletal muscle sarcoplasmic reticulum ryanodine receptor Ca(2+) release channels contain thiols that are potential targets of endogenously produced reactive oxygen and nitrogen intermediates. Previously, we showed that the skeletal muscle ryanodine receptor (RyR1) has O(2)-sensitive thiols; only when these thiols are in the reduced state (pO(2) approximately 10 mmHg) can physiological concentrations of NO (nanomolar) activate RyR1. Here, we report that cardiac muscle ryanodine receptor (RyR2) activity also depends on pO(2), but unlike RyR1, RyR2 was not activated or S-nitrosylated directly by NO. Rather, activation and S-nitrosylation of RyR2 required S-nitrosoglutathione. The effects of peroxynitrite were indiscriminate on RyR1 and RyR2. Our results indicate that both RyR1 and RyR2 are pO(2)-responsive yet point to different mechanisms by which NO and S-nitrosoglutathione influence cardiac and skeletal muscle sarcoplasmic reticulum Ca(2+) release.


Subject(s)
Myocardium/chemistry , Oxygen/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , S-Nitrosoglutathione/pharmacology , Animals , Calcium/metabolism , Dogs , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Myocardium/metabolism , Nitric Oxide/pharmacology , Partial Pressure , Peroxynitrous Acid/pharmacology , Sarcoplasmic Reticulum/chemistry
15.
Electrophoresis ; 29(13): 2768-78, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18615785

ABSTRACT

Current algorithms for the calculation of peptide or protein pI, based on the charge associated with individual amino acids, can calculate pI values to within +/-0.2 pI units. Here, we present a new pI calculation algorithm that takes into account the effect of adjacent amino acids on the pI value. The algorithm accounts for the effect of adjacent amino acids+/-3 residues away from a charged aspartic or glutamic acid, as well as effects on the free C terminus, and applies a correction term to the corresponding pK values. The correction increments are derived from a 5000-peptide training set using a genetic optimization approach. The accuracy of the new pI values obtained with this method approaches the error associated with the manufacture of the IPG strip (<+/-0.03 pI units). The approach is demonstrated for cytosolic cell extracts derived from the breast-cancer cell line DU4475, and from membrane preparations from human lung-tissue samples. One potential application of a more highly accurate pI calculation is data filtering of MS/MS outputs that will allow for more complex database searches including gene finding, and validation, and detection of coding single-nucleotide polymorphisms in their expressed form.


Subject(s)
Amino Acids/chemistry , Isoelectric Focusing/methods , Isoelectric Point , Protein Hydrolysates/chemistry , Algorithms , Animals , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Male , Rats , Testis/chemistry , Trypsin/metabolism
16.
Nat Cell Biol ; 10(6): 688-97, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18488020

ABSTRACT

It is now well established that stromal interaction molecule 1 (STIM1) is the calcium sensor of endoplasmic reticulum stores required to activate store-operated calcium entry (SOC) channels at the surface of non-excitable cells. However, little is known about STIM1 in excitable cells, such as striated muscle, where the complement of calcium regulatory molecules is rather disparate from that of non-excitable cells. Here, we show that STIM1 is expressed in both myotubes and adult skeletal muscle. Myotubes lacking functional STIM1 fail to show SOC and fatigue rapidly. Moreover, mice lacking functional STIM1 die perinatally from a skeletal myopathy. In addition, STIM1 haploinsufficiency confers a contractile defect only under conditions where rapid refilling of stores would be needed. These findings provide insight into the role of STIM1 in skeletal muscle and suggest that STIM1 has a universal role as an ER/SR calcium sensor in both excitable and non-excitable cells.


Subject(s)
Calcium/metabolism , Membrane Glycoproteins/physiology , Animals , Calcium Channels/metabolism , Calcium Signaling , Cell Line , Gene Silencing , Membrane Glycoproteins/metabolism , Mice , Models, Biological , Models, Genetic , Muscle Contraction , Muscles/metabolism , Patch-Clamp Techniques , Sarcoplasmic Reticulum/metabolism , Signal Transduction , Stromal Interaction Molecule 1
17.
Mol Cell Biol ; 28(8): 2637-47, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18268005

ABSTRACT

Transient receptor potential (TRP) channels are nonselective cation channels, several of which are expressed in striated muscle. Because the scaffolding protein Homer 1 has been implicated in TRP channel regulation, we hypothesized that Homer proteins play a significant role in skeletal muscle function. Mice lacking Homer 1 exhibited a myopathy characterized by decreased muscle fiber cross-sectional area and decreased skeletal muscle force generation. Homer 1 knockout myotubes displayed increased basal current density and spontaneous cation influx. This spontaneous cation influx in Homer 1 knockout myotubes was blocked by reexpression of Homer 1b, but not Homer 1a, and by gene silencing of TRPC1. Moreover, diminished Homer 1 expression in mouse models of Duchenne's muscular dystrophy suggests that loss of Homer 1 scaffolding of TRP channels may contribute to the increased stretch-activated channel activity observed in mdx myofibers. These findings provide direct evidence that Homer 1 functions as an important scaffold for TRP channels and regulates mechanotransduction in skeletal muscle.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Deletion , Muscular Dystrophies/physiopathology , TRPC Cation Channels/metabolism , Animals , Calcium Signaling , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Homer Scaffolding Proteins , Mice , Mice, Knockout , Muscle Contraction , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Protein Binding , TRPC Cation Channels/genetics
18.
J Biol Chem ; 281(40): 30143-51, 2006 Oct 06.
Article in English | MEDLINE | ID: mdl-16891657

ABSTRACT

Excitation-contraction (EC) coupling in striated muscles is mediated by the cardiac or skeletal muscle isoform of voltage-dependent L-type Ca(2+) channel (Ca(v)1.2 and Ca(v)1.1, respectively) that senses a depolarization of the cell membrane, and in response, activates its corresponding isoform of intracellular Ca(2+) release channel/ryanodine receptor (RyR) to release stored Ca(2+), thereby initiating muscle contraction. Specifically, in cardiac muscle following cell membrane depolarization, Ca(v)1.2 activates cardiac RyR (RyR2) through an influx of extracellular Ca(2+). In contrast, in skeletal muscle, Ca(v)1.1 activates skeletal muscle RyR (RyR1) through a direct physical coupling that negates the need for extracellular Ca(2+). Since airway smooth muscle (ASM) expresses Ca(v)1.2 and all three RyR isoforms, we examined whether a cardiac muscle type of EC coupling also mediates contraction in this tissue. We found that the sustained contractions of rat ASM preparations induced by depolarization with KCl were indeed partially reversed ( approximately 40%) by 200 mum ryanodine, thus indicating a functional coupling of L-type channels and RyRs in ASM. However, KCl still caused transient ASM contractions and stored Ca(2+) release in cultured ASM cells without extracellular Ca(2+). Further analyses of rat ASM indicated that this tissue expresses as many as four L-type channel isoforms, including Ca(v)1.1. Moreover, Ca(v)1.1 and RyR1 in rat ASM cells have a similar distribution near the cell membrane in rat ASM cells and thus may be directly coupled as in skeletal muscle. Collectively, our data implicate that EC-coupling mechanisms in striated muscles may also broadly transduce diverse smooth muscle functions.


Subject(s)
Bronchi/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Amino Acid Sequence , Animals , Bronchi/drug effects , Calcium Channels/genetics , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/physiology , Cells, Cultured , Male , Molecular Sequence Data , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Rats , Rats, Wistar , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/physiology
19.
Curr Alzheimer Res ; 3(3): 201-4, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16842096

ABSTRACT

Memantine, an aminodamantane, has recently been approved to treat moderate-to-severe Alzheimer's disease in the US after over 20 years on the market in Europe for treatment of Parkinson's disease. The unique properties of Memantine allow for its selective inhibition of abnormally active NMDA receptor channels while preserving normal glutamate activity and healthy neuronal function. Recently, it has been shown that compounds such as nitroglycerin, used for years for ischemic coronary disease, can also regulate the NMDA receptor channel. Novel compounds have been synthesized in an attempt to combine these activities, in an attempt to synergistically improve upon the activities of both nitrates and aminoadamantanes. We have subjected these compounds to several laboratory tests to compare their ability to affect the function of the NMDA receptor and to dilate blood vessels. These tests provide an initial indication of which of the compounds may have enhanced activity relative to memantine. The results also provide guidance for the synthesis of additional compounds that are likely to have the properties that are being sought.


Subject(s)
Adamantane/analogs & derivatives , Adamantane/pharmacology , Aorta/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Vasodilation/drug effects , Adamantane/chemical synthesis , Adamantane/metabolism , Alzheimer Disease/drug therapy , Animals , Drug Evaluation, Preclinical , In Vitro Techniques , Nitrates/chemical synthesis , Nitrates/metabolism , Nitrates/pharmacology , Rabbits , Recombinant Proteins , Sodium Channels/drug effects
20.
J Biol Chem ; 281(22): 15572-81, 2006 Jun 02.
Article in English | MEDLINE | ID: mdl-16595676

ABSTRACT

We examined the roles of type 1 and type 2 calsequestrins (CSQ1 and CSQ2) in stored Ca2+ release of C2C12 skeletal muscle myotubes. Transduction of C2C12 myoblasts with CSQ1 or CSQ2 small interfering RNAs effectively reduced the expression of targeted CSQ protein to near undetectable levels. As compared with control infected or CSQ1 knockdown myotubes, CSQ2 and CSQ1/CSQ2 knockdown myotubes had significantly reduced stored Ca2+ release evoked by activators of intracellular Ca2+ release channel/ryanodine receptor (10 mM caffeine, 200 microM 4-chloro-m-cresol, or 10 mM KCl). Thus, CSQ1 is not essential for effective stored Ca2+ release in C2C12 myotubes despite our in vitro studies suggesting that CSQ1 may enhance ryanodine receptor channel activity. To determine the basis of the reduced stored Ca2+ release in CSQ2 knockdown myotubes, we performed immunoblot analyses and found a significant reduction in both sarco/endoplasmic reticulum Ca2+-ATPase and skeletal muscle ryanodine receptor proteins in CSQ2 and CSQ1/CSQ2 knockdown myotubes. Moreover, these knockdown myotubes exhibited reduced Ca2+ uptake and reduced stored Ca2+ release by UTP (400 microM) that activates a different family of intracellular Ca2+ release channels (inositol 1,4,5-trisphosphate receptors). Taken together, our data suggest that knocking down CSQ2, but not CSQ1, leads to reduced Ca2+ storage and release in C2C12 myotubes.


Subject(s)
Calcium Signaling/physiology , Calsequestrin/antagonists & inhibitors , Muscle Fibers, Skeletal/metabolism , Animals , Base Sequence , Calcium Signaling/drug effects , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calsequestrin/classification , Calsequestrin/genetics , Calsequestrin/metabolism , Cell Line , Mice , Muscle Fibers, Skeletal/drug effects , RNA, Small Interfering/genetics , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...