Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Am J Transplant ; 12(11): 3076-84, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22883104

ABSTRACT

Bronchiolitis obliterans syndrome (BOS) is a condition of progressive airflow obstruction that affects a majority of lung transplant recipients and limits long-term posttransplant survival. Although epithelial injury appears central to the development of BOS, little is known regarding the specific epithelial cell types that are affected in this condition. We hypothesized that BOS would involve preferential injury to the secretory Clara cells that function in innate defense and epithelial repair. To test this hypothesis, we assessed tissue transcript, tissue protein and lung fluid protein expression of Clara cell secretory protein (CCSP), a marker for Clara cells, in lung transplant recipients with BOS, BOS-free patients and in donor controls. Our results demonstrate that CCSP tissue transcript and protein expression are significantly reduced in lung transplant recipients with BOS compared to BOS-free or donor controls. In addition, we demonstrate that CCSP protein levels are significantly reduced in the lung fluid of patients with BOS compared to BOS-free controls, in cross-sectional and longitudinal analysis. Collectively, these complementary results illustrate that BOS involves a selective alteration in the distribution and function of bronchiolar Clara cells.


Subject(s)
Bronchiolitis Obliterans/etiology , Bronchiolitis Obliterans/pathology , Bronchoalveolar Lavage Fluid/cytology , Epithelial Cells/metabolism , Lung Transplantation/adverse effects , Uteroglobin/metabolism , Adult , Biomarkers/metabolism , Case-Control Studies , Epithelial Cells/pathology , Female , Fluorescent Antibody Technique , Graft Rejection , Graft Survival , Humans , Immunohistochemistry , Lung Transplantation/methods , Male , Middle Aged , Prognosis , Prospective Studies , Reference Values , Risk Factors , Severity of Illness Index , Syndrome , Uteroglobin/genetics
2.
Am J Transplant ; 8(9): 1946-50, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18671675

ABSTRACT

Lymphangiomatosis is a rare disease of lymphatic proliferation for which no adequate treatment is known. We report the first successful case of bilateral lung transplantation for the treatment of end-stage pulmonary lymphangiomatosis. A successful outcome was achieved with continued survival beyond 4 years posttransplant and stable lung function. The primary obstacles to significant gains in pulmonary function were thoracic, skeletal and abdominal lymphangiomatosis, which led to pulmonary restriction. Our report demonstrates that pulmonary lymphangiomatosis should be included among those diseases for which lung transplantation is considered potentially beneficial treatment but also emphasizes the importance of screening patients carefully for chest wall and abdominal lymphangiomas that may impede recovery.


Subject(s)
Lung Neoplasms/surgery , Lung Transplantation , Lymphangioma/surgery , Adult , Female , Follow-Up Studies , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Lymphangioma/diagnostic imaging , Lymphangioma/pathology , Radiography , Time Factors , Treatment Outcome
3.
Proc Natl Acad Sci U S A ; 98(20): 11158-62, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11562475

ABSTRACT

We have shown previously that at physiologically relevant oxygen tension (pO(2) approximately 10 mmHg), NO S-nitrosylates 1 of approximately 50 free cysteines per ryanodine receptor 1 (RyR1) subunit and transduces a calcium-sensitizing effect on the channel by means of calmodulin (CaM). It has been suggested that cysteine-3635 is part of a CaM-binding domain, and its reactivity is attenuated by CaM [Porter Moore, C., Zhang, J. Z., Hamilton, S. L. (1999) J. Biol. Chem. 274, 36831-36834]. Therefore, we tested the hypothesis that the effect of NO was mediated by C3635. The full-length RyR1 single-site C3635A mutant was generated and expressed in HEK293 cells. The mutation resulted in the loss of CaM-dependent NO modulation of channel activity and reduced S-nitrosylation by NO to background levels but did not affect NO-independent channel modulation by CaM or the redox sensitivity of the channel to O(2) and glutathione. Our results reveal that different cysteines within the channel have been adapted to serve in nitrosative and oxidative responses, and that S-nitrosylation of the cysteine-containing CaM-binding domain underlies the mechanism of CaM-dependent regulation of RyR1 by NO.


Subject(s)
Calmodulin/metabolism , Cysteine , Muscle, Skeletal/metabolism , Nitric Oxide/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine/metabolism , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Glutathione/pharmacology , Humans , Intracellular Membranes/metabolism , Microsomes/metabolism , Mutagenesis, Site-Directed , Nitric Oxide/pharmacology , Oxidation-Reduction , Oxygen/pharmacology , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/drug effects , Sarcoplasmic Reticulum/metabolism , Transfection
4.
Biochemistry ; 40(16): 4904-10, 2001 Apr 24.
Article in English | MEDLINE | ID: mdl-11305905

ABSTRACT

Nitric oxide (NO) and related molecules play important roles in vascular biology. NO modifies proteins through nitrosylation of free cysteine residues, and such modifications are important in mediating NO's biologic activity. Tissue transglutaminase (tTG) is a sulfhydryl rich protein that is expressed by endothelial cells and secreted into the extracellular matrix (ECM) where it is bound to fibronectin. Tissue TG exhibits a Ca(2+)-dependent transglutaminase activity (TGase) that cross-links proteins involved in wound healing, tissue remodeling, and ECM stabilization. Since tTG is in proximity to sites of NO production, has 18 free cysteine residues, and utilizes a cysteine for catalysis, we investigated the factors that regulated NO binding and tTG activity. We report that TGase activity is regulated by NO through a unique Ca(2+)-dependent mechanism. Tissue TG can be poly-S-nitrosylated by the NO carrier, S-nitrosocysteine (CysNO). In the absence of Ca(2+), up to eight cysteines were nitrosylated without modifying TGase activity. In the presence of Ca(2+), up to 15 cysteines were found to be nitrosylated and this modification resulted in an inhibition of TGase activity. The addition of Ca(2+) to nitrosylated tTG was able to trigger the release of NO groups (i.e. denitrosylation). tTG nitrosylated in the absence of Ca(2+) was 6-fold more susceptible to inhibition by Mg-GTP. When endothelial cells in culture were incubated with tTG and stimulated to produce NO, the exogenous tTG was S-nitrosylated. Furthermore, S-nitrosylated tTG inhibited platelet aggregation induced by ADP. In conclusion, we provide evidence that Ca(2+) regulates the S-nitrosylation and denitrosylation of tTG and thereby TGase activity. These data suggest a novel allosteric role for Ca(2+) in regulating the inhibition of tTG by NO and a novel function for tTG in dispensing NO bioactivity.


Subject(s)
Calcium/physiology , GTP-Binding Proteins/metabolism , Mercaptoethanol , Nitric Oxide/metabolism , Phosphorylcholine/analogs & derivatives , S-Nitrosothiols , Sphingosine/analogs & derivatives , Transglutaminases/metabolism , Adenosine Diphosphate/physiology , Adenosine Triphosphate/pharmacology , Animals , Cations, Divalent/pharmacology , Cattle , Cells, Cultured , Cysteine/analogs & derivatives , Cysteine/pharmacology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Guanosine Triphosphate/pharmacology , Guinea Pigs , Humans , Kinetics , Nitroso Compounds/metabolism , Nitroso Compounds/pharmacology , Phosphorylcholine/metabolism , Platelet Aggregation , Protein Conformation , Protein Glutamine gamma Glutamyltransferase 2 , Recombinant Proteins/metabolism , Sphingosine/metabolism , Transglutaminases/antagonists & inhibitors , Transglutaminases/chemistry , Transglutaminases/genetics
5.
J Biol Chem ; 276(19): 15625-30, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11278999

ABSTRACT

The skeletal muscle Ca(2+) release channel/ryanodine receptor (RyR1) is a prototypic redox-responsive ion channel. Nearly half of the 101 cysteines per RyR1 subunit are kept in a reduced (free thiol) state under conditions comparable with resting muscle. Here we assessed the effects of physiological determinants of cellular redox state (oxygen tension, reduced (GSH) or oxidized (GSSG) glutathione, and NO/O(2) (released by 3-morpholinosydnonimine)) on RyR1 redox state and activity. Oxidation of approximately 10 RyR1 thiols (from approximately 48 to approximately 38 thiols/RyR1 subunit) had little effect on channel activity. Channel activity increased reversibly as the number of thiols was further reduced to approximately 23/subunit, whereas more extensive oxidation (to approximately 13 thiols/subunit) inactivated the channel irreversibly. Neither S-nitrosylation nor tyrosine nitration contributed to these effects. The results identify at least three functional classes of RyR1 thiols and suggest that 1) the channel may be protected from oxidation by a large reservoir of functionally inert thiols, 2) the channel may be designed to respond to moderate oxidative stress by a change in activation setpoint, and 3) the channel is susceptible to oxidative injury under more extensive conditions.


Subject(s)
Glutathione/metabolism , Molsidomine/pharmacology , Muscle, Skeletal/physiology , Nitric Oxide Donors/pharmacology , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/physiology , Sulfhydryl Compounds/metabolism , Animals , Glutathione Disulfide/metabolism , Kinetics , Molsidomine/analogs & derivatives , Nitric Oxide/metabolism , Oxidation-Reduction , Protein Subunits , Rabbits , Ryanodine/pharmacokinetics , Ryanodine Receptor Calcium Release Channel/drug effects , Superoxides/metabolism
6.
Cell ; 102(4): 499-509, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10966111

ABSTRACT

Ion channels have been studied extensively in ambient O2 tension (pO2), whereas tissue PO2 is much lower. The skeletal muscle calcium release channel/ryanodine receptor (RyR1) is one prominent example. Here we report that PO2 dynamically controls the redox state of 6-8 out of 50 thiols in each RyR1 subunit and thereby tunes the response to NO. At physiological pO2, nanomolar NO activates the channel by S-nitrosylating a single cysteine residue. Among sarcoplasmic reticulum proteins, S-nitrosylation is specific to RyR1 and its effect on the channel is calmodulin dependent. Neither activation nor S-nitrosylation of the channel occurs at ambient PO2. The demonstration that channel cysteine residues subserve coupled O2 sensor and NO regulatory functions and that these operate through the prototypic allosteric effector calmodulin may have general implications for the regulation of redox-related systems.


Subject(s)
Muscle, Skeletal/physiology , Nitric Oxide/metabolism , Oxygen/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Signal Transduction , Animals , Calcium/metabolism , Calcium-Transporting ATPases/metabolism , Calmodulin/metabolism , Oxidation-Reduction , Rabbits , Ryanodine/metabolism , Sarcoplasmic Reticulum/metabolism
7.
Biochemistry ; 39(5): 1040-7, 2000 Feb 08.
Article in English | MEDLINE | ID: mdl-10653649

ABSTRACT

Nitric oxide overproduction has been implicated in the pathogenesis of many disorders, including artherosclerosis, neurodegenerative diseases, inflammatory and autoimmune diseases, and cancer. The common view holds that nitric oxide-induced cellular injury is caused by oxidative stress. This theory predicts that interactions between reactive nitrogen species and reactive oxygen species produce powerful oxidants that initiate cell death programs. Cytokine-treated murine macrophages are the prototype of this form of cellular injury. Here we report that generation of reactive nitrogen species upon lipopolysacharide/interferon-gamma stimulation of RAW 264.7 cells is largely divorced from production of reactive oxygen species, and that oxidative stress is not principally responsible for cell death (in this model). Rather, the death program is induced mainly by a nitrosative challenge, characterized by the accrual of nitrosylated proteins without a major alteration in cellular redox state. Moreover, interactions between reactive oxygen and nitrogen species may alter the balance between pathways that yield nitrite and nitrate, without impacting the level of S-nitrosylation or extent of cell death. Our results thus (1) provide new insights into NO-related metabolic pathways, (2) demonstrate that apoptotic injury can be caused by nitrosative mechanisms, and (3) establish a model for nitrosative stress in mammalian cells.


Subject(s)
Apoptosis , Macrophages/cytology , Macrophages/metabolism , Nitroso Compounds/metabolism , 2-Naphthylamine/analogs & derivatives , 2-Naphthylamine/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cytokines/toxicity , Free Radicals/metabolism , Glutathione/analogs & derivatives , Glutathione/toxicity , Macrophages/drug effects , Macrophages/pathology , Mice , Nitrates/metabolism , Nitric Oxide Donors/toxicity , Nitrites/metabolism , Nitrosation , Nitroso Compounds/toxicity , Oxidation-Reduction , Reactive Oxygen Species/metabolism , S-Nitrosoglutathione
8.
Biochem Pharmacol ; 57(10): 1079-84, 1999 May 15.
Article in English | MEDLINE | ID: mdl-11230795

ABSTRACT

The ryanodine receptors (RyRs) are large intracellular calcium release channels that play an important role in the control of the calcium levels in excitable and non-excitable cells. Many endogenous modulators such as Mg2+, ATP, or calmodulin can affect the channel activities of the three known mammalian RyR isoforms. RyRs also are known to be redox-responsive. However, the molecular basis and the physiological relevance of redox modulation of RyRs are unclear. Recent evidence suggests that nitric oxide (NO) and related molecules may be endogenous regulators of the skeletal and cardiac muscle RyRs. The two tissues express nitric oxide synthases (NOSs), and NO or NO-related species have been shown to affect Ca2+ release channel activities directly via covalent modifications of thiol groups. Both an oxidative and a nitrosative modification of RyRs have been described, leading to either a reversible or irreversible alteration of RyR ion channel activity. Additional mechanisms of regulation may include cyclic GMP-dependent signaling pathways and NO modification of RyR regulatory proteins such as the surface membrane L-type Ca2+ channel. Modification of RyRs by NO may influence a variety of physiological functions such as insulin release, vasomotor control, and muscle contraction.


Subject(s)
Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium Channels, L-Type/metabolism , Cyclic GMP/metabolism , Humans , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Oxidation-Reduction
9.
Science ; 279(5348): 234-7, 1998 Jan 09.
Article in English | MEDLINE | ID: mdl-9422697

ABSTRACT

Several ion channels are reportedly redox responsive, but the molecular basis for the changes in activity is not known. The mechanism of nitric oxide action on the cardiac calcium release channel (ryanodine receptor) (CRC) in canines was explored. This tetrameric channel contains approximately 84 free thiols and is S-nitrosylated in vivo. S-Nitrosylation of up to 12 sites (3 per CRC subunit) led to progressive channel activation that was reversed by denitrosylation. In contrast, oxidation of 20 to 24 thiols per CRC (5 or 6 per subunit) had no effect on channel function. Oxidation of additional thiols (or of another class of thiols) produced irreversible activation. The CRC thus appears to be regulated by poly-S-nitrosylation (multiple covalent attachments), whereas oxidation can lead to loss of control. These results reveal that ion channels can differentiate nitrosative from oxidative signals and indicate that the CRC is regulated by posttranslational chemical modification(s) of sulfurs.


Subject(s)
Calcium/metabolism , Mercaptoethanol , Myocardium/metabolism , Nitric Oxide/metabolism , Nitroso Compounds/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , S-Nitrosothiols , Animals , Cyclic GMP/metabolism , Cysteine/analogs & derivatives , Cysteine/pharmacology , Dithiothreitol/pharmacology , Dogs , Electric Conductivity , Ethylmaleimide/pharmacology , Glutathione/analogs & derivatives , Glutathione/pharmacology , Liposomes/metabolism , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Nitrosation , Nitroso Compounds/pharmacology , Oxidation-Reduction , Proteolipids/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , S-Nitrosoglutathione , Sulfhydryl Compounds/metabolism
10.
Science ; 276(5321): 2034-7, 1997 Jun 27.
Article in English | MEDLINE | ID: mdl-9197264

ABSTRACT

The binding of oxygen to heme irons in hemoglobin promotes the binding of nitric oxide (NO) to cysteinebeta93, forming S-nitrosohemoglobin. Deoxygenation is accompanied by an allosteric transition in S-nitrosohemoglobin [from the R (oxygenated) to the T (deoxygenated) structure] that releases the NO group. S-nitrosohemoglobin contracts blood vessels and decreases cerebral perfusion in the R structure and relaxes vessels to improve blood flow in the T structure. By thus sensing the physiological oxygen gradient in tissues, hemoglobin exploits conformation-associated changes in the position of cysteinebeta93 SNO to bring local blood flow into line with oxygen requirements.


Subject(s)
Cerebrovascular Circulation , Hemodynamics , Hemoglobins/physiology , Mercaptoethanol , Oxygen/blood , S-Nitrosothiols , Animals , Blood Pressure , Cysteine/chemistry , Cysteine/metabolism , Hemoglobins/analysis , Hemoglobins/chemistry , Models, Molecular , Nitric Oxide/blood , Nitric Oxide/metabolism , Nitroso Compounds/blood , Oxyhemoglobins/chemistry , Protein Conformation , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL