Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Mhealth ; 10: 6, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38323145

RESUMEN

Depression is more common in youth with type 1 diabetes (T1D) compared to youth without diabetes. This study aims to assess the efficacy of Competent Adulthood Transition with Cognitive Humanistic and Interpersonal Teaching (CATCH-IT), an internet-based cognitive behavioral therapy (CBT) intervention, in adolescents with T1D and depressive symptoms. Adolescents (13 to 17 years old) with T1D and mild (score 5-9) or moderate (score 10-14) depressive symptoms on Patient Health Questionnaire-Adolescent (PHQ-A) screening assessment were recruited to participate and received online access to the CATCH-IT modules for 6 months (requested to complete in 12 weeks). Statistical analyses included paired t-test for changes in Center for Epidemiologic Studies Depression Scale (CES-D), PHQ-A, Problem Areas in Diabetes-Teen version (PAID-T), and hemoglobin A1c (HbA1c). Nineteen patients were consented, 15 met inclusion criteria and received the intervention. In the seven participants that completed the modules, there was a trend towards improvements in PHQ-A, CES-D and HbA1c. Participants provided robust qualitative feedback on the modules and areas for improvement in subsequent iterations, such as inclusion of diabetes-related content. Given the prevalence of depression in diabetes, feasible, low resource interventions are needed. Internet programs such as CATCH-IT can serve as an effective first line intervention in this high-risk population. A modified version of CATCH-IT tailored for adolescents with T1D may be beneficial in this patient population.

3.
J Emerg Trauma Shock ; 16(1): 26-28, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37181738

RESUMEN

Trauma to the adrenal glands is very rare. The variation in clinical manifestations is marked and markers for its diagnosis being limited, makes it tough to be diagnosed. Computed tomography remains the gold standard for detecting this injury. Prompt recognition and the potential for mortality with adrenal insufficiency can provide the best guidance for the treatment and care of the severely injured. We present a case of a 33-year-old trauma patient who was not responding to the management of his shock. He was finally found to have a right adrenal haemorrhage leading to adrenal crisis. The patient was resuscitated in the Emergency Department but succumbed 10 days post admission.

4.
Reumatologia ; 61(1): 80-81, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36998576
5.
Cureus ; 14(4): e24625, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35664407

RESUMEN

Randomized controlled trials (RCTs) have reported conflicting outcomes with the use of vitamin D in critically ill patients. With reporting of newer RCTs, we conducted this updated meta-analysis. Electronic databases were searched for RCTs comparing vitamin D with placebo in critically ill patients admitted to the intensive care unit (ICU). A random-effects meta-analysis was used to calculate the risk ratio (RR) and standardized mean difference (SMD) with a 95% confidence interval (CI). Eleven RCTs with a total of 2,187 patients (vitamin D: n = 1,120; placebo: n = 1,067) were included. Vitamin D when compared to placebo was associated with the decreased duration of mechanical ventilation (SMD = -0.50; 95% CI = [-0.97, -0.03]; p = 0.04) and ICU stay (SMD = -0.60; 95% CI = [-1.03, -0.16]; p = 0.007) without any difference in the mortality (RR = 0.85; 95% CI = [0.68, 1.04]; p = 0.12) and length of hospital stay (SMD = -0.21; 95% CI = (-0.51, 0.09); p = 0.18]. Subgroup analysis showed that parenteral vitamin D may reduce the risk of mortality (RR = 0.54; 95% CI = [0.35, 0.83], p = 0.005). Vitamin D supplementation in critically ill patients decreases the duration of mechanical ventilation and ICU stay. Further studies should identify specific groups of patients who will derive the most benefit from vitamin D supplementation.

6.
Clin Cancer Res ; 28(15): 3256-3267, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35583555

RESUMEN

PURPOSE: Despite promising activity in hematopoietic malignancies, efficacy of the B-cell lymphoma 2 (BCL2) inhibitor venetoclax in solid tumors is unknown. We report the prespecified VERONICA primary results, a randomized phase II clinical trial evaluating venetoclax and fulvestrant in estrogen receptor (ER)-positive, HER2-negative metastatic breast cancer, post-cyclin-dependent kinase (CDK) 4/6 inhibitor progression. PATIENTS AND METHODS: Pre-/postmenopausal females ≥18 years were randomized 1:1 to venetoclax (800 mg orally daily) plus fulvestrant (500 mg intramuscular; cycle 1: days 1 and 15; subsequent 28-day cycles: day 1) or fulvestrant alone. The primary endpoint was clinical benefit rate (CBR); secondary endpoints were progression-free survival (PFS), overall survival, and safety. Exploratory biomarker analyses included BCL2 and BCL extra-large (BCLXL) tumor expression, and PIK3CA circulating tumor DNA mutational status. RESULTS: At primary analysis (cutoff: August 5, 2020; n = 103), venetoclax did not significantly improve CBR [venetoclax plus fulvestrant: 11.8% (n = 6/51; 95% confidence interval (CI), 4.44-23.87); fulvestrant: 13.7% (7/51; 5.70-26.26); risk difference -1.96% (95% CI, -16.86 to 12.94)]. Median PFS was 2.69 months (95% CI, 1.94-3.71) with venetoclax plus fulvestrant versus 1.94 months (1.84-3.55) with fulvestrant (stratified HR, 0.94; 95% CI, 0.61-1.45; P = 0.7853). Overall survival data were not mature. A nonsignificant improvement of CBR and PFS was observed in patients whose tumors had strong BCL2 expression (IHC 3+), a BCL2/BCLXL Histoscore ratio ≥1, or PIK3CA-wild-type status. CONCLUSIONS: Our findings do not indicate clinical utility for venetoclax plus fulvestrant in endocrine therapy-resistant, CDK4/6 inhibitor-refractory metastatic breast tumors, but suggest possible increased dependence on BCLXL in this setting.


Asunto(s)
Neoplasias de la Mama , Inhibidores de Proteínas Quinasas , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Compuestos Bicíclicos Heterocíclicos con Puentes , Fosfatidilinositol 3-Quinasa Clase I/genética , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Femenino , Fulvestrant/uso terapéutico , Humanos , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2 , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Sulfonamidas
7.
Breast ; 64: 151-158, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35487845

RESUMEN

AIM: To report the final results of the 5-year follow-up of the non-randomized SafeHER Phase III study (NCT01566721) describing the safety, tolerability, and efficacy of subcutaneous (SC) trastuzumab alone and in combination with concurrent or sequential chemotherapy. METHODS: Patients with human epidermal growth factor receptor 2 (HER2)-positive early breast cancer (EBC) with no prior anti-HER2 therapy were included. SC trastuzumab was administered every 3 weeks for 18 cycles as adjuvant therapy with or without chemotherapy (concurrent or sequential). The primary objective was overall safety and tolerability of SC trastuzumab; efficacy was a secondary objective. RESULTS: No new safety signals were observed during the final evaluation. The majority of adverse events (AEs) were grade 1 or 2 across the chemotherapy subgroups. Treatment discontinuation due to AEs was 5.1% for the intent-to-treat (ITT) population and similar for all chemotherapy subgroups. The overall disease-free survival (DFS) 5-year event-free rate in the ITT population (n = 2573) was 86.6% (95% CI, 85.2%-87.9%) with a median follow-up of 72 months. Based on chemotherapy timing, the no (n = 235), concurrent (n = 1533), and sequential (n = 805) chemotherapy subgroups had DFS 5-year event-free rates (95% CI) of 88.5% (83.4%-92.2%), 88.4% (86.6%-89.9%), and 82.6 (79.7%-85.2%), respectively. CONCLUSIONS: The 5-year follow-up analysis of the SafeHER trial demonstrating that SC trastuzumab has an acceptable safety profile, including cardiac toxicity, and efficacy for the treatment of HER2-positive EBC with and without chemotherapy, corresponding with historical data with trastuzumab.


Asunto(s)
Neoplasias de la Mama , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/metabolismo , Quimioterapia Adyuvante , Ensayos Clínicos Fase III como Asunto , Supervivencia sin Enfermedad , Femenino , Humanos , Inyecciones Subcutáneas , Receptor ErbB-2/metabolismo , Trastuzumab/uso terapéutico
8.
J R Coll Physicians Edinb ; 51(4): 369-372, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34882136

RESUMEN

Infectious and autoimmune diseases are distinct entities that require opposite therapeutic approaches. However, differentiating between the two can be a challenge, especially when the histopathology misguides the clinician. We highlight the case of a 66-year-old female who presented with fever, shortness of breath and cough for three months duration. She had raised inflammatory markers, and imaging revealed bilateral cavitating nodules. Histopathology revealed necrotising granulomatous inflammation along with acid-fast bacilli on Ziehl-Neelsen staining. The patient was diagnosed with tuberculosis and started on anti-tubercular therapy. When no response was seen, a rheumatologist was consulted, and a suspected diagnosis of granulomatosis with polyangiitis was made on the basis of clinical and laboratory features. Dramatic response to steroids and a negative mycobacteria culture confirmed the diagnosis. The patient responded to a combination of steroids and cyclophosphamide. This case highlights the importance of recognising the possibility of a false-positive acid-fast bacilli report.


Asunto(s)
Tuberculosis , Anciano , Tos , Ciclofosfamida , Femenino , Humanos
9.
Cancer Treat Res Commun ; 28: 100439, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34333246

RESUMEN

The understanding of cancer biology and the identification of various molecular pathways and targeted oncogenic drivers have led to a paradigm shift in treatment of non-small cell lung cancer. In the last two decades, the therapeutic approach for non-small cell lung cancer (NSCLC) has gradually transitioned from empiric treatment with chemotherapeutic regimens to personalized medicine with precision targets. The major key players in these novel approaches involve targeted therapy, such as tyrosine kinase inhibitors (TKI) and immunotherapy, such as immune checkpoint inhibitors (ICI) blocking intrinsic down regulators of immunity, to achieve anti-cancer effects. These novel agents are generally better tolerated than chemotherapeutics and it is essential to be cognizant of the various drug related adverse effects. Regular follow up of patients with NSCLC by chest computed tomography (CT) surveillance to monitor for disease progression or recurrence is a prerequisite. It is becoming increasingly challenging to identify pulmonary complications related to the use of novel TKI and ICI. Our review focuses on various pulmonary complications of TKI and ICI in patients undergoing treatment for NSCLC, chest CT manifestations, management strategies, and treatment outcomes described in various case reports and case series.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma de Pulmón de Células no Pequeñas/complicaciones , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/efectos adversos , Humanos
10.
Clin Breast Cancer ; 21(6): 539-551, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34154926

RESUMEN

BACKGROUND: Despite advances, there continues to be unmet need in breast cancer. Combining anti-programmed death-ligand 1 (PD-L1) cancer immunotherapy atezolizumab with other targeted therapies may enhance T-cell-dependent cytolytic antitumor activity. METHODS: This open-label, phase Ib study evaluated the safety of atezolizumab-based combinations with antibody-dependent cellular cytotoxicity or antibody-drug conjugate (ADC) agents. Patients with unresectable human epidermal growth factor receptor 2-positive (HER2+) locally advanced or metastatic breast cancer (mBC) received atezolizumab with trastuzumab/pertuzumab, atezolizumab with the ADC ado-trastuzumab emtansine (T-DM1), or atezolizumab with trastuzumab/pertuzumab and docetaxel. In an early-breast cancer (eBC) "window of opportunity" study, patients with operable HER2+ locally advanced or inflammatory eBC received neoadjuvant atezolizumab with trastuzumab/pertuzumab or atezolizumab/T-DM1, followed by docetaxel, carboplatin, and trastuzumab/pertuzumab. Exploratory outcomes included tumor response and biomarkers. RESULTS: By March 15, 2019, 73 patients were enrolled. Safety findings were consistent with the treatment components' individual profiles. Objective responses were observed in 2 of 6 and 5 of 14 patients in 2 mBC cohorts receiving atezolizumab/T-DM1 and in 6 of 6 patients with mBC receiving atezolizumab, trastuzumab/pertuzumab, and docetaxel. PD-L1 in immune cells was the only biomarker that increased with atezolizumab/T-DM1. In the window of opportunity cohorts, PD-L1 levels and CD8+ T-cell infiltration increased from baseline in HER2+ eBC tumors receiving atezolizumab with trastuzumab/pertuzumab or T-DM1, irrespective of response. Despite increases in T-cell and B-cell gene signatures with trastuzumab/pertuzumab, but not T-DM1, neither combination promoted T-cell receptor clonal expansion. CONCLUSION: Atezolizumab with antibody-dependent cellular cytotoxicity or ADC agents appears safe and may activate the adaptive immune system of patients with HER2+ eBC tumors more than those with mBC tumors.


Asunto(s)
Ado-Trastuzumab Emtansina/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Microambiente Tumoral , Adulto , Neoplasias de la Mama/patología , Docetaxel/uso terapéutico , Femenino , Humanos , Persona de Mediana Edad , Terapia Neoadyuvante , Receptor ErbB-2/metabolismo , Taxoides/uso terapéutico , Trastuzumab/uso terapéutico
11.
Reumatologia ; 59(6): 420-422, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35079187

RESUMEN

Post-vaccination inflammatory myositis is a rare but known entity in the literature. We encountered a 46-year-old female patient, who presented with complains of fever, arthralgia, and weakness 1 week after taking the second dose of COVID-19 (Oxford-AstraZeneca) vaccine. On workup the patient had raised inflammatory markers, evidence of myositis on magnetic resonance imaging of thighs, and evidence of interstitial lung disease on high-resolution computed tomography of the chest. The patient was further found to be positive for anti-Jo-1 antibody. The initial treatment was glucocorticosteroids and methotrexate initially. The patient briefly developed pneumocystis pneumonia and recovered. The treatment was switched to mycophenolate mofetil with good response. We presented the first case of anti-Jo-1 syndrome reported following COVID-19 vaccination in the literature. Our aim is to sensitise the clinicians to such rare but occasionally life-threatening complications that may arise in the post-vaccination period.

12.
13.
FASEB J ; 33(3): 3562-3574, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30462529

RESUMEN

Fatty acid binding protein 4 (FABP4), an intracellular lipid chaperone and adipokine, is expressed by lung macrophages, but the function of macrophage-FABP4 remains elusive. We investigated the role of FABP4 in host defense in a murine model of Pseudomonas aeruginosa pneumonia. Compared with wild-type (WT) mice, FABP4-deficient (FABP4-/-) mice exhibited decreased bacterial clearance and increased mortality when challenged intranasally with P. aeruginosa. These findings in FABP4-/- mice were associated with a delayed neutrophil recruitment into the lungs and were followed by greater acute lung injury and inflammation. Among leukocytes, only macrophages expressed FABP4 in WT mice with P. aeruginosa pneumonia. Chimeric FABP4-/- mice with WT bone marrow were protected from increased mortality seen in chimeric WT mice with FABP4-/- bone marrow during P. aeruginosa pneumonia, thus confirming the role of macrophages as the main source of protective FABP4 against that infection. There was less production of C-X-C motif chemokine ligand 1 (CXCL1) in FABP4-/- alveolar macrophages and lower airway CXCL1 levels in FABP4-/- mice. Delivering recombinant CXCL1 to the airways protected FABP4-/- mice from increased susceptibility to P. aeruginosa pneumonia. Thus, macrophage-FABP4 has a novel role in pulmonary host defense against P. aeruginosa infection by facilitating crosstalk between macrophages and neutrophils via regulation of macrophage CXCL1 production.-Liang, X., Gupta, K., Rojas Quintero, J., Cernadas, M., Kobzik, L., Christou, H., Pier, G. B., Owen, C. A., Çataltepe, S. Macrophage FABP4 is required for neutrophil recruitment and bacterial clearance in Pseudomonas aeruginosa pneumonia.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/inmunología , Macrófagos Alveolares/inmunología , Neutrófilos/inmunología , Neumonía/inmunología , Pseudomonas aeruginosa/inmunología , Lesión Pulmonar Aguda/inmunología , Animales , Médula Ósea/inmunología , Quimiocina CXCL1/inmunología , Inflamación/inmunología , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/inmunología , Infecciones por Pseudomonas/inmunología
14.
Am J Respir Crit Care Med ; 198(10): 1254-1267, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29750543

RESUMEN

RATIONALE: ADAM8 (a disintegrin and metalloproteinase domain-8) is expressed by leukocytes and epithelial cells in health, but its contribution to the pathogenesis of chronic obstructive pulmonary disease (COPD) is unknown. OBJECTIVES: To determine whether the expression of ADAM8 is increased in the lungs of patients with COPD and cigarette smoke (CS)-exposed mice, and whether ADAM8 promotes the development of COPD. METHODS: ADAM8 levels were measured in lung, sputum, plasma, and/or BAL fluid samples from patients with COPD, smokers, and nonsmokers, and wild-type (WT) mice exposed to CS versus air. COPD-like lung pathologies were compared in CS-exposed WT versus Adam8-/- mice. MEASUREMENTS AND MAIN RESULTS: ADAM8 immunostaining was reduced in macrophages, and alveolar and bronchial epithelial cells in the lungs of patients with COPD versus control subjects, and CS- versus air-exposed WT mice. ADAM8 levels were similar in plasma, sputum, and BAL fluid samples from patients with COPD and control subjects. CS-exposed Adam8-/- mice had greater airspace enlargement and airway mucus cell metaplasia than WT mice, but similar small airway fibrosis. CS-exposed Adam8-/- mice had higher lung macrophage counts, oxidative stress levels, and alveolar septal cell death rates, but lower alveolar septal cell proliferation rates and soluble epidermal growth factor receptor BAL fluid levels than WT mice. Adam8 deficiency increased lung inflammation by reducing CS-induced activation of the intrinsic apoptosis pathway in macrophages. Human ADAM8 proteolytically shed the epidermal growth factor receptor from bronchial epithelial cells to reduce mucin expression in vitro. Adam8 bone marrow chimera studies revealed that Adam8 deficiency in leukocytes and lung parenchymal cells contributed to the exaggerated COPD-like disease in Adam8-/- mice. CONCLUSIONS: Adam8 deficiency increases CS-induced lung inflammation, emphysema, and airway mucus cell metaplasia. Strategies that increase or prolong ADAM8's expression in the lung may have therapeutic efficacy in COPD.


Asunto(s)
Proteínas ADAM/genética , Antígenos CD/genética , Proteínas de la Membrana/genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Anciano , Animales , Fumar Cigarrillos/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad
15.
Proc Natl Acad Sci U S A ; 113(32): E4681-7, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27444019

RESUMEN

Genetic variants in Hedgehog interacting protein (HHIP) have consistently been associated with the susceptibility to develop chronic obstructive pulmonary disease and pulmonary function levels, including the forced expiratory volume in 1 s (FEV1), in general population samples by genome-wide association studies. However, in vivo evidence connecting Hhip to age-related FEV1 decline and emphysema development is lacking. Herein, using Hhip heterozygous mice (Hhip(+/-)), we observed increased lung compliance and spontaneous emphysema in Hhip(+/-) mice starting at 10 mo of age. This increase was preceded by increases in oxidative stress levels in the lungs of Hhip(+/-) vs. Hhip(+/+) mice. To our knowledge, these results provide the first line of evidence that HHIP is involved in maintaining normal lung function and alveolar structures. Interestingly, antioxidant N-acetyl cysteine treatment in mice starting at age of 5 mo improved lung function and prevented emphysema development in Hhip(+/-) mice, suggesting that N-acetyl cysteine treatment limits the progression of age-related emphysema in Hhip(+/-) mice. Therefore, reduced lung function and age-related spontaneous emphysema development in Hhip(+/-) mice may be caused by increased oxidative stress levels in murine lungs as a result of haploinsufficiency of Hhip.


Asunto(s)
Proteínas Portadoras/genética , Enfisema/etiología , Haploinsuficiencia , Glicoproteínas de Membrana/genética , Acetilcisteína/farmacología , Factores de Edad , Animales , Glutatión/metabolismo , Gutatión-S-Transferasa pi/fisiología , Pulmón/patología , Pulmón/fisiología , Rendimiento Pulmonar , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo
16.
Am J Respir Crit Care Med ; 194(2): 185-97, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-26862784

RESUMEN

RATIONALE: A genetic locus within the FAM13A gene has been consistently associated with chronic obstructive pulmonary disease (COPD) in genome-wide association studies. However, the mechanisms by which FAM13A contributes to COPD susceptibility are unknown. OBJECTIVES: To determine the biologic function of FAM13A in human COPD and murine COPD models and discover the molecular mechanism by which FAM13A influences COPD susceptibility. METHODS: Fam13a null mice (Fam13a(-/-)) were generated and exposed to cigarette smoke. The lung inflammatory response and airspace size were assessed in Fam13a(-/-) and Fam13a(+/+) littermate control mice. Cellular localization of FAM13A protein and mRNA levels of FAM13A in COPD lungs were assessed using immunofluorescence, Western blotting, and reverse transcriptase-polymerase chain reaction, respectively. Immunoprecipitation followed by mass spectrometry identified cellular proteins that interact with FAM13A to reveal insights on FAM13A's function. MEASUREMENTS AND MAIN RESULTS: In murine and human lungs, FAM13A is expressed in airway and alveolar type II epithelial cells and macrophages. Fam13a null mice (Fam13a(-/-)) were resistant to chronic cigarette smoke-induced emphysema compared with Fam13a(+/+) mice. In vitro, FAM13A interacts with protein phosphatase 2A and recruits protein phosphatase 2A with glycogen synthase kinase 3ß and ß-catenin, inducing ß-catenin degradation. Fam13a(-/-) mice were also resistant to elastase-induced emphysema, and this resistance was reversed by coadministration of a ß-catenin inhibitor, suggesting that FAM13A could increase the susceptibility of mice to emphysema development by inhibiting ß-catenin signaling. Moreover, human COPD lungs had decreased protein levels of ß-catenin and increased protein levels of FAM13A. CONCLUSIONS: We show that FAM13A may influence COPD susceptibility by promoting ß-catenin degradation.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , beta Catenina/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estabilidad Proteica , Transducción de Señal , beta Catenina/genética , beta Catenina/fisiología
17.
Genome Med ; 7(1): 12, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25763110

RESUMEN

BACKGROUND: The HHIP gene, encoding Hedgehog interacting protein, has been implicated in chronic obstructive pulmonary disease (COPD) by genome-wide association studies (GWAS), and our subsequent studies identified a functional upstream genetic variant that decreased HHIP transcription. However, little is known about how HHIP contributes to COPD pathogenesis. METHODS: We exposed Hhip haploinsufficient mice (Hhip (+/-) ) to cigarette smoke (CS) for 6 months to model the biological consequences caused by CS in human COPD risk-allele carriers at the HHIP locus. Gene expression profiling in murine lungs was performed followed by an integrative network inference analysis, PANDA (Passing Attributes between Networks for Data Assimilation) analysis. RESULTS: We detected more severe airspace enlargement in Hhip (+/-) mice vs. wild-type littermates (Hhip (+/+) ) exposed to CS. Gene expression profiling in murine lungs suggested enhanced lymphocyte activation pathways in CS-exposed Hhip (+/-) vs. Hhip (+/+) mice, which was supported by increased numbers of lymphoid aggregates and enhanced activation of CD8+ T cells after CS-exposure in the lungs of Hhip (+/-) mice compared to Hhip (+/+) mice. Mechanistically, results from PANDA network analysis suggested a rewired and dampened Klf4 signaling network in Hhip (+/-) mice after CS exposure. CONCLUSIONS: In summary, HHIP haploinsufficiency exaggerated CS-induced airspace enlargement, which models CS-induced emphysema in human smokers carrying COPD risk alleles at the HHIP locus. Network modeling suggested rewired lymphocyte activation signaling circuits in the HHIP haploinsufficiency state.

18.
Eur Respir J ; 45(6): 1544-56, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25700379

RESUMEN

Club cell secretory protein-16 (CC16) is the major secreted product of airway club cells, but its role in the pathogenesis of chronic obstructive pulmonary disease (COPD) is unclear. We measured CC16 airway expression in humans with and without COPD and CC16 function in a cigarette smoke (CS)-induced COPD murine model. Airway CC16 expression was measured in COPD patients, smokers without COPD and non-smokers. We exposed wildtype (WT) and CC16(-/-)mice to CS or air for up to 6 months, and measured airway CC16 expression, pulmonary inflammation, alveolar septal cell apoptosis, airspace enlargement, airway mucin 5AC (MUC5AC) expression, small airway remodelling and pulmonary function. Smokers and COPD patients had reduced airway CC16 immunostaining that decreased with increasing COPD severity. Exposing mice to CS reduced airway CC16 expression. CC16(-/-) mice had greater CS-induced emphysema, airway remodelling, pulmonary inflammation, alveolar cell apoptosis, airway MUC5AC expression, and more compliant lungs than WT mice. These changes were associated with increased nuclear factor-κB (NF-κB) activation in CC16(-/-) lungs. CS-induced acute pulmonary changes were reversed by adenoviral-mediated over-expression of CC16. CC16 protects lungs from CS-induced injury by reducing lung NF-κB activation. CS-induced airway CC16 deficiency increases CS-induced pulmonary inflammation and injury and likely contributes to the pathogenesis of COPD.


Asunto(s)
Pulmón/metabolismo , FN-kappa B/metabolismo , Nicotiana , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Humo , Fumar/metabolismo , Uteroglobina/genética , Uteroglobina/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Apoptosis , Estudios de Casos y Controles , Caspasa 3/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Técnicas de Sustitución del Gen , Humanos , Pulmón/fisiopatología , Ratones , Ratones Noqueados , Mucina 5AC/metabolismo , Estrés Oxidativo , Fosfolipasas A2 Secretoras/metabolismo , Alveolos Pulmonares/citología , Enfisema Pulmonar/metabolismo , Pruebas de Función Respiratoria , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
19.
J Immunol ; 193(5): 2469-82, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25063875

RESUMEN

A disintegrin and a metalloproteinase domain (ADAM) 9 is known to be expressed by monocytes and macrophages. In this study, we report that ADAM9 is also a product of human and murine polymorphonuclear neutrophils (PMNs). ADAM9 is not synthesized de novo by circulating PMNs. Rather, ADAM9 protein is stored in the gelatinase and specific granules and the secretory vesicles of human PMNs. Unstimulated PMNs express minimal quantities of surface ADAM9, but activation of PMNs with degranulating agonists rapidly (within 15 min) increases PMN surface ADAM9 levels. Human PMNs produce small quantities of soluble forms of ADAM9. Surprisingly, ADAM9 degrades several extracellular matrix (ECM) proteins, including fibronectin, entactin, laminin, and insoluble elastin, as potently as matrix metalloproteinase-9. However, ADAM9 does not degrade types I, III, or IV collagen or denatured collagens in vitro. To determine whether Adam9 regulates PMN recruitment or ECM protein turnover during inflammatory responses, we compared wild-type and Adam9(-/-) mice in bacterial LPS- and bleomycin-mediated acute lung injury (ALI). Adam9 lung levels increase 10-fold during LPS-mediated ALI in wild-type mice (due to increases in leukocyte-derived Adam9), but Adam9 does not regulate lung PMN (or macrophage) counts during ALI. Adam9 increases mortality, promotes lung injury, reduces lung compliance, and increases degradation of lung elastin during LPS- and/or bleomycin-mediated ALI. Adam9 does not regulate collagen accumulation in the bleomycin-treated lung. Thus, ADAM9 is expressed in an inducible fashion on PMN surfaces where it degrades some ECM proteins, and it promotes alveolar-capillary barrier injury during ALI in mice.


Asunto(s)
Proteínas ADAM/inmunología , Lesión Pulmonar Aguda/inmunología , Matriz Extracelular/inmunología , Proteínas de la Membrana/inmunología , Neutrófilos/inmunología , Proteolisis , Proteínas ADAM/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/farmacología , Bleomicina/efectos adversos , Bleomicina/farmacología , Barrera Alveolocapilar/inmunología , Barrera Alveolocapilar/patología , Colágeno/genética , Colágeno/inmunología , Elastina/genética , Elastina/inmunología , Matriz Extracelular/genética , Humanos , Lipopolisacáridos/toxicidad , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Neutrófilos/patología
20.
Am J Respir Cell Mol Biol ; 51(2): 262-72, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24617927

RESUMEN

Elafin (peptidase inhibitor 3 [PI3]) and its biologically active precursor, pre-elafin, are neutrophil serine proteinase inhibitors with an important role in preventing excessive tissue injury during inflammatory events. Recently, we reported an association between single-nucleotide polymorphism (SNP) rs2664581 in the PI3 gene, increased risk of acute respiratory distress syndrome (ARDS) and pre-elafin circulating levels. This study aims to validate the legitimacy of this association by using a cohort of patients who met the criteria for systemic inflammatory response syndrome and were at risk of developing ARDS (n = 840). A comprehensive functional study of SNPs in PI3 gene was also performed. Luciferase assays and electrophoretic mobility shift assays were conducted to determine the functional relevance of promoter region variants. The effect of the coding SNP rs2664581 on the neutrophil elastase inhibitory activity and transglutaminase binding properties of pre-elafin was also investigated. The variant allele of rs2664581 (C) was significantly associated with increased ARDS risk, mainly among subjects with sepsis (odds ratio = 1.44; 95% confidence interval = 1.04-1.99; P = 0.0276, adjusted by age, sex, and Acute Physiology and Chronic Health Evaluation III). Pre-elafin recombinant protein carrying the amino acid change associated with rs2664581 (Thr34Pro, mutant protein [MT]) had greater capacity to undergo transglutaminase-mediated cross-linking to immobilized fibronectin than wild-type protein in vitro (P < 0.003). No differences were observed in the neutrophil elastase inhibitory activities of wild-type versus MT proteins. In addition, the risk allele-promoter construct had significantly lower cytokine-induced transcriptional activity. Electrophoretic mobility shift assay results indicated a differential binding of nuclear proteins to the G and A alleles of SNP -338G > A. Our results confirm the association between SNP rs2664581 and enhanced risk of ARDS, further supporting the role of PI3 in ARDS development. SNPs in the PI3 locus may act synergistically by regulating PI3 gene expression and pre-elafin biological functions.


Asunto(s)
Elafina/genética , Polimorfismo de Nucleótido Simple , Síndrome de Dificultad Respiratoria/genética , Síndrome de Respuesta Inflamatoria Sistémica/genética , Adulto , Anciano , Sitios de Unión , Estudios de Casos y Controles , Línea Celular Tumoral , Citocinas/metabolismo , Elafina/metabolismo , Femenino , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Frecuencia de los Genes , Genes Reporteros , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Elastasa de Leucocito/metabolismo , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Oportunidad Relativa , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica , Síndrome de Dificultad Respiratoria/metabolismo , Factores de Riesgo , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Transcripción Genética , Transfección , Transglutaminasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...