Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nature ; 580(7803): E7, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32296181

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Nature ; 579(7798): 265-269, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32015508

RESUMEN

Emerging infectious diseases, such as severe acute respiratory syndrome (SARS) and Zika virus disease, present a major threat to public health1-3. Despite intense research efforts, how, when and where new diseases appear are still a source of considerable uncertainty. A severe respiratory disease was recently reported in Wuhan, Hubei province, China. As of 25 January 2020, at least 1,975 cases had been reported since the first patient was hospitalized on 12 December 2019. Epidemiological investigations have suggested that the outbreak was associated with a seafood market in Wuhan. Here we study a single patient who was a worker at the market and who was admitted to the Central Hospital of Wuhan on 26 December 2019 while experiencing a severe respiratory syndrome that included fever, dizziness and a cough. Metagenomic RNA sequencing4 of a sample of bronchoalveolar lavage fluid from the patient identified a new RNA virus strain from the family Coronaviridae, which is designated here 'WH-Human 1' coronavirus (and has also been referred to as '2019-nCoV'). Phylogenetic analysis of the complete viral genome (29,903 nucleotides) revealed that the virus was most closely related (89.1% nucleotide similarity) to a group of SARS-like coronaviruses (genus Betacoronavirus, subgenus Sarbecovirus) that had previously been found in bats in China5. This outbreak highlights the ongoing ability of viral spill-over from animals to cause severe disease in humans.


Asunto(s)
Betacoronavirus/clasificación , Enfermedades Transmisibles Emergentes/complicaciones , Enfermedades Transmisibles Emergentes/virología , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/virología , Neumonía Viral/complicaciones , Neumonía Viral/virología , Síndrome Respiratorio Agudo Grave/etiología , Síndrome Respiratorio Agudo Grave/virología , Adulto , Betacoronavirus/genética , COVID-19 , China , Enfermedades Transmisibles Emergentes/diagnóstico por imagen , Enfermedades Transmisibles Emergentes/patología , Infecciones por Coronavirus/diagnóstico por imagen , Infecciones por Coronavirus/patología , Genoma Viral/genética , Humanos , Pulmón/diagnóstico por imagen , Masculino , Filogenia , Neumonía Viral/diagnóstico por imagen , Neumonía Viral/patología , ARN Viral/genética , Recombinación Genética/genética , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/diagnóstico por imagen , Síndrome Respiratorio Agudo Grave/patología , Tomografía Computarizada por Rayos X , Secuenciación Completa del Genoma
3.
Appl Opt ; 58(34): 9370-9375, 2019 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-31873527

RESUMEN

An adjustable mounting structure is proposed to compensate for surface deformation of a mirror caused by the assembly process. The mount adopts a six-point support based on the kinematic mount principle. Three of the support points are adjustable, and they are moved along the axial direction by actuators. Surface deformation is expressed by Zernike coefficients in this paper, and a sensitivity matrix of the surface deformation is established by varying the unit displacement of each adjustment support point and getting the corresponding Zernike coefficient changes. The surface deformation is measured, and the compensation adjustment of each adjustable support point is then obtained by anti-sensitivity calculation. Finally, the feasibility of present support structure design and surface figure compensating method are verified by experiments. The experimental results show that the present structure and method could significantly reduce the surface deformation caused by the assembly process. The surface deformation is 4.6 nm RMS after assembly and it is decreased to 1.3 nm RMS after four iterations of compensation, which is close to the 1.1 nm RMS after optical polishing.

4.
Am J Respir Crit Care Med ; 186(11): 1168-79, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23065014

RESUMEN

RATIONALE: IL-9-producing CD4(+) T cells (Th9 cells) have been reported to be involved in inflammation and immune diseases. However, the involvement of Th9 cells in malignancy has not been investigated. OBJECTIVES: To elucidate the mechanism by which Th9 cells differentiate in malignant pleural effusion (MPE) and to explore the immune regulation of Th9 cells on lung cancer cells. METHODS: Distribution of Th9 cells in relation to Th17 and Th1 cells in both MPE and blood were determined. The effects and mechanisms of proinflammatory cytokines and regulatory T cells on differentiation of Th9 cells in vitro were explored. The impacts and signal transductions of IL-9, IL-17, and IFN-γ on lung cancer cell lines were also investigated. MEASUREMENTS AND MAIN RESULTS: The numbers of Th9, Th17, and Th1 cells were all increased in MPE when compared with blood. The increase in Th9 cells in MPE was due to the promotion by cytokines and regulatory T cells. By activating STAT3 signaling, both IL-9 and IL-17 substantially promoted the proliferation and migratory activity of lung cancer cells, whereas IFN-γ, which activated STAT1 signaling, was noted to suppress lung cancer cell proliferation and migration. IFN-γ could induce lung cancer cell apoptosis. Moreover, IL-9 and IFN-γ, but not IL-17, could strongly facilitate intercellular adhesion of lung cancer cells to pleural mesothelial cell monolayers. CONCLUSIONS: Our data revealed that Th9 cells were increased in MPE and that Th9 cells exerted an important immune regulation on lung cancer cells in human tumor environment.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Neoplasias Pulmonares/inmunología , Derrame Pleural Maligno/inmunología , Células TH1/inmunología , Anciano , Biomarcadores/análisis , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-9/inmunología , Interleucina-9/metabolismo , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Derrame Pleural Maligno/citología , Derrame Pleural Maligno/patología , Pronóstico , Sensibilidad y Especificidad , Transducción de Señal , Células TH1/metabolismo , Células Tumorales Cultivadas
5.
Am J Respir Crit Care Med ; 185(6): 660-9, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22199006

RESUMEN

RATIONALE: IL-22-producing helper T cells (Th22 cells) have been reported to be involved in tuberculosis infection. However, differentiation and immune regulation of Th22 cells in tuberculous pleural effusion (TPE) remain unknown. OBJECTIVES: To elucidate the mechanism by which Th22 cells differentiate and recruit into the pleural space. METHODS: The distribution and phenotypic features of Th22 cells in both TPE and blood were determined. The impacts of proinflammatory cytokines and antigen presentation by pleural mesothelial cells (PMCs) on Th22-cell differentiation were explored. The chemoattractant activity of chemokines produced by PMCs for Th22 cells was observed. MEASUREMENTS AND MAIN RESULTS: Th22 cells were significantly higher in TPE than in blood. IL-1ß, IL-6, and/or tumor necrosis factor-α promoted Th22-cell differentiation from CD4(+) T cells. It was found that PMCs expressed CCL20, CCL22, and CCL27, and that TPE and PMC supernatants were chemotactic for Th22 cells. This activity was partly blocked by anti-CCL20, anti-CCL22, and anti-CCL27 antibodies. IL-22 and IL-17 significantly improved PMC wound healing. Moreover, PMCs were able to stimulate CD4(+) T-cell proliferation and Th22-cell differentiation by presenting tuberculosis-specific antigen. CONCLUSIONS: The overrepresentation of Th22 cells in TPE may be due to pleural cytokines and to PMC-produced chemokines. Our data suggest a collaborative loop between PMCs and Th22 cells in TPE. In particular, PMCs were able to function as antigen-presenting cells to stimulate CD4(+) T-cell proliferation and Th22-cell differentiation.


Asunto(s)
Inmunidad Celular , Interleucinas/biosíntesis , Activación de Linfocitos/inmunología , Pleura/patología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Tuberculosis Pleural/inmunología , Células Presentadoras de Antígenos/metabolismo , Diferenciación Celular , Epitelio/inmunología , Epitelio/metabolismo , Epitelio/patología , Humanos , Pleura/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología , Tuberculosis Pleural/patología , Interleucina-22
6.
Front Plant Sci ; 14: 1276918, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37929165

RESUMEN

Medicinal plants are invaluable resources for mankind and play a crucial role in combating diseases. Arbuscular mycorrhizal fungi (AMF) are widely recognized for enhancing the production of medicinal active ingredients in medicinal plants. However, there is still a lack of comprehensive understanding regarding the quantitative effects of AMF on the accumulation of medicinal active ingredients. Here we conducted a comprehensive global analysis using 233 paired observations to investigate the impact of AMF inoculation on the accumulation of medicinal active ingredients. This study revealed that AMF inoculation significantly increased the contents of medicinal active ingredients by 27%, with a particularly notable enhancement observed in flavonoids (68%) and terpenoids (53%). Furthermore, the response of medicinal active ingredients in belowground organs (32%) to AMF was more pronounced than that in aboveground organs (18%). Notably, the AMF genus Rhizophagus exhibited the strongest effect in improving the contents of medicinal active ingredients, resulting in an increase of over 50% in both aboveground and belowground organs. Additionally, the promotion of medicinal active ingredients by AMF was attributed to improvements in physiological factors, such as chlorophyll, stomatal conductance and net photosynthetic rate. Collectively, this research substantially advanced our comprehension of the pivotal role of AMF in improving the medicinal active ingredients of plants and provided valuable insights into the potential mechanisms driving these enhancements.

7.
Chin Med J (Engl) ; 133(9): 1032-1038, 2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32118640

RESUMEN

BACKGROUND: Since early December 2019, the 2019 novel coronavirus disease (COVID-19) has caused pneumonia epidemic in Wuhan, Hubei province of China. This study aimed to investigate the factors affecting the progression of pneumonia in COVID-19 patients. Associated results will be used to evaluate the prognosis and to find the optimal treatment regimens for COVID-19 pneumonia. METHODS: Patients tested positive for the COVID-19 based on nucleic acid detection were included in this study. Patients were admitted to 3 tertiary hospitals in Wuhan between December 30, 2019, and January 15, 2020. Individual data, laboratory indices, imaging characteristics, and clinical data were collected, and statistical analysis was performed. Based on clinical typing results, the patients were divided into a progression group or an improvement/stabilization group. Continuous variables were analyzed using independent samples t-test or Mann-Whitney U test. Categorical variables were analyzed using Chi-squared test or Fisher's exact test. Logistic regression analysis was performed to explore the risk factors for disease progression. RESULTS: Seventy-eight patients with COVID-19-induced pneumonia met the inclusion criteria and were included in this study. Efficacy evaluation at 2 weeks after hospitalization indicated that 11 patients (14.1%) had deteriorated, and 67 patients (85.9%) had improved/stabilized. The patients in the progression group were significantly older than those in the disease improvement/stabilization group (66 [51, 70] vs. 37 [32, 41] years, U = 4.932, P = 0.001). The progression group had a significantly higher proportion of patients with a history of smoking than the improvement/stabilization group (27.3% vs. 3.0%, χ = 9.291, P = 0.018). For all the 78 patients, fever was the most common initial symptom, and the maximum body temperature at admission was significantly higher in the progression group than in the improvement/stabilization group (38.2 [37.8, 38.6] vs. 37.5 [37.0, 38.4]°C, U = 2.057, P = 0.027). Moreover, the proportion of patients with respiratory failure (54.5% vs. 20.9%, χ = 5.611, P = 0.028) and respiratory rate (34 [18, 48] vs. 24 [16, 60] breaths/min, U = 4.030, P = 0.004) were significantly higher in the progression group than in the improvement/stabilization group. C-reactive protein was significantly elevated in the progression group compared to the improvement/stabilization group (38.9 [14.3, 64.8] vs. 10.6 [1.9, 33.1] mg/L, U = 1.315, P = 0.024). Albumin was significantly lower in the progression group than in the improvement/stabilization group (36.62 ±â€Š6.60 vs. 41.27 ±â€Š4.55 g/L, U = 2.843, P = 0.006). Patients in the progression group were more likely to receive high-level respiratory support than in the improvement/stabilization group (χ = 16.01, P = 0.001). Multivariate logistic analysis indicated that age (odds ratio [OR], 8.546; 95% confidence interval [CI]: 1.628-44.864; P = 0.011), history of smoking (OR, 14.285; 95% CI: 1.577-25.000; P = 0.018), maximum body temperature at admission (OR, 8.999; 95% CI: 1.036-78.147, P = 0.046), respiratory failure (OR, 8.772, 95% CI: 1.942-40.000; P = 0.016), albumin (OR, 7.353, 95% CI: 1.098-50.000; P = 0.003), and C-reactive protein (OR, 10.530; 95% CI: 1.224-34.701, P = 0.028) were risk factors for disease progression. CONCLUSIONS: Several factors that led to the progression of COVID-19 pneumonia were identified, including age, history of smoking, maximum body temperature at admission, respiratory failure, albumin, and C-reactive protein. These results can be used to further enhance the ability of management of COVID-19 pneumonia.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus , Pandemias , Neumonía Viral , Adulto , Anciano , COVID-19 , Femenino , Hospitales , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Factores de Riesgo , SARS-CoV-2
8.
Ann Am Thorac Soc ; 17(7): 839-846, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32255382

RESUMEN

Rationale: The current outbreak of coronavirus disease (COVID-19) pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Wuhan, China, spreads across national and international borders. The overall death rate of COVID-19 pneumonia in the Chinese population was 4%.Objectives: To describe the process of hospitalization and critical care of patients who died of COVID-19 pneumonia.Methods: This was a multicenter observational study of 109 decedents with COVID-19 pneumonia from three hospitals in Wuhan. Demographic, clinical, laboratory, and treatment data were collected and analyzed, and the final date of follow-up was February 24, 2020.Results: The mean age of 109 decedents with COVID-19 pneumonia was 70.7 years, 35 patients (32.1%) were female, and 85 patients (78.0%) suffered from one or more underlying comorbidities. Multiple organ failure, especially respiratory failure and heart failure, appeared in all patients even at the early stage of disease. Overall, the mean time from onset of symptoms to death was 22.3 days. All 109 hospitalized patients needed admission to an intensive care unit (ICU); however, because of limited availability, only 51 (46.8%) could be admitted. The period from hospitalization to death in the ICU group and non-ICU group was 15.9 days (standard deviation = 8.8 d) and 12.5 days (8.6 d, P = 0.044), respectively.Conclusions: Mortality due to COVID-19 pneumonia was concentrated in patients above the age of 65 years, especially those with major comorbidities. Patients who were admitted to the ICU lived longer than those who were not. Our findings should aid in the recognition and clinical management of such infections, especially with regard to ICU resource allocation.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus , Cuidados Críticos/métodos , Insuficiencia Multiorgánica , Pandemias , Neumonía Viral , Insuficiencia Respiratoria , Anciano , Betacoronavirus/aislamiento & purificación , Betacoronavirus/patogenicidad , COVID-19 , China/epidemiología , Comorbilidad , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/fisiopatología , Infecciones por Coronavirus/terapia , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Unidades de Cuidados Intensivos/estadística & datos numéricos , Masculino , Mortalidad , Insuficiencia Multiorgánica/diagnóstico , Insuficiencia Multiorgánica/etiología , Evaluación de Procesos y Resultados en Atención de Salud , Neumonía Viral/diagnóstico , Neumonía Viral/epidemiología , Neumonía Viral/etiología , Neumonía Viral/mortalidad , Neumonía Viral/fisiopatología , Neumonía Viral/terapia , Pronóstico , Insuficiencia Respiratoria/etiología , Insuficiencia Respiratoria/terapia , Medición de Riesgo , Factores de Riesgo , SARS-CoV-2
9.
Indian J Pathol Microbiol ; 60(2): 285-287, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28631658

RESUMEN

Aspergillus tracheobronchitis (AT) is a unique form of invasive pulmonary aspergillosis, which is commonly found in patients with impaired immunity. Early-stage AT presents in a nonspecific way, both clinically and radiographically, thereby delaying diagnosis and resulting in a high mortality. Owing to impaired mucociliary clearance, previous nonfungal infections, and administration of corticosteroids, among other aspects, patients with chronic obstructive pulmonary disease (COPD) are predisposed to AT, although they are mostly immunocompetent. AT in COPD patients has not been well recognized and the condition is often misdiagnosed or missed. We herein report a case of AT diagnosed in a male with past COPD, with the features of pseudomembranous AT upon bronchoscopy. This contradicts the opinion that pseudomembranous AT is found in severely immunocompromised hosts with hematologic malignancies.


Asunto(s)
Aspergilosis/diagnóstico , Aspergilosis/patología , Aspergillus/aislamiento & purificación , Bronquitis/patología , Traqueítis/patología , Anciano , Aspergilosis/microbiología , Bronquitis/microbiología , Broncoscopía , Humanos , Masculino , Traqueítis/microbiología
10.
Tuberculosis (Edinb) ; 94(2): 131-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24406080

RESUMEN

Programmed death 1 (PD-1), PD-ligand 1 (PD-L1), and PD-L2 have been demonstrated to be involved in tuberculosis immunity, however, the expression and regulation of PD-1/PD-Ls pathways in pleural mesothelial cells (PMCs) and CD4(+) T cells in tuberculous pleural effusion (TPE) have not been investigated. Expression of PD-1 on CD4(+) T cells and expressions of PD-L1 and PD-L2 on PMCs in TPE were determined. The impacts of PD-1/PD-Ls pathways on proliferation, apoptosis, adhesion, and migration of CD4(+) T cells were explored. Concentrations of soluble PD-l, but not of soluble PD-Ls, were much higher in TPE than in serum. Expressions of PD-1 on CD4(+) T cells in TPE were significantly higher than those in blood. Expressions of PD-Ls were much higher on PMCs from TPE when compared with those from transudative effusion. Interferon-γ not only upregulated the expression of PD-1 on CD4(+) T cells, but also upregulated the expressions of PD-Ls on PMCs. Blockage PD-1/PD-Ls pathways abolished the inhibitory effects on proliferation and adhesion activity of CD4(+) T cells induced by PMCs. PD-1/PD-Ls pathways on PMCs inhibited proliferation and adhesion activity of CD4(+) T cells, suggesting that Mycobacterium tuberculosis might exploit PD-1/PD-Ls pathways to evade host cell immune response in human.


Asunto(s)
Antígeno B7-H1/inmunología , Linfocitos T CD4-Positivos/inmunología , Interferón gamma/inmunología , Mycobacterium tuberculosis/inmunología , Proteína 2 Ligando de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Tuberculosis Pleural/inmunología , Adulto , Apoptosis , Adhesión Celular , Proliferación Celular , Células Cultivadas , Femenino , Citometría de Flujo , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Activación de Linfocitos , Masculino , Mycobacterium tuberculosis/aislamiento & purificación , Transducción de Señal , Tuberculosis Pleural/fisiopatología , Molécula 1 de Adhesión Celular Vascular/inmunología
11.
PLoS One ; 8(9): e74624, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24069325

RESUMEN

BACKGROUND: Intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) have been demonstrated to be expressed on pleural mesothelial cells (PMCs), and to mediate leukocyte adhesion and migration; however, little is known about whether adhesion molecule-dependent mechanisms are involved in the regulation of CD4(+) T cells by PMCs in tuberculous pleural effusion (TPE). METHODS: Expressions of ICAM-1 and VCAM-1 on PMCs, as well as expressions of CD11a and CD29, the counter-receptors for ICAM-1 and VCAM-1, respectively, expressed on CD4(+) T cells in TPE were determined using flow cytometry. The immune regulations on adhesion, proliferation, activation, selective expansion of CD4(+) helper T cell subgroups exerted by PMCs via adhesion molecule-dependent mechanisms were explored. RESULTS: Percentages of ICAM-1-positive and VCAM-1‒positive PMCs in TPE were increased compared with PMC line. Interferon-γ enhanced fluorescence intensity of ICAM-1, while IL-4 promoted VCAM-1 expression on PMCs. Percentages of CD11a(high)CD4(+) and CD29(high)CD4(+) T cells in TPE significantly increased as compared with peripheral blood. Prestimulation of PMCs with anti‒ICAM-1 or ‒VCAM-1 mAb significantly inhibited adhesion, activation, as well as effector regulatory T cell expansion induced by PMCs. CONCLUSIONS: Our current data showed that adhesion molecule pathways on PMCs regulated adhesion and activation of CD4(+) T cells, and selectively promoted the expansion of effector regulatory T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/metabolismo , Tuberculosis Pleural/inmunología , Tuberculosis Pleural/metabolismo , Antígeno CD11a/metabolismo , Adhesión Celular , Células Cultivadas , Humanos , Integrina beta1/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Activación de Linfocitos/inmunología , Transducción de Señal , Molécula 1 de Adhesión Celular Vascular/metabolismo
12.
Cancer Lett ; 326(1): 23-32, 2012 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-22809567

RESUMEN

Th22 cells have been reported to be involved in human cancers. However, differentiation and immune regulation of Th22 cells in malignant pleural effusion (MPE) remain unknown. We noted that Th22 cell numbers were increased in MPE, and that IL-22 substantially promoted the proliferation and migratory activity of A549 cells. Moreover, IL-22 could strongly facilitate intercellular adhesion of A549 cells to pleural mesothelial cell monolayers. Our data revealed that the increase in Th22 cells in MPE was due to pleural cytokines and chemokines, and that Th22 exerted an important immune regulation on cancer cells in human pleural malignant environment.


Asunto(s)
Interleucinas/metabolismo , Neoplasias Pulmonares/inmunología , Derrame Pleural Maligno/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Adulto , Animales , Adhesión Celular , Diferenciación Celular , Línea Celular Tumoral , Quimiotaxis , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Modelos Animales , Interleucina-22
13.
PLoS One ; 7(2): e31710, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22363712

RESUMEN

Newly discovered IL-9-producing CD4(+) helper T cells (Th9 cells) have been reported to contribute to tissue inflammation and immune responses, however, differentiation and immune regulation of Th9 cells in tuberculosis remain unknown. In the present study, our data showed that increased Th9 cells with the phenotype of effector memory cells were found to be in tuberculous pleural effusion as compared with blood. TGF-ß was essential for Th9 cell differentiation from naïve CD4(+) T cells stimulated with PMA and ionomycin in vitro for 5 h, and addition of IL-1ß, IL-4 or IL-6 further augmented Th9 cell differentiation. Tuberculous pleural effusion and supernatants of cultured pleural mesothelial cells were chemotactic for Th9 cells, and this activity was partly blocked by anti-CCL20 antibody. IL-9 promoted the pleural mesothelial cell repairing and inhibited IFN-γ-induced pleural mesothelial cell apoptosis. Moreover, pleural mesothelial cells promoted Th9 cell differentiation by presenting antigen. Collectively, these data provide new information concerning Th9 cells, in particular the collaborative immune regulation between Th9 cells and pleural mesothelial cells in human M. tuberculosis infection. In particular, pleural mesothelial cells were able to function as antigen-presenting cells to stimulate Th9 cell differentiation.


Asunto(s)
Diferenciación Celular , Células Epiteliales/inmunología , Interleucina-9/inmunología , Activación de Linfocitos/inmunología , Pleura/patología , Linfocitos T Colaboradores-Inductores/inmunología , Tuberculosis/inmunología , Adulto , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Antígenos Bacterianos/inmunología , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Quimiocina CCL20/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Células Epiteliales/patología , Humanos , Interferón gamma/farmacología , Interleucina-4/farmacología , Ionomicina/farmacología , Activación de Linfocitos/efectos de los fármacos , Persona de Mediana Edad , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/fisiología , Fenotipo , Derrame Pleural/inmunología , Derrame Pleural/microbiología , Derrame Pleural/patología , Receptores de Quimiocina/metabolismo , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Tuberculosis/microbiología , Tuberculosis/patología , Cicatrización de Heridas/efectos de los fármacos , Adulto Joven
14.
Journal of Experimental Hematology ; (6): 1003-1010, 2017.
Artículo en Zh | WPRIM | ID: wpr-271878

RESUMEN

<p><b>OBJECTIVE</b>To explore the effect of arsenic trioxide combined with itraconazole on proliferation and apoptosis of KG1a cells and its potential mechanism.</p><p><b>METHODS</b>The cell morphology was observed with Wrighe-Giemsa staining; cell survival rate was examined by CCK-8; and colony formation capacity was measured by methylcellulose colony formation test; the flow cytometry was used to analyse the cell apoptosis rate and cell cycle; the protein expressions of BCL-2,caspase-3,BAX,SMO,Gli1 and Gli2 were detected by Western-blot.</p><p><b>RESULTS</b>The arsenic trioxide and itraconazole alone both could inhibit the KG1a cell proliferation in dose-and time-dependent manner. In comparison between single and combined drug-treatment group, both the cell survival rate and the colony number of the single drug-treatment group were significantly lower(P<0.05), and the apoptosis rate was higher in the combined drug-treatment group. In the combined-treatment group, the protein expression of Caspase-3 and BAX was upregulated, while the protein expression of BCL-2,SMO,Gli1 and Gli2 was downregulated.</p><p><b>CONCLUSION</b>Arsenic trioxide combined with itraconazole can inhibit the KG1a cell proliferation and induce apoptosis, which may be related with the inhibition of Hh signaling pathway and upregulation of both Caspase-3 and BAX protein expression, and provided experimental data of arsenic trioxide combined with itraconazole for the treatment of refractory AML.</p>

15.
Journal of Experimental Hematology ; (6): 1267-1272, 2014.
Artículo en Zh | WPRIM | ID: wpr-340516

RESUMEN

This study was aimed to explore the effect of arsenic trioxide combined with curcumin on proliferation and apoptosis of KG1a cells and its potential mechanism. The cell survival rate was mesured by MTT; colony formation capacity was examined by methylcellulose colony formation test; flow cytometry was used to analyse the cell surface molecules, cell apoptosis rate and cell cycle; the cell morphology was observed with Wright-Giemsa staining and the protein expression of BCL-2, BAX, PARP was detected by Western blot. The results showed that the phenotype of KG1a cells was CD34(+)CD38(-), while the phenotype of HL-60 cell was CD34(+)CD38(+). The former possessed a stronger colony ability than the latter. Effect of curcumin and arsenic trioxide alone on cell proliferation and inhibition was in dose-dependent manner. Compared with single drug-treatment group, the cell survival rate and colony number were lower, and the apoptosis rate was higher in combined drug-treatment group. Protein expression of BCL-2 and PARP was upregulated, while the protein expression of PARP was downregulated in the combined treatment group. It is concluded that compared with HL-60 cells, KG1a cells are the earlier leukemia stem/progenitor cells. Arsenic trioxide combined with curcumin can effectively inhibit the KG1a cell proliferation and induce apoptosis, which may be associated with the downregulation of BCL-2 and PARP protein expression and the upregulation of BAX protein expression.


Asunto(s)
Humanos , Apoptosis , Arsenicales , Farmacología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Curcumina , Farmacología , Óxidos , Farmacología , Proteína X Asociada a bcl-2
16.
Journal of Experimental Hematology ; (6): 1577-1583, 2014.
Artículo en Zh | WPRIM | ID: wpr-340455

RESUMEN

This study was aimed to investigate the effect of Honokiol (HNK) on proliferation and apoptosis of acute myeloid leukemia HL-60 cells and its potential mechanism. Inhibitory effect of HNK on the HL-60 cell proliferation was detected by MTT assay. Flow cytometry was used to detect the change of cell cycle and AnnexinV/PI staining was used to detect apoptosis. Western blot was applied to analyze the cell cycle protein (cyclins), cyclin-dependent kinase (CDK), P53, P21, P27, BCL-2, BCL-XL, Bax, caspase-3/9 and proteins for MAPK signal pathway. The results showed that HNK could inhibit the proliferation of HL-60 cells in time- and dose dependent ways. HNK arrested HL-60 cells in G0/G1 phase, and S phase cells decreased significantly (P < 0.05). The expression of cyclin D1, cyclin A, cyclin E and CDK2/4/6 were significantly down-regulated (P < 0.05), the expression of P53 and P21 was significantly upregulated after treating for 24 h with HNK (P < 0.05). After 24 h treatment with HNK, HL-60 cell apoptosis increased significantly with the upregulation of activated caspase-3, -9, BAX expression and the downregulation of BCL-2, BCL-XL expression. The MAPK subfamily, P38 and JNK were not significantly changed, but the expression of MEK1/2-ERK1/2 was significantly downregulated (P < 0.05). It is concluded that HNK arrestes the cells at G0/G1 phase and induces HL-60 cell apoptosis through the intervention of MEK1/2-ERK1/2 signaling pathway.


Asunto(s)
Humanos , Apoptosis , Compuestos de Bifenilo , Farmacología , Caspasa 3 , Ciclo Celular , Proliferación Celular , Ciclina D1 , Ciclina E , Quinasa 2 Dependiente de la Ciclina , Células HL-60 , Lignanos , Farmacología , Proteínas Oncogénicas , Transducción de Señal , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA