Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Ann Am Thorac Soc ; 21(2): 261-270, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37962905

RESUMO

Rationale: Bronchiectasis is a chronic, progressive disease of bronchial dilation, inflammation, and scarring leading to impaired mucociliary clearance and increased susceptibility to infection. Identified causes include previous severe respiratory infections. A small, single-center UK study demonstrated a reduction in bronchiectasis exacerbations during the first year of the coronavirus disease (COVID-19) pandemic. No studies have been conducted in a U.S. (commercially insured) cohort to date. Objectives: To explore the impact of the COVID-19 pandemic on the frequency of exacerbations in a large cohort of commercially insured U.S. patients with bronchiectasis by testing the hypothesis that U.S. patients with bronchiectasis had fewer exacerbations during the pandemic. Methods: This retrospective observational cohort study used health insurance claims data from Optum's deidentified Clinformatics Data Mart database, which included U.S. patients and their covered dependents. Eligible patients were ⩾18 years of age with bronchiectasis; patients with other respiratory conditions were excluded. The main study cohort excluded patients with frequent asthma and/or chronic obstructive pulmonary disease diagnoses. The primary objective was to compare the bronchiectasis exacerbation rates before and during the COVID-19 pandemic. Results: The median number of exacerbations per patient per year decreased significantly from the year before the COVID-19 pandemic to the first year of the pandemic (1 vs. 0; P < 0.01). More patients had zero exacerbations during the first year of the pandemic than the year prior (57% vs. 24%; McNemar's chi-square = 122.56; P < 0.01). Conclusions: In a U.S. population-based study of patients with International Classification of Diseases codes for bronchiectasis, the rate of exacerbations during Year 1 of the COVID-19 pandemic was reduced compared with the 2-year time period preceding the pandemic.


Assuntos
Bronquiectasia , COVID-19 , Seguro , Humanos , Estudos Retrospectivos , Pandemias , COVID-19/epidemiologia , Bronquiectasia/epidemiologia , Progressão da Doença
2.
J Med Chem ; 66(20): 14188-14207, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37797307

RESUMO

Histone deacetylase 6 (HDAC6) is a unique member of the HDAC family mainly targeting cytosolic nonhistone substrates, such as α-tubulin, cortactin, and heat shock protein 90 to regulate cell proliferation, metastasis, invasion, and mitosis in tumors. We describe the identification and characterization of a series of 2-(difluoromethyl)-1,3,4-oxadiazoles (DFMOs) as selective nonhydroxamic acid HDAC6 inhibitors. By comparing structure-activity relationships and performing quantum mechanical calculations of the HDAC6 catalytic mechanism, we show that potent oxadiazoles are electrophilic substrates of HDAC6 and propose a mechanism for the bioactivation. We also observe that the inherent electrophilicity of the oxadiazoles makes them prone to degradation in water solution and the generation of potentially toxic products cannot be ruled out, limiting the developability for chronic diseases. However, the oxadiazoles demonstrate high oral bioavailability and low in vivo clearance and are excellent tools for studying the role of HDAC6 in vitro and in vivo in rats and mice.


Assuntos
Neoplasias , Oxidiazóis , Ratos , Camundongos , Animais , Desacetilase 6 de Histona , Oxidiazóis/farmacologia , Tubulina (Proteína)/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/química
3.
PLoS One ; 17(10): e0266310, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36223404

RESUMO

Airway epithelial damage is a common feature in respiratory diseases such as COPD and has been suggested to drive inflammation and progression of disease. These features manifest as remodeling and destruction of lung epithelial characteristics including loss of small airways which contributes to chronic airway inflammation. Histone deacetylase 6 (HDAC6) has been shown to play a role in epithelial function and dysregulation, such as in cilia disassembly, epithelial to mesenchymal transition (EMT) and oxidative stress responses, and has been implicated in several diseases. We thus used ACY-1083, an inhibitor with high selectivity for HDAC6, and characterized its effects on epithelial function including epithelial disruption, cytokine production, remodeling, mucociliary clearance and cell characteristics. Primary lung epithelial air-liquid interface cultures from COPD patients were used and the impacts of TNF, TGF-ß, cigarette smoke and bacterial challenges on epithelial function in the presence and absence of ACY-1083 were tested. Each challenge increased the permeability of the epithelial barrier whilst ACY-1083 blocked this effect and even decreased permeability in the absence of challenge. TNF was also shown to increase production of cytokines and mucins, with ACY-1083 reducing the effect. We observed that COPD-relevant stimulations created damage to the epithelium as seen on immunohistochemistry sections and that treatment with ACY-1083 maintained an intact cell layer and preserved mucociliary function. Interestingly, there was no direct effect on ciliary beat frequency or tight junction proteins indicating other mechanisms for the protected epithelium. In summary, ACY-1083 shows protection of the respiratory epithelium during COPD-relevant challenges which indicates a future potential to restore epithelial structure and function to halt disease progression in clinical practice.


Assuntos
Inibidores de Histona Desacetilases , Doença Pulmonar Obstrutiva Crônica , Citocinas/metabolismo , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Desacetilase 6 de Histona/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Humanos , Inflamação/metabolismo , Pulmão/metabolismo , Mucinas/metabolismo , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/metabolismo , Mucosa Respiratória/metabolismo , Proteínas de Junções Íntimas/metabolismo , Fator de Crescimento Transformador beta/metabolismo
4.
Am J Physiol Lung Cell Mol Physiol ; 323(5): L536-L547, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36098422

RESUMO

Airway dehydration causes mucus stasis and bacterial overgrowth in cystic fibrosis (CF), resulting in recurrent respiratory infections and exacerbations. Strategies to rehydrate airway mucus including inhibition of the epithelial sodium channel (ENaC) have the potential to improve mucosal defense by enhancing mucociliary clearance (MCC) and reducing the risk of progressive decline in lung function. In the current work, we evaluated the effects of AZD5634, an ENaC inhibitor that shows extended lung retention and safety profile as compared with previously evaluated candidate drugs, in healthy and CF preclinical model systems. We found that AZD5634 elicited a potent inhibition of amiloride-sensitive current in non-CF airway cells and airway cells derived from F508del-homozygous individuals with CF that effectively increased airway surface liquid volume and improved mucociliary transport (MCT) rate. AZD5634 also demonstrated efficacious inhibition of ENaC in sheep bronchial epithelial cells, translating to dose-dependent improvement of mucus clearance in healthy sheep in vivo. Conversely, nebulization of AZD5634 did not notably improve airway hydration or MCT in CF rats that exhibit an MCC defect, consistent with findings from a first single-dose evaluation of AZD5634 on MCC in people with CF. Overall, these findings suggest that CF animal models demonstrating impaired mucus clearance translatable to the human situation may help to successfully predict and promote the successful translation of ENaC-directed therapies to the clinic.


Assuntos
Fibrose Cística , Canais Epiteliais de Sódio , Humanos , Ratos , Ovinos , Animais , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Bloqueadores dos Canais de Sódio/uso terapêutico , Amilorida/farmacologia , Depuração Mucociliar/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística , Fibrose Cística/tratamento farmacológico , Mucosa Respiratória
5.
Elife ; 112022 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-35587260

RESUMO

Chronic autoimmune diseases are associated with mutations in PTPN22, a modifier of T cell receptor (TCR) signaling. As with all protein tyrosine phosphatases, the activity of PTPN22 is redox regulated, but if or how such regulation can modulate inflammatory pathways in vivo is not known. To determine this, we created a mouse with a cysteine-to-serine mutation at position 129 in PTPN22 (C129S), a residue proposed to alter the redox regulatory properties of PTPN22 by forming a disulfide with the catalytic C227 residue. The C129S mutant mouse showed a stronger T-cell-dependent inflammatory response and development of T-cell-dependent autoimmune arthritis due to enhanced TCR signaling and activation of T cells, an effect neutralized by a mutation in Ncf1, a component of the NOX2 complex. Activity assays with purified proteins suggest that the functional results can be explained by an increased sensitivity to oxidation of the C129S mutated PTPN22 protein. We also observed that the disulfide of native PTPN22 can be directly reduced by the thioredoxin system, while the C129S mutant lacking this disulfide was less amenable to reductive reactivation. In conclusion, we show that PTPN22 functionally interacts with Ncf1 and is regulated by oxidation via the noncatalytic C129 residue and oxidation-prone PTPN22 leads to increased severity in the development of T-cell-dependent autoimmunity.


Assuntos
Doenças Autoimunes , Linfócitos T , Animais , Dissulfetos/metabolismo , Inflamação/metabolismo , Camundongos , Oxirredução , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo
6.
J Cyst Fibros ; 21(4): 684-690, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35227647

RESUMO

BACKGROUND: Epithelial sodium channel (ENaC) inhibitors may offer clinical benefit in cystic fibrosis (CF); however, data are limited. We report the outcomes of a Phase I (NCT02679729) and a Phase Ib (NCT02950805) study of AZD5634, a novel inhaled ENaC inhibitor. METHODS: A Phase I, first-in-human, single-blind, placebo-controlled, single ascending dose, sequential dose group study assessed the safety, tolerability, and pharmacokinetics of AZD5634 in healthy subjects (n=53) in part A following inhaled doses up to 1700 µg, and, in part B, following administration of single inhaled (1700 µg) and intravenous (65 µg) doses. A Phase Ib, randomized, double-blind, placebo-controlled, single-dose, 2-way cross-over study assessed the effects of a single dose (600 µg) of inhaled AZD5634 on mucociliary clearance (MCC), pharmacokinetics and safety and tolerability in patients with CF (n=11). Nasal potential difference (NPD) was assessed as an in situ target engagement exploratory biomarker. RESULTS: Absolute bioavailability of AZD5634 after inhalation was approximately 3%, indicating minimal distribution into the systemic circulation. Urinary excretion was a minor elimination pathway. Administration of inhaled AZD5634 did not improve MCC in CF patients, but AZD5634 inhibited ENaC in the nasal epithelium, as measured by NPD. AZD5634 was safe and well tolerated in both studies. CONCLUSIONS: AZD5634 showed favorable pharmacokinetics and safety in healthy subjects and patients with CF. However, despite achieving target engagement, proof of mechanism was not achieved after a single dose in patients with CF. Further evaluation into multiple dose studies is warranted to explore its therapeutic potential.


Assuntos
Fibrose Cística , Administração por Inalação , Estudos Cross-Over , Fibrose Cística/diagnóstico , Fibrose Cística/tratamento farmacológico , Método Duplo-Cego , Voluntários Saudáveis , Humanos , Método Simples-Cego
7.
PLoS One ; 16(7): e0254248, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34242292

RESUMO

We have modified the periplasmic Escherichia coli glucose/galactose binding protein (GBP) and labelled with environmentally sensitive fluorophores to further explore its potential as a sensor for the evaluation of glucose concentration in airway surface liquid (ASL). We identified E149C/A213R GBP labelled with N,N'-Dimethyl-N-(iodoacetyl)-N'-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)ethylenediamine (IANBD, emission wavelength maximum 536nm) with a Kd for D-glucose of 1.02mM and a fluorescence dynamic range of 5.8. This sensor was specific for D-glucose and exhibited fluorescence stability in experiments for several hours. The use of E149C/A213R GBP-IANBD in the ASL of airway cells grown at air-liquid-interface (ALI) detected an increase in glucose concentration 10 minutes after raising basolateral glucose from 5 to 15mM. This sensor also reported a greater change in ASL glucose concentration in response to increased basolateral glucose in H441 airway cells compared to human bronchial epithelial cells (HBEC) and there was less variability with HBEC data than that of H441 indicating that HBEC more effectively regulate glucose movement into the ASL. The sensor detected glucose in bronchoalveolar lavage fluid (BALf) from diabetic db/db mice but not normoglycaemic wildtype mice, indicating limited sensitivity of the sensor at glucose concentrations <50µM. Using nasal inhalation of the sensor and spectral unmixing to generate images, E149C/A213R GBP-IANBD fluorescence was detected in luminal regions of cryosections of the murine distal lung that was greater in db/db than wildtype mice. In conclusion, this sensor provides a useful tool for further development to measure luminal glucose concentration in models of lung/airway to explore how this may change in disease.


Assuntos
Técnicas Biossensoriais , Glucose , Animais , Proteínas de Ligação ao Cálcio , Células Epiteliais , Camundongos , Proteínas de Transporte de Monossacarídeos , Proteínas Periplásmicas de Ligação
8.
Eur J Pharmacol ; 904: 174123, 2021 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-33974881

RESUMO

Cystic fibrosis (CF) is a recessive inherited disease caused by mutations affecting anion transport by the epithelial ion channel cystic fibrosis transmembrane conductance regulator (CFTR). The disease is characterized by mucus accumulation in the airways and intestine, but the major cause of mortality in CF is airway mucus accumulation, leading to bacterial colonization, inflammation and respiratory failure. Several drug targets are under evaluation to alleviate airway mucus obstruction in CF and one of these targets is the epithelial sodium channel ENaC. To explore effects of ENaC inhibitors on mucus properties, we used two model systems to investigate mucus characteristics, mucus attachment in mouse ileum and mucus bundle transport in piglet airways. We quantified mucus attachment in explants from CFTR null (CF) mice and tracheobronchial explants from newborn CFTR null (CF) piglets to evaluate effects of ENaC or sodium/hydrogen exchanger (NHE) inhibitors on mucus attachment. ENaC inhibitors detached mucus in the CF mouse ileum, although the ileum lacks ENaC expression. This effect was mimicked by two NHE inhibitors. Airway mucus bundles were immobile in untreated newborn CF piglets but were detached by the therapeutic drug candidate AZD5634 (patent WO, 2015140527). These results suggest that the ENaC inhibitor AZD5634 causes detachment of CF mucus in the ileum and airway via NHE inhibition and that drug design should focus on NHE instead of ENaC inhibition.


Assuntos
Fibrose Cística/tratamento farmacológico , Fibrose Cística/metabolismo , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Canais Epiteliais de Sódio/metabolismo , Pulmão/metabolismo , Muco/metabolismo , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Bicarbonatos/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Canais Epiteliais de Sódio/efeitos dos fármacos , Feminino , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Íleo/efeitos dos fármacos , Íleo/metabolismo , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Muco/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/genética , Suínos
9.
JCI Insight ; 3(17)2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30185674

RESUMO

The respiratory tract is normally kept essentially free of bacteria by cilia-mediated mucus transport, but in chronic obstructive pulmonary disease (COPD) and cystic fibrosis (CF), bacteria and mucus accumulates instead. To address the mechanisms behind the mucus accumulation, the proteome of bronchoalveolar lavages from COPD patients and mucus collected in an elastase-induced mouse model of COPD was analyzed, revealing similarities with each other and with the protein content in colonic mucus. Moreover, stratified laminated sheets of mucus were observed in airways from patients with CF and COPD and in elastase-exposed mice. On the other hand, the mucus accumulation in the elastase model was reduced in Muc5b-KO mice. While mucus plugs were removed from airways by washing with hypertonic saline in the elastase model, mucus remained adherent to epithelial cells. Bacteria were trapped on this mucus, whereas, in non-elastase-treated mice, bacteria were found on the epithelial cells. We propose that the adherence of mucus to epithelial cells observed in CF, COPD, and the elastase-induced mouse model of COPD separates bacteria from the surface cells and, thus, protects the respiratory epithelium.


Assuntos
Bactérias , Células Epiteliais/metabolismo , Muco/microbiologia , Muco/fisiologia , Doença Pulmonar Obstrutiva Crônica/complicações , Animais , Líquido da Lavagem Broncoalveolar , Fibrose Cística/complicações , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Feminino , Humanos , Pulmão , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucina-5B/genética , Elastase Pancreática , Pseudomonas aeruginosa , Mucosa Respiratória
10.
Expert Opin Ther Pat ; 27(2): 145-161, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27774822

RESUMO

INTRODUCTION: Janus kinases (JAKs) are a family of four enzymes; JAK1, JAK2, JAK3 and tyrosine kinase 2 (TYK2) that are critical in cytokine signalling and are strongly linked to both cancer and inflammatory diseases. There are currently two launched JAK inhibitors for the treatment of human conditions: tofacitinib for Rheumatoid arthritis (RA) and ruxolitinib for myeloproliferative neoplasms including intermediate or high risk myelofibrosis and polycythemia vera. Areas covered: This review covers patents claiming activity against one or more JAK family members in the period 2013-2015 inclusive, and covers 95 patents from 42 applicants, split over two parts. The authors have ordered recent patents according to the primary applicant's name, with part 2 covering J through Z. Expert opinion: Inhibition of JAK-family kinases is an area of growing interest, catalysed by the maturity of data on marketed inhibitors ruxolitinib and tofacitinib in late stage clinical trials. Many applicants are pursuing traditional fast-follower strategies around these inhibitors, with a range of chemical strategies adopted. The challenge will be to show sufficient differentiation to the originator compounds, since dose limiting toxicities with such agents appear to be on target and mechanism-related and also considering that such agents may be available as generic compounds by the time follower agents reach market.


Assuntos
Desenho de Fármacos , Janus Quinases/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Animais , Asma/tratamento farmacológico , Asma/enzimologia , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/enzimologia , Relação Dose-Resposta a Droga , Humanos , Patentes como Assunto , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos
11.
Expert Opin Ther Pat ; 27(2): 127-143, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27774824

RESUMO

INTRODUCTION: Janus kinases (JAKs) are a family of four enzymes; JAK1, JAK2, JAK3 and tyrosine kinase 2 (TYK2) that are critical in cytokine signalling and are strongly linked to both cancer and inflammatory diseases. There are currently two launched JAK inhibitors for the treatment of human conditions: tofacitinib for Rheumatoid arthritis (RA) and ruxolitinib for myeloproliferative neoplasms including intermediate or high risk myelofibrosis and polycythemia vera. Areas covered: This review covers patents claiming activity against one or more JAK family members in the period 2013-2015 inclusive, and covers 95 patents from 42 applicants, split over two parts. The authors have ordered recent patents according to the primary applicant's name, with part 1 covering A through to I. Expert opinion: Inhibition of JAK-family kinases is an area of growing interest, catalysed by the maturity of data on marketed inhibitors ruxolitinib and tofacitinib in late stage clinical trials. Many applicants are pursuing traditional fast-follower strategies around these inhibitors, with a range of chemical strategies adopted. The challenge will be to show sufficient differentiation to the originator compounds, since dose limiting toxicities with such agents appear to be on target and mechanism-related and also considering that such agents may be available as generic compounds by the time follower agents reach market.


Assuntos
Desenho de Fármacos , Janus Quinases/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Animais , Relação Dose-Resposta a Droga , Humanos , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Patentes como Assunto , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos
12.
Pharmacol Res Perspect ; 3(3): e00134, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26236482

RESUMO

Although the anti-inflammatory role of the A2a receptor is well established, controversy remains with regard to the therapeutic value for A2a agonists in treatment of inflammatory lung diseases, also as a result of unwanted A2a-mediated cardiovascular effects. In this paper, we describe the discovery and characterization of a new, potent and selective A2a agonist (compound 2) with prolonged lung retention and limited systemic exposure following local administration. To support the lead optimization chemistry program with compound selection and profiling, multiple in vitro and in vivo assays were used, characterizing compound properties, pharmacodynamics (PD), and drug concentrations. Particularly, pharmacokinetic-PD modeling was applied to quantify the effects on the cardiovascular system, and an investigative toxicology study in rats was performed to explore potential myocardial toxicities. Compound 2, in comparison to a reference A2a agonist, UK-432,097, demonstrated higher solubility, lower lipophilicity, lower plasma protein binding, high rat lung retention (28% remaining after 24 h), and was efficacious in a lung inflammatory rat model following intratracheal dosing. Despite these properties, compound 2 did not provide a sufficient therapeutic index, that is, separation of local anti-inflammatory efficacy in the lung from systemic side effects in the cardiovascular system. The plasma concentration that resulted in induction of hypotension (half maximal effective concentration; EC50 0.5 nmol/L) correlated to the in vitro A2a potency (rIC50 0.6 nmol/L). Histopathological lesions in the heart were observed at a dose level which is threefold above the efficacious dose level in the inflammatory rat lung model. In conclusion, compound 2 is a highly potent and selective A2a agonist with significant lung retention after intratracheal administration. Despite its local anti-inflammatory efficacy in rat lung, small margins to the cardiovascular effects suggested limited therapeutic value of this compound for treatment of inflammatory lung disease by the inhaled route.

13.
Am J Physiol Lung Cell Mol Physiol ; 308(1): L22-32, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25361567

RESUMO

Airway dehydration causes mucus stasis and bacterial overgrowth in cystic fibrosis and chronic bronchitis (CB). Rehydration by hypertonic saline is efficacious but suffers from a short duration of action. We tested whether epithelial sodium channel (ENaC) inhibition would rehydrate normal and dehydrated airways to increase mucociliary clearance (MCC) over a significant time frame. For this, we used a tool compound (Compound A), which displays nanomolar ENaC affinity and retention in the airway surface liquid (ASL). Using normal human bronchial epithelial cultures (HBECs) grown at an air-liquid interface, we evaluated in vitro potency and efficacy using short-circuit current (I(sc)) and ASL height measurements where it inhibited I(sc) and increased ASL height by ∼ 50% (0.052 µM at 6 h), respectively. The in vivo efficacy was investigated in a modified guinea pig tracheal potential difference model, where we observed an effective dose (ED50) of 5 µg/kg (i.t.), and by MCC measures in rats and sheep, where we demonstrated max clearance rates at 100 µg/kg (i.t.) and 75 µg/kg (i.t.), respectively. Acute cigarette smoke-induced ASL height depletion in HBECs was used to mimic the situation in patients with CB, and pretreatment prevented both cigarette smoke-induced ASL dehydration and lessened the decrease in ciliary beat frequency. Furthermore, when added after cigarette smoke exposure, Compound A increased the rate of ASL rehydration. In conclusion, Compound A demonstrated significant effects and a link between increased airway hydration, ciliary function, and MCC. These data support the hypothesis that ENaC inhibition may be efficacious in the restoration of mucus hydration and transport in patients with CB.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Muco/metabolismo , Mucosa Respiratória/metabolismo , Fumar/metabolismo , Animais , Transporte Biológico Ativo , Células Cultivadas , Cílios/metabolismo , Cílios/patologia , Feminino , Cobaias , Humanos , Ratos , Ratos Wistar , Mucosa Respiratória/patologia , Ovinos , Fumar/efeitos adversos , Fumar/patologia
14.
Biochem Biophys Res Commun ; 318(4): 964-9, 2004 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-15147966

RESUMO

It is well recognized that the hypothalamus is of central importance in the regulation of food intake and fat mass. Recent studies indicate that it also plays an important role in the regulation of bone mass. Melanin concentrating hormone (MCH) is highly expressed in the hypothalamus and has been implicated in regulation of energy homeostasis. We developed MCHR1 inactivated mice to evaluate the physiological role of this receptor. Interestingly, the MCHR1(-/-) mice have osteoporosis, caused by a reduction in the cortical bone mass, while the amount of trabecular bone is unaffected. The reduction in cortical bone mass is due to decreased cortical thickness. Serum levels of c-telopeptide, a marker of bone resorption, are increased in MCHR1(-/-) mice, indicating that the MCHR1(-/-) mice have a high bone turnover osteoporosis. In conclusion, the MCHR1(-/-) mice have osteoporosis, indicating that MCHR1-signalling is involved in a tonic stimulation of bone mass.


Assuntos
Osteoporose/metabolismo , Receptores do Hormônio Hipofisário/deficiência , Receptores do Hormônio Hipofisário/fisiologia , Absorciometria de Fóton , Animais , Biomarcadores/sangue , Densidade Óssea/fisiologia , Reabsorção Óssea/sangue , Colágeno/sangue , Colágeno Tipo I , Feminino , Fêmur/química , Fêmur/diagnóstico por imagem , Fêmur/patologia , Componentes do Gene , Expressão Gênica , Hormônios Hipotalâmicos/metabolismo , Masculino , Melaninas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteocalcina/sangue , Osteoporose/sangue , Osteoporose/genética , Peptídeos/sangue , Hormônios Hipofisários/metabolismo , RNA Mensageiro/biossíntese , Receptores do Hormônio Hipofisário/genética , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA