Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Mol Med (Berl) ; 102(7): 913-926, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38753040

ABSTRACT

DEAD-box helicase 53 (DDX53) is a member of the DEAD-box protein family of RNA helicases. Unlike other family members that are responsible for RNA metabolism, the biological function of DDX53 and its impact on the human condition are unclear. Herein, we found a full-length DDX53 deletion mutation in a hereditary spastic paraplegia-like (HSP-like) patient with lower extremity spasticity, walking disorder, visual impairment, and lateral ventricular white matter lesions. Bioinformatic analysis revealed that DDX53 was mainly expressed in the cerebellar cortex and may function as a tissue-specific RNA helicase. Transcriptome analysis showed that the expression of multiple brain-associated genes involved in synapse organization, neuron function, and neuromuscular junctions was affected by DDX53 depletion. Moreover, RNA immunoprecipitation sequencing (RIP-seq) analysis showed that DDX53 interacted with 176 genes, and 96 of these genes were associated with the execution of neurofunction, particularly in the regulation of cell projection organization and nervous system development. Collectively, although a more specified cell or animal model is required to fully understand the functional role of DDX53 in the human brain, we report for the first time that the patient with DDX53 defects exhibits HSP-like symptoms and that DDX53 is essential for maintaining neuronal function, with loss-of-function mutation in DDX53 potentially leading to HSP due to impaired RNA metabolism in the nervous system. KEY MESSAGES: DDX53 deficiency was first reported to be associated with HSP disorder. DDX53 exhibited minimal impact on mitochondrial function. DDX53 impaired RNA metabolism in the nervous system.


Subject(s)
DEAD-box RNA Helicases , Spastic Paraplegia, Hereditary , Female , Humans , Male , Brain/metabolism , Brain/pathology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Loss of Function Mutation , Pedigree , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Adult
2.
STAR Protoc ; 4(1): 102088, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36853693

ABSTRACT

Here, we provide a protocol to isolate mitochondria from cultured cells and extract differently located mitochondrial proteins. We detail steps to separate both integral and peripheral membrane proteins from soluble proteins using sonication. We describe the separation of integral membrane proteins from the peripheral membrane and soluble proteins using sodium carbonate extraction. Furthermore, we detail the use of proteinase K and Triton X-100 to distinguish outer membrane proteins from mitochondrial proteins.


Subject(s)
Mitochondria , Mitochondrial Proteins , Membrane Proteins , Octoxynol , Endopeptidase K
3.
Am J Physiol Lung Cell Mol Physiol ; 292(1): L278-86, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16920889

ABSTRACT

Endothelin-1 (ET-1), a G protein-coupled receptor-activating peptide, is increased in airway epithelium, plasma, and bronchoalveolar lavage fluid of asthmatic patients. We hypothesized that ET-1 may contribute to the increased airway smooth muscle mass found in severe asthma by inducing hypertrophy and inhibiting apoptosis of smooth muscle cells. To investigate this hypothesis, we determined that treatment of primary human bronchial smooth muscle cells with ET-1 dose dependently [10(-11)-10(-7) M] inhibited the apoptosis induced by serum withdrawal. ET-1 treatment also resulted in a significant increase in total protein synthesis, mediated through both ET(A) and ET(B) receptors, cell size, as well as increased expression of myosin heavy chain, alpha-smooth muscle actin, and calponin. ET-1-induced hypertrophy was accompanied by activation of JAK1/STAT-3 and MAPK1/2 (ERK1/2) cell signaling pathways. Inhibition of JAK1/STAT-3 pathways by piceatannol or ERK1/2 by the MAPK/ERK kinase 1/2 inhibitor U0126 blunted the increase in total protein synthesis. The hypertrophic effect of ET-1 was equivalent to that of the gp130 cytokine oncostatin M and greater than that induced by cardiotrophin-1. ET-1 induced release of IL-6 but not IL-11, leukemia inhibitory factor, oncostatin M, or cardiotrophin-1, although treatment of cells with IL-6 alone did not induce hypertrophy. These results suggest that ET-1 is a candidate mediator for the induction of increased smooth muscle mass in asthma and identify signaling pathways activated by this mediator.


Subject(s)
Bronchi/drug effects , Bronchi/pathology , Endothelin-1/pharmacology , Muscle, Smooth/drug effects , Muscle, Smooth/pathology , Apoptosis/drug effects , Butadienes/pharmacology , Cell Enlargement/drug effects , Cells, Cultured , Cytokines/pharmacology , Humans , Interleukin-6/biosynthesis , Interleukin-6/pharmacology , Janus Kinase 1/metabolism , MAP Kinase Signaling System/drug effects , Muscle Proteins/metabolism , Nitriles/pharmacology , Oncostatin M/pharmacology , Recombinant Proteins/pharmacology , STAT3 Transcription Factor/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 287(6): L1165-71, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15273080

ABSTRACT

Induction of hypertrophy and inhibition of apoptosis may be important mechanisms contributing to increased airway smooth muscle (ASM) mass in asthma. Data from our laboratory indicate that cardiotrophin-1 (CT-1) induces hypertrophy and inhibits apoptosis in isolated human ASM cells. To determine whether these novel effects of CT-1 also occur in the airway tissue milieu and to determine whether structural changes are accompanied by functional changes, matched pairs of guinea pig airway explants were treated with or without CT-1 for 7 days, and structural features as well as isometric and isotonic contractile and relaxant mechanical properties were measured. CT-1 (0.2-5 ng/ml) increased both myocyte mass and extracellular matrix in a concentration-dependent fashion. CT-1 (10 ng/ml)-treated tissues exhibited a significant increase in passive tension at all lengths on day 7; at optimal length, passive tension generated by CT-1-treated tissues was 1.72 +/- 0.12 vs. 1.0 +/- 0.1 g for control. Maximal isometric stress was decreased in the CT-1-treated group on day 7 (0.39 +/- 0.10 kg/cm(2)) vs. control (0.77 +/- 0.15 kg/cm(2), P < 0.05). Isoproterenol-induced relaxant potency was reduced in CT-1-treated tissues, log EC(50) being -7.28 +/- 0.34 vs. -8.12 +/- 0.25 M in control, P < 0.05. These data indicate that CT-1 alters ASM structural and mechanical properties in the tissue environment and suggest that structural changes found in the airway wall in asthma are not necessarily associated with increased responsiveness.


Subject(s)
Cytokines/pharmacology , Muscle, Smooth/physiology , Trachea/physiology , Animals , Female , Guinea Pigs , Isoproterenol/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Organ Culture Techniques , Trachea/cytology , Trachea/drug effects
5.
Br J Pharmacol ; 140(7): 1237-44, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14597603

ABSTRACT

1. Cellular hypertrophy and/or a reduced rate of apoptosis could increase airway smooth muscle mass. As cardiotrophin-1 (CT-1) induces hypertrophy and inhibits apoptosis in cardiomyocytes, we tested for the expression and effects of CT-1 in human bronchial smooth muscle cells (HBSMC). 2. CT-1 was detected in abundance in normal adult human lung and was expressed in both fetal and adult HBSMC. 3. Following serum deprivation, CT-1 was released by reintroduction of serum and by TGF-beta 2/IL-4 in fetal but not adult cells. TGF-beta 2/IL-4 triggered the release of CT-1 in serum-fed adult cells. Hypoxia and strain had no effect on the release of CT-1. 4. CT-1 reduced the apoptosis induced both by serum deprivation and by Fas antibody/TNF-alpha treatment in adult cells, with greater efficacy than other members of the IL-6 superfamily. The MAPK/ERK kinase inhibitor PD98059 (1-10 microM) reduced the effect of CT-1. Fetal cells were more resistant to apoptosis. 5. CT-1 (10 ng ml-1) induced a significant increase in cell size as judged by protein/DNA ratios and flow cytometry. No effects on smooth muscle alpha-actin or vimentin proteins were noted, although CT-1 qualitatively alters the cytostructural distribution of SM22, an actin filament-associated protein, and increased SM22 protein abundance. No effect on proliferation or migration was evident. 6. These data suggest CT-1 expression primarily in fetal and synthetic HBSMC phenotypes. By reducing the rates of apoptosis and inducing hypertrophy, CT-1 may contribute to increased smooth muscle mass in airway disease.


Subject(s)
Bronchi/cytology , Cardiolipins/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Adolescent , Adult , Apoptosis/drug effects , Cardiolipins/metabolism , Cell Size/drug effects , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Enzyme Inhibitors/pharmacology , Fetus , Flavonoids/pharmacology , Fluorescent Antibody Technique, Direct , Humans , Immunohistochemistry , Male , Mitogen-Activated Protein Kinases/drug effects , Muscle, Smooth, Vascular/physiology , Tumor Necrosis Factor-alpha/pharmacology
6.
Pharmacotherapy ; 23(5): 561-71, 2003 May.
Article in English | MEDLINE | ID: mdl-12741429

ABSTRACT

STUDY OBJECTIVES: To characterize asthma symptoms and pulmonary function throughout two menstrual cycles, with and without exogenous estradiol administration, in women with premenstrual asthma, and to determine the effect of estradiol administration on asthma symptoms, pulmonary function, quality of life, and biomarkers of airway inflammation. DESIGN: Double-blind, randomized, placebo-controlled, crossover study. SETTING: Respiratory clinic and clinical research center. SUBJECTS: Twelve women with documented premenstrual asthma (> or = 20% premenstrual worsening of asthma symptoms and/or of peak expiratory flow [PEF] during a 1-month screening phase). INTERVENTION: Each woman received either estradiol 2 mg or placebo orally between cycle days 23 and 28 (i.e., premenstrually, or before the onset of menses) in the first cycle and then crossed over to the other arm in the second cycle. Throughout both cycles, the women recorded daily morning and evening PEF readings and asthma symptoms. MEASUREMENTS AND MAIN RESULTS: Spirometry testing and measurement of serum estradiol and biomarkers of airway inflammation were performed on days 8 (follicular phase), 22 (luteal phase), and 28 (premenstrually) of both the estradiol and placebo cycles. During the two premenstrual visits, the Asthma Quality of Life Questionnaire was administered. No notable differences were observed between the estradiol and placebo cycles in daily PEF recordings or composite asthma symptoms scores. The area under the curve (AUC) for the composite asthma symptoms versus time profile was numerically, but not statistically, lower (denoting less severe symptoms) during the estradiol cycle than during the placebo cycle. Likewise, no significant difference in AUC values for morning PEF or evening PEF was found between the estradiol cycle and the placebo cycle. Despite differences (p<0.05) in day-28 estradiol concentrations for estradiol and placebo cycles, no significant differences were found in forced expiratory volume in 1 second, serum endothelin-1, serum and urine eosinophil protein X, urine leukotriene E4, or quality-of-life scores. CONCLUSION: Exogenously administered estradiol did not have a significant effect in women with premenstrual asthma whose asthma was classified predominantly as mild and under excellent control. As in the case of premenstrual syndrome, the placebo effect may be prominent in premenstrual asthma. Further trials, involving women with more severe asthma under poorer control, are warranted to discern underlying mechanisms for the worsening of asthma in relation to menstruation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Asthma/drug therapy , Estradiol/therapeutic use , Menstrual Cycle , Adult , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/blood , Antioxidants/administration & dosage , Antioxidants/analysis , Biomarkers/analysis , Creatinine/urine , Cross-Over Studies , Double-Blind Method , Endothelin-1/blood , Eosinophil-Derived Neurotoxin , Estradiol/administration & dosage , Estradiol/blood , Female , Humans , Leukotriene E4/urine , Progesterone/blood , Quality of Life , Ribonucleases/blood , Ribonucleases/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...