Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
BMJ Open Respir Res ; 8(1)2021 11.
Article in English | MEDLINE | ID: mdl-34764199

ABSTRACT

Singing is an increasingly popular activity for people with chronic obstructive pulmonary disease (COPD). Research to date suggests that 'Singing for Lung Health' may improve various health measures, including health-related quality-of-life. Singing and breathing are closely linked processes affecting one another. In this narrative review, we explore the physiological rationale for 'Singing for Lung Health' as an intervention, focusing on the abnormalities of pulmonary mechanics seen in COPD and how these might be impacted by singing. The potential beneficial physiological mechanisms outlined here require further in-depth evaluation.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Singing , Humans , Lung , Pulmonary Disease, Chronic Obstructive/therapy , Quality of Life
2.
NPJ Prim Care Respir Med ; 26: 16080, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27906158

ABSTRACT

There is growing interest in Singing for Lung Health (SLH), an approach where patients with respiratory disease take part in singing groups, intended to improve their condition. A consensus group was convened in early 2016 to address issues including: the specific features that make SLH distinct from other forms of participation in singing; the existing evidence base via a systematic review; gaps in the evidence base including the need to define value-based outcome measures for sustainable commissioning of SLH; defining the measures needed to evaluate both individuals' responses to SLH and the quality of singing programmes. and core training, expertise and competencies required by singing group leaders to deliver high-quality programmes. A systematic review to establish the extent of the evidence base for SLH was undertaken. Electronic databases, including Pubmed, OVID Medline and Embase, Web of Science, Cochrane central register of controlled trials and PEDro, were used. Six studies were included in the final review. Quantitative data suggest that singing has the potential to improve health-related quality of life, particularly related to physical health, and levels of anxiety without causing significant side effects. There is a significant risk of bias in many of the existing studies with small numbers of subjects overall. Little comparison can be made between studies owing to their heterogeneity in design. Qualitative data indicate that singing is an enjoyable experience for patients, who consistently report that it helps them to cope with their condition better. Larger and longer-term trials are needed.


Subject(s)
Lung Diseases/therapy , Singing , Humans , Lung/physiology , Lung Diseases/psychology , Quality of Life
3.
Cell Signal ; 23(4): 673-82, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21130869

ABSTRACT

Intracellular signaling pathways that regulate the production of lethal proteins in central neurons are not fully characterized. Previously, we reported induction of a novel neuronal protein neuronal pentraxin 1 (NP1) in neonatal brain injury following hypoxia-ischemia (HI); however, how NP1 is induced in hypoxic-ischemic neuronal death remains elusive. Here, we have elucidated the intracellular signaling regulation of NP1 induction in neuronal death. Primary cortical neurons showed a hypoxic-ischemia time-dependent increase in cell death and that NP1 induction preceded the actual neuronal death. NP1 gene silencing by NP1-specific siRNA significantly reduced neuronal death. The specificity of NP1 induction in neuronal death was further confirmed by using NP1 (-/-) null primary cortical neurons. Declines in phospho-Akt (i.e. deactivation) were observed concurrent with decreased phosphorylation of its downstream substrate GSK-3α/ß (at Ser21/Ser9) (i.e. activation) and increased GSK-3α and GSK-3ß kinase activities, which occurred prior to NP1 induction. Expression of a dominant-negative inhibitor of Akt (Akt-kd) blocked phosphorylation of GSK-3α/ß and subsequently enhanced NP1 induction. Whereas, overexpression of constitutively activated Akt (Akt-myr) or wild-type Akt (wtAkt) increased GSK-α/ß phosphorylation and attenuated NP1 induction. Transfection of neurons with GSK-3α siRNA completely blocked NP1 induction and cell death. Similarly, overexpression of the GSK-3ß inhibitor Frat1 or the kinase mutant GSK-3ßKM, but not the wild-type GSK-3ßWT, blocked NP1 induction and rescued neurons from death. Our findings clearly implicate both GSK-3α- and GSK-3ß-dependent mechanism of NP1 induction and point to a novel mechanism in the regulation of hypoxic-ischemic neuronal death.


Subject(s)
C-Reactive Protein/metabolism , Glycogen Synthase Kinase 3/metabolism , Hypoxia-Ischemia, Brain/pathology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , C-Reactive Protein/genetics , Cell Culture Techniques , Cell Death , Cell Hypoxia , Cell Survival , Cells, Cultured , Enzyme Activation , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta , Hypoxia-Ischemia, Brain/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Rats , Rats, Inbred F344
4.
J Neurochem ; 106(3): 1357-70, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18485100

ABSTRACT

The inhibitors of apoptosis (IAPs) are emerging as key proteins in the control of cell death. In this study, we evaluated the expression and subcellular distribution of the antiapoptotic protein X-linked IAP (XIAP), and its interactions with the XIAP-associated factor 1 (XAF1) in neonatal rat brain following hypoxia-ischemia (HI). HI triggered the mitochondrial release of cytochrome c, Smac/DIABLO, and caspase 3 activation. Confocal microscopy detected XIAP-specific immunofluorescence in the cytoplasm under normal condition, which exhibited a diffuse distribution at 6 h post-HI and by 12 h the majority of XIAP was redistributed into the nucleus. XIAP nuclear translocation was confirmed by subcellular fractionations and by expressing FLAG-tagged XIAP in primary cortical neurons. Over-expression of XIAP significantly reduced, whereas XIAP gene silencing further enhanced cell death, demonstrating a specific requirement of cytoplasmic XIAP for cell survival. An elevated level of cytosolic XIAP was also evident under the conditions of neuroprotection by fibroblast growth factor-1. XAF1 expression was increased temporally and there was increased nuclear co-localization with XIAP in hypoxic-ischemic cells. XIAP co-immunoprecipitated > 9-fold XAF1 protein concurrent with decreased association with caspases 9 and 3. This is evidenced by the enhanced caspase 3 activity and neuronal death. Our findings implicate XIAP nuclear translocation in neuronal death and point to a novel mechanism in the regulation of hypoxic-ischemic brain injury.


Subject(s)
Cell Nucleus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Neurons/cytology , Neurons/physiology , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , Animals , Animals, Newborn , Apoptosis/physiology , Cell Death/physiology , Cell Nucleus/genetics , Cell Nucleus/pathology , Cell Survival/physiology , Cells, Cultured , Cytoplasm/metabolism , Cytoplasm/physiology , Humans , Hypoxia-Ischemia, Brain/genetics , Neurons/metabolism , Rats , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/physiology
5.
Neurobiol Dis ; 22(3): 677-90, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16635575

ABSTRACT

Perinatal hypoxia-ischemia (HI) is a major cause of neurological disability and mortality in infant and children. In the present study, we explored the neuroprotective efficacy of FGF-1 in a rat model of perinatal HI. Carotid ligation combined with hypoxia caused marked infarctions in the ipsilateral cerebral hemisphere with significant loss of ipsilateral striatal, cortical and hippocampal volumes. Morphological analyses revealed both apoptotic and necrotic form of neuronal death determined by Nissl histology, dark-field microscopy and TUNEL staining. HI induced a marked increase in activated caspase-9, caspase-3 and PARP cleavage at 12 h to 7 days after HI in brain areas displaying TUNEL (+) cells. In addition, expression of the anti-apoptotic protein X-linked inhibitor of apoptosis (XIAP) was decreased under similar conditions of HI. Expression of human FGF-1 in brain significantly reduced the extent of both apoptotic and necrotic injury caused by HI. FGF-1 attenuated the HI-induced increase in activated caspase-3, caspase-9 and cleaved PARP protein levels and markedly blocked the HI-induced decrease in XIAP expression under the conditions at which FGF-1 showed significant neuroprotection. These findings demonstrate that FGF-1 prevents the onset of both apoptotic and necrotic death in neurons otherwise "destined to die" following hypoxic-ischemic injury by intervening at the level of caspase-signaling cascades and by restoring prosurvival protein XIAP expression in central neurons.


Subject(s)
Caspases/drug effects , Fibroblast Growth Factor 1/biosynthesis , Hypoxia-Ischemia, Brain/drug therapy , Signal Transduction/drug effects , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , Animals , Animals, Genetically Modified , Animals, Newborn , Apoptosis/drug effects , Blotting, Western , Cell Transplantation , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Fibroblast Growth Factor 1/genetics , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Rats , Signal Transduction/physiology , Transgenes
6.
Ann Neurol ; 55(5): 660-7, 2004 May.
Article in English | MEDLINE | ID: mdl-15122706

ABSTRACT

Brain injury from inorganic Pb(2+) is considered the most important environmental childhood health hazard worldwide. The microvasculature of the developing brain is uniquely susceptible to high level Pb(2+) toxicity (ie, Pb(2+) encephalopathy) characterized by cerebellar hemorrhage, increased blood-brain barrier permeability, and vasogenic edema. However, the specific molecular mediators of Pb(2+) encephalopathy have been elusive. We found that Pb(2+) induces vascular endothelial growth factor/vascular permeability factor (VEGF) in cultured astrocytes (J Biol Chem, 2000;275:27874-27882). The study presented here asks if VEGF dysregulation contributes mechanistically to Pb(2+) encephalopathy. Neonatal rats exposed to 4% Pb-carbonate develop the histopathological features of Pb(2+) encephalopathy seen in children. Cerebellar VEGF expression increased approximately twofold (p < 0.01) concurrent with the development of cerebellar microvascular hemorrhage, enhanced vascular permeability to serum albumin, and vasogenic cerebellar edema (p < 0.01). No change in VEGF expression occurred in cerebral cortex that does not develop these histopathological complications of acute Pb(2+) intoxication. Pb(2+) exposure increased phosphorylation of cerebellar Flk-1 VEGF receptors and the Flk-1 inhibitor CEP-3967 completely blocked cerebellar edema formation without affecting microhemorrhage formation or blood-brain barrier permeability. This establishes that Pb(2+)-induced vasogenic edema formation develops via a Flk-1-dependent mechanism and suggests that the vascular permeability caused by Pb(2+) is Flk-1 independent.


Subject(s)
Brain Edema/metabolism , Lead Poisoning/metabolism , Lead/toxicity , Vascular Endothelial Growth Factor A/biosynthesis , Acute Disease , Animals , Animals, Newborn , Brain Edema/pathology , Female , Lead Poisoning/pathology , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/genetics
7.
J Neurosci ; 24(17): 4187-96, 2004 Apr 28.
Article in English | MEDLINE | ID: mdl-15115814

ABSTRACT

Neonatal hypoxic-ischemic brain injury is a major cause of neurological disability and mortality. Its therapy will likely require a greater understanding of the discrete neurotoxic molecular mechanism(s) triggered by hypoxia-ischemia (HI). Here, we investigated the role of neuronal pentraxin 1 (NP1), a member of a newly recognized subfamily of "long pentraxins," in the HI injury cascade. Neonatal brains developed marked infarcts in the ipsilateral cerebral hemisphere at 24 hr and showed significant loss of ipsilateral striatal, cortical, and hippocampal volumes at 7 d after HI compared with the contralateral hemisphere and sham controls. Immunofluorescence analyses revealed elevated neuronal expression of NP1 in the ipsilateral cerebral cortex from 6 hr to 7 d and in the hippocampal CA1 and CA3 regions from 24 hr to 7 d after HI. These same brain areas developed infarcts and terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling-positive cells within 24-48 hr of HI. In primary cortical neurons, NP1 protein was induced >2.5-fold (p < 0.001) after their exposure to hypoxia that caused approximately 30-40% neuronal death. Transfecting cortical neurons with antisense oligodeoxyribonucleotides directed against NP1 mRNA (NP1AS) significantly inhibited (p < 0.01) hypoxia-induced NP1 protein induction and neuronal death (p < 0.001), demonstrating a specific requirement of NP1 in hypoxic neuronal injury. NP1 protein colocalized and coimmunoprecipitated with the fast excitatory AMPA glutamate receptor subunit (GluR1) in primary cortical neurons, and hypoxia induced a time-dependent increase in NP1-GluR1 interactions. NPIAS also protected against AMPA-induced neuronal death (p < 0.05), implicating a role for NP1 in the excitotoxic cascade. Our results show that NP1 induction mediates hypoxic-ischemic injury probably by interacting with and modulating GluR1 and potentially other excitatory glutamate receptors.


Subject(s)
Brain/physiopathology , C-Reactive Protein/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Brain/drug effects , Brain/pathology , C-Reactive Protein/genetics , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/prevention & control , In Situ Nick-End Labeling , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/pathology , Neurotoxins/antagonists & inhibitors , Neurotoxins/toxicity , Oligonucleotides, Antisense/pharmacology , Protein Binding/physiology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Receptors, AMPA/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/antagonists & inhibitors , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity
8.
J Neurochem ; 81(2): 365-78, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12064484

ABSTRACT

Neuroprotective actions of scatter factor/hepatocyte growth factor (SF/HGF) have not been described. We examined the effects of SF/HGF in comparison to acidic fibroblast growth factor-1 (FGF-1) on N-methyl-D-aspartate (NMDA) and quinolinic acid (QUIN)-induced excitotoxicity in primary cerebellar granule neurons. Exposure to NMDA or QUIN for 24 h resulted in concentration-dependent cell death (p < 0.001) that was completely attenuated (p < 0.001) by pre-treatment of cells with SF/HGF (50 ng/mL) or FGF-1 (40 ng/mL). SF/ HGF and FGF-1 activated both Akt and MAP-kinase > threefold (p < 0.001). Neither SF/HGF nor FGF-1 activated cyclic AMP-response element binding protein (CREB), a downstream target of MAP-kinase, whereas brain-derived neurotrophic factor (BDNF) activated both MAP-kinase and CREB in granule neurons. Neuroprotection against NMDA or QUIN by SF/HGF and FGF-1 was negated by the addition of LY294002 (10 microM) or wortmannin (100 microM), two distinct inhibitors of phosphatidylinositol 3-kinase (P13-K), but not by the MAP-kinase kinase (MEK) inhibitor PD98059 (33 microm). Likewise, expression of a dominant-negative mutant of Akt (Akt-kd) completely prevented the neuroprotective actions of SF/HGF and FGF-1. Overexpression of a constitutively activated Akt (Akt-myr) or wild-type Akt (wtAkt) attenuated excitotoxic cell death. These data show that both SF/HGF and FGF-1 protect cerebellar granule neurons against excitotoxicity with similar potency in a P13-K/Akt-dependent and MAP-kinase/CREB-independent manner.


Subject(s)
Cerebellum/drug effects , Fibroblast Growth Factor 1/pharmacology , Hepatocyte Growth Factor/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Animals , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cytoprotection , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/toxicity , Neurons/cytology , Neurons/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-met/biosynthesis , Rats , Rats, Inbred Lew
SELECTION OF CITATIONS
SEARCH DETAIL
...