Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Sci Transl Med ; 16(746): eadk4728, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38718131

RESUMEN

Group 2 innate lymphoid cells (ILC2s) rapidly induce a type 2 inflammation in the lungs in response to allergens. Here, we focused on the role of iron, a critical nutritional trace element, on ILC2 function and asthma pathogenesis. We found that transferrin receptor 1 (TfR1) is rapidly up-regulated and functional during ILC2 activation in the lungs, and blocking transferrin uptake reduces ILC2 expansion and activation. Iron deprivation reprogrammed ILC2 metabolism, inducing a HIF-1α-driven up-regulation of glycolysis and inhibition of oxidative mitochondrial activity. Consequently, we observed that in vivo iron chelation or induction of hypoferremia reduced the development of airway hyperreactivity in experimental models of ILC2-driven allergic asthma. Human circulating ILC2s rapidly induced TfR1 during activation, whereas inhibition of iron uptake or iron deprivation reduced effector functions. Last, we found a negative relationship between circulating ILC2 TfR1 expression and airway function in cohorts of patients with asthma. Collectively, our studies define cellular iron as a critical regulator of ILC2 function.


Asunto(s)
Asma , Hierro , Linfocitos , Receptores de Transferrina , Receptores de Transferrina/metabolismo , Hierro/metabolismo , Animales , Linfocitos/metabolismo , Humanos , Asma/inmunología , Asma/metabolismo , Pulmón/metabolismo , Pulmón/patología , Inmunidad Innata , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL
2.
J Exp Med ; 221(5)2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38530239

RESUMEN

Mechanosensitive ion channels sense force and pressure in immune cells to drive the inflammatory response in highly mechanical organs. Here, we report that Piezo1 channels repress group 2 innate lymphoid cell (ILC2)-driven type 2 inflammation in the lungs. Piezo1 is induced on lung ILC2s upon activation, as genetic ablation of Piezo1 in ILC2s increases their function and exacerbates the development of airway hyperreactivity (AHR). Conversely, Piezo1 agonist Yoda1 reduces ILC2-driven lung inflammation. Mechanistically, Yoda1 inhibits ILC2 cytokine secretion and proliferation in a KLF2-dependent manner, as we found that Piezo1 engagement reduces ILC2 oxidative metabolism. Consequently, in vivo Yoda1 treatment reduces the development of AHR in experimental models of ILC2-driven allergic asthma. Human-circulating ILC2s express and induce Piezo1 upon activation, as Yoda1 treatment of humanized mice reduces human ILC2-driven AHR. Our studies define Piezo1 as a critical regulator of ILC2s, and we propose the potential of Piezo1 activation as a novel therapeutic approach for the treatment of ILC2-driven allergic asthma.


Asunto(s)
Asma , Inmunidad Innata , Humanos , Animales , Ratones , Linfocitos , Inflamación , Canales Iónicos/genética
3.
Mucosal Immunol ; 16(6): 788-800, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37634572

RESUMEN

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are serious health problems that manifest as acute respiratory failure in response to different conditions, including viral respiratory infections. Recently, the inhibitory properties of leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) were demonstrated in allergic and viral airway inflammation. In this study, we investigate the implication of LAIR-1 in ALI/ARDS and explore the underlying mechanisms. Polyinosinic:polycytidylic acid, a synthetic analog of double-stranded RNA, was used to mimic acute inflammation in viral infections. We demonstrate that LAIR-1 is predominantly expressed on macrophages and regulates their recruitment to the lungs as well as their activation in response to polyinosinic:polycytidylic acid. Interestingly, LAIR-1 deficiency increases neutrophil recruitment as well as lung resistance and permeability. In particular, we highlight the capacity of LAIR-1 to regulate the secretion of CXCL10, considered a key marker of macrophage overactivation in acute lung inflammation. We also reveal in COVID-19-induced lung inflammation that LAIR1 is upregulated on lung macrophages in correlation with relevant immune regulatory genes. Altogether, our findings demonstrate the implication of LAIR-1 in the pathogenesis of ALI/ARDS by means of the regulation of macrophages, thereby providing the basis of a novel therapeutic target.


Asunto(s)
Lesión Pulmonar Aguda , Neumonía , Síndrome de Dificultad Respiratoria , Humanos , Activación de Macrófagos , Pulmón , Inflamación/patología , Poli C
4.
Cell Rep ; 42(8): 112990, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37590140

RESUMEN

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe clinical disorders that mainly develop from viral respiratory infections, sepsis, and chest injury. Antigen-presenting cells play a pivotal role in propagating uncontrolled inflammation and injury through the excess secretion of pro-inflammatory cytokines and recruitment of immune cells. Autophagy, a homeostatic process that involves the degradation of cellular components, is involved in many processes including lung inflammation. Here, we use a polyinosinic-polycytidylic acid (poly(I:C))-induced lung injury mouse model to mimic viral-induced ALI/ARDS and show that disruption of autophagy in macrophages exacerbates lung inflammation and injury, whereas autophagy induction attenuates this process. Therefore, induction of autophagy in macrophages can be a promising therapeutic strategy in ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Síndrome de Dificultad Respiratoria , Animales , Ratones , Células Presentadoras de Antígenos , Macrófagos , Autofagia , Poli I-C/farmacología
5.
J Allergy Clin Immunol ; 151(2): 526-538.e8, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35963455

RESUMEN

BACKGROUND: Neutrophilic asthma is associated with disease severity and corticosteroid insensitivity. Novel therapies are required to manage this life-threatening asthma phenotype. Programmed cell death protein-1 (PD-1) is a key homeostatic modulator of the immune response for T-cell effector functions. OBJECTIVE: We sought to investigate the role of PD-1 in the regulation of acute neutrophilic inflammation in a murine model of airway hyperreactivity (AHR). METHODS: House dust mite was used to induce and compare neutrophilic AHR in wild-type and PD-1 knockout mice. Then, the therapeutic potential of a human PD-1 agonist was tested in a humanized mouse model in which the PD-1 extracellular domain is entirely humanized. Single-cell RNA sequencing and flow cytometry were mainly used to investigate molecular and cellular mechanisms. RESULTS: PD-1 was highly induced on pulmonary T cells in our inflammatory model. PD-1 deficiency was associated with an increased neutrophilic AHR and high recruitment of inflammatory cells to the lungs. Consistently, PD-1 agonist treatment dampened AHR, decreased neutrophil recruitment, and modulated cytokine production in a humanized PD-1 mouse model. Mechanistically, we demonstrated at the transcriptional and protein levels that the inhibitory effect of PD-1 agonist is associated with the reprogramming of pulmonary effector T cells that showed decreased number and activation. CONCLUSIONS: PD-1 agonist treatment is efficient in dampening neutrophilic AHR and lung inflammation in a preclinical humanized mouse model.


Asunto(s)
Asma , Receptor de Muerte Celular Programada 1 , Humanos , Animales , Ratones , Receptor de Muerte Celular Programada 1/metabolismo , Pulmón , Células Th2 , Modelos Animales de Enfermedad
6.
Nat Commun ; 13(1): 1440, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35301333

RESUMEN

There has been a global increase in rates of obesity with a parallel epidemic of non-alcoholic fatty liver disease (NAFLD). Autophagy is an essential mechanism involved in the degradation of cellular material and has an important function in the maintenance of liver homeostasis. Here, we explore the effect of Autophagy-related 5 (Atg5) deficiency in liver CD11c+ cells in mice fed HFD. When compared to control mice, Atg5-deficient CD11c+ mice exhibit increased glucose intolerance and decreased insulin sensitivity when fed HFD. This phenotype is associated with the development of NAFLD. We observe that IL-23 secretion is induced in hepatic CD11c+ myeloid cells following HFD feeding. We demonstrate that both therapeutic and preventative IL-23 blockade alleviates glucose intolerance, insulin resistance and protects against NAFLD development. This study provides insights into the function of autophagy and IL-23 production by hepatic CD11c+ cells in NAFLD pathogenesis and suggests potential therapeutic targets.


Asunto(s)
Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Animales , Autofagia , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/genética , Interleucina-23/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo
7.
J Allergy Clin Immunol ; 149(5): 1628-1642.e10, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34673048

RESUMEN

BACKGROUND: Cannabinoids modulate the activation of immune cells and physiologic processes in the lungs. Group 2 innate lymphoid cells (ILC2s) are central players in type 2 asthma, but how cannabinoids modulate ILC2 activation remains to be elucidated. OBJECTIVE: Our goal was to investigate the effects of cannabinoids on ILC2s and their role in asthma. METHODS: A combination of cannabinoid receptor (CB)2 knockout (KO) mice, CB2 antagonist and agonist were used in the mouse models of IL-33, IL-25, and Alternaria alternata ILC2-dependent airway inflammation. RNA sequencing was performed to assess transcriptomic changes in ILC2s, and humanized mice were used to assess the role of CB2 signaling in human ILC2s. RESULTS: We provide evidence that CB2 signaling in ILC2s is important for the development of ILC2-driven airway inflammation in both mice and human. We showed that both naive and activated murine pulmonary ILC2s express CB2. CB2 signaling did not affect ILC2 homeostasis at steady state, but strikingly it stimulated ILC2 proliferation and function upon activation. As a result, ILC2s lacking CB2 induced lower lung inflammation, as we made similar observations using a CB2 antagonist. Conversely, CB2 agonism remarkably exacerbated ILC2-driven airway hyperreactivity and lung inflammation. Mechanistically, transcriptomic and protein analysis revealed that CB2 signaling induced cyclic adenosine monophosphate-response element binding protein (CREB) phosphorylation in ILC2s. Human ILC2s expressed CB2, as CB2 antagonism and agonism showed opposing effects on ILC2 effector function and development of airway hyperreactivity in humanized mice. CONCLUSION: Collectively, our results define CB2 signaling in ILC2s as an important modulator of airway inflammation.


Asunto(s)
Asma , Cannabinoides , Neumonía , Animales , Proliferación Celular , Citocinas , Humanos , Inmunidad Innata , Inflamación , Interleucina-33 , Pulmón , Linfocitos , Ratones , Ratones Noqueados , Receptor Cannabinoide CB2 , Receptores de Cannabinoides
8.
J Allergy Clin Immunol ; 149(1): 223-236.e6, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34144112

RESUMEN

BACKGROUND: Type 2 innate lymphoid cells (ILC2s) are relevant players in type 2 asthma. They initiate eosinophil infiltration and airway hyperreactivity (AHR) through cytokine secretion. Leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1) is an inhibitory receptor considered to be an immune checkpoint in different inflammatory diseases. OBJECTIVE: Our aim here was to investigate the expression of LAIR-1 and assess its role in human and murine ILC2s. METHODS: Wild-type and LAIR-1 knockout mice were intranasally challenged with IL-33, and pulmonary ILC2s were sorted to perform an ex vivo comparative study based on RNA sequencing and flow cytometry. We next studied the impact of LAIR-1 deficiency on AHR and lung inflammation by using knockout mice and adoptive transfer experiments in Rag2-/-Il2rg-/- mice. Knockdown antisense strategies and humanized mice were used to assess the role of LAIR-1 in human ILC2s. RESULTS: We have demonstrated that LAIR-1 is inducible on activated ILC2s and downregulates cytokine secretion and effector function. LAIR-1 signaling in ILC2s was mediated via inhibitory pathways, including SHP1/PI3K/AKT, and LAIR-1 deficiency led to exacerbated ILC2-dependent AHR in IL-33 and Alternaria alternata models. In adoptive transfer experiments, we confirmed the LAIR-1-mediated regulation of ILC2s in vivo. Interestingly, LAIR-1 was expressed and inducible in human ILC2s, and knockdown approaches of Lair1 resulted in higher cytokine production. Finally, engagement of LAIR-1 by physiologic ligand C1q significantly reduced ILC2-dependent AHR in a humanized ILC2 murine model. CONCLUSION: Our results unravel a novel regulatory axis in ILC2s with the capacity to reduce allergic AHR and lung inflammation.


Asunto(s)
Alternariosis/inmunología , Linfocitos/inmunología , Neumonía/inmunología , Receptores Inmunológicos/inmunología , Hipersensibilidad Respiratoria/inmunología , Traslado Adoptivo , Alternaria , Alternariosis/fisiopatología , Animales , Citocinas/inmunología , Femenino , Humanos , Inmunidad Innata , Interleucina-33/farmacología , Pulmón/inmunología , Pulmón/fisiopatología , Transfusión de Linfocitos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/fisiopatología , Receptores Inmunológicos/genética , Hipersensibilidad Respiratoria/fisiopatología
9.
Front Immunol ; 12: 733136, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34531874

RESUMEN

While pulmonary ILC2s represent one of the major tissue-resident innate lymphoid cell populations at steady state and are key drivers of cytokine secretion in their occupational niche, their role in pulmonary cancer progression remains unclear. As the programmed cell death protein-1 (PD-1) plays a major role in cancer immunotherapy and immunoregulatory properties, here we investigate the specific effect of PD-1 inhibition on ILC2s during pulmonary B16 melanoma cancer metastasis. We demonstrate that PD-1 inhibition on ILC2s suppresses B16 tumor growth. Further, PD-1 inhibition upregulates pulmonary ILC2-derived TNF-α production, a cytotoxic cytokine that directly induces cell death in B16 cells, independent of adaptive immunity. Together, these results highlight the importance of ILC2s and their anti-tumor role in pulmonary B16 cancer progression during PD-1 inhibitory immunotherapy.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Linfocitos/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Microambiente Tumoral , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Linfocitos/inmunología , Linfocitos/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/secundario , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor de Muerte Celular Programada 1/metabolismo , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Carga Tumoral
10.
Nat Commun ; 12(1): 2526, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33953190

RESUMEN

The prevalence of asthma and airway hyperreactivity (AHR) is increasing at an alarming rate. Group 2 innate lymphoid cells (ILC2s) are copious producers of type 2 cytokines, which leads to AHR and lung inflammation. Here, we show that mouse ILC2s express CD200 receptor (CD200R) and this expression is inducible. CD200R engagement inhibits activation, proliferation and type 2 cytokine production, indicating an immunoregulatory function for the CD200-CD200R axis on ILC2s. Furthermore, CD200R engagement inhibits both canonical and non-canonical NF-κB signaling pathways in activated ILC2s. Additionally, we demonstrate both preventative and therapeutic approaches utilizing CD200R engagement on ILC2s, which lead to improved airway resistance, dynamic compliance and eosinophilia. These results show CD200R is expressed on human ILC2s, and its engagement ameliorates AHR in humanized mouse models, emphasizing the translational applications for treatment of ILC2-related diseases such as allergic asthma.


Asunto(s)
Antígenos CD/metabolismo , Asma/metabolismo , Inmunidad Innata/inmunología , Linfocitos/metabolismo , Receptores de Orexina/metabolismo , Neumonía/metabolismo , Animales , Antígenos CD/genética , Asma/inmunología , Proliferación Celular , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Eosinofilia , Femenino , Humanos , Interleucina-33/metabolismo , Pulmón/metabolismo , Glicoproteínas de Membrana , Ratones , Ratones Endogámicos BALB C , Receptores de Orexina/genética , Neumonía/inmunología
11.
Oncol Rep ; 46(1)2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33955525

RESUMEN

MDM2 proto­oncogene, E3 ubiquitin protein ligase (MDM2) is a well­known oncogene and has been reported to be closely associated with epithelial­to­mesenchymal transition (EMT). The present study first demonstrated that the expression levels of MDM2 were markedly increased in TGF­ß­induced EMT using quantitative PCR and western blotting. In addition, MDM2 was demonstrated to be associated with pathological grade in clinical glioma samples by immunohistochemical staining. Furthermore, overexpression of MDM2 promoted EMT in glioma, lung cancer and breast cancer cell lines using a scratch wound migration assay. Subsequently, the present study explored the mechanism by which MDM2 promoted EMT and revealed that MDM2 induced EMT by upregulating EMT­related transcription factors via activation of the B­Raf signaling pathway through tyrosine 3­monooxygenase activation protein ε using RNA sequencing and western blotting. This mechanism depended on the p53 gene. Furthermore, in vivo experiments and the colony formation experiment demonstrated that MDM2 could promote tumor progression and induce EMT via the B­Raf signaling pathway. Since EMT contributes to increased drug resistance in tumor cells, the present study also explored the relationship between MDM2 and drug sensitivity using an MTT assay, and identified that MDM2 promoted cell insensitivity to silibinin treatment in an EMT­dependent manner. This finding is crucial for the development of cancer therapies and can also provide novel research avenues for future biological and clinical studies.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Regulación hacia Arriba , Proteínas 14-3-3/genética , Células A549 , Adulto , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/metabolismo , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Células MCF-7 , Masculino , Ratones , Persona de Mediana Edad , Clasificación del Tumor , Trasplante de Neoplasias , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Adulto Joven
12.
Mucosal Immunol ; 14(4): 899-911, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33731828

RESUMEN

Allergic asthma is a chronic inflammatory disorder associated with airway hyperreactivity (AHR) whose global prevalence is increasing at an alarming rate. Group 2 innate lymphoid cells (ILC2s) and T helper 2 (TH2) cells are producers of type 2 cytokines, which may contribute to development of AHR. In this study, we explore the potential of CD52-targeted depletion of type 2 immune cells for treating allergic AHR. Here we show that anti-CD52 therapy can prevent and remarkably reverse established IL-33-induced AHR by reducing airway resistance and alleviating lung inflammation. We further show that CD52 depletion prevents and treats allergic AHR induced by clinically relevant allergens such as Alternaria alternata and house dust mite. Importantly, we leverage various humanized mice models of AHR to show new therapeutic applications for Alemtuzumab, an anti-CD52 depleting antibody that is currently FDA approved for treatment of multiple sclerosis. Our results demonstrate that CD52 depletion is a viable therapeutic option for reduction of pulmonary inflammation, abrogation of eosinophilia, improvement of lung function, and thus treatment of allergic AHR. Taken together, our data suggest that anti-CD52 depleting monoclonal antibodies, such as Alemtuzumab, can serve as viable therapeutic drugs for amelioration of TH2- and ILC2-dependent AHR.


Asunto(s)
Alemtuzumab/farmacología , Antiinflamatorios/farmacología , Antineoplásicos Inmunológicos/farmacología , Asma/etiología , Antígeno CD52/antagonistas & inhibidores , Neumonía/etiología , Inmunidad Adaptativa/inmunología , Alérgenos/inmunología , Animales , Asma/tratamiento farmacológico , Asma/metabolismo , Asma/patología , Proteínas de Unión al ADN/deficiencia , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Humanos , Inmunidad Innata , Subgrupos Linfocitarios , Ratones , Ratones Noqueados , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Neumonía/patología , Pyroglyphidae/inmunología , Células Th2/inmunología , Células Th2/metabolismo
13.
Autophagy ; 16(2): 387-388, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31905312

RESUMEN

Macroautophagy/autophagy deregulation has been observed in perpetuated inflammation and the proliferation of tumor cells. However, the mechanisms underlying these changes have yet to be well-identified. UVRAG is one of the key players of autophagy, but its role in vivo remained puzzling. Our recent study utilized a mouse model with inducible expression of a cancer-derived frameshift (FS) mutation in UVRAG that dominant-negatively inhibits wild-type UVRAG, resulting in impaired stimulus-induced autophagy. The systemically compromised autophagy, particularly mitophagy, notably increases inflammation and associated pathologies. Furthermore, our discovery indicates that time-dependent autophagy suppression and ensuing CTNNB1/ß-catenin activation may serve as one tumor-promoting mechanism underpinning age-related cancer susceptibility.


Asunto(s)
Autofagia , Inflamación/metabolismo , Inflamación/patología , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Supresoras de Tumor/metabolismo , Animales , Autofagosomas/metabolismo , Mutación del Sistema de Lectura/genética , Ratones , Transducción de Señal , Proteínas Supresoras de Tumor/genética
14.
Nat Commun ; 10(1): 5681, 2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31831743

RESUMEN

Aberrant autophagy is a major risk factor for inflammatory diseases and cancer. However, the genetic basis and underlying mechanisms are less established. UVRAG is a tumor suppressor candidate involved in autophagy, which is truncated in cancers by a frameshift (FS) mutation and expressed as a shortened UVRAGFS. To investigate the role of UVRAGFS in vivo, we generated mutant mice that inducibly express UVRAGFS (iUVRAGFS). These mice are normal in basal autophagy but deficient in starvation- and LPS-induced autophagy by disruption of the UVRAG-autophagy complex. iUVRAGFS mice display increased inflammatory response in sepsis, intestinal colitis, and colitis-associated cancer development through NLRP3-inflammasome hyperactivation. Moreover, iUVRAGFS mice show enhanced spontaneous tumorigenesis related to age-related autophagy suppression, resultant ß-catenin stabilization, and centrosome amplification. Thus, UVRAG is a crucial autophagy regulator in vivo, and autophagy promotion may help prevent/treat inflammatory disease and cancer in susceptible individuals.


Asunto(s)
Autofagia/genética , Carcinogénesis/genética , Inflamación/genética , Mutación , Proteínas Supresoras de Tumor/genética , Animales , Carcinogénesis/patología , Proliferación Celular , Centrosoma , Colitis , Neoplasias del Colon/patología , Neoplasias Colorrectales/genética , Femenino , Mutación del Sistema de Lectura , Inflamasomas , Lipopolisacáridos/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Sepsis , Inanición , Receptor Toll-Like 4/metabolismo
15.
J Antimicrob Chemother ; 74(9): 2657-2665, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31219553

RESUMEN

OBJECTIVES: The optimal selection of antibacterials during polymicrobial infections is poorly defined. The objective of the current investigation was to quantify the pharmacodynamics of relevant antimicrobials during co-culture of Pseudomonas aeruginosa with two separate Staphylococcus aureus phenotypes. METHODS: Time-kill experiments were conducted against co-cultures of the P. aeruginosa strain PA01 paired with either the normal phenotype (NP) MRSA isolate COL or the small colony variant phenotype (SCVP) MRSA isolate Ia48. The killing by levofloxacin, gentamicin, clindamycin, vancomycin and polymyxin B was evaluated to investigate drugs with activity against one or both pathogens. A Hill-type function and a mechanism-based model were used to describe bacterial killing. RESULTS: P. aeruginosa attenuated the activity of clindamycin against NP MRSA, with a reduction in the Emax (maximal killing) from 3.67 (95% CI 2.79-4.56) in monoculture to 1.86 (95% CI 1.35-2.37) during co-culture, whereas a significant protective effect was not observed for other antibacterials. The reduction in NP MRSA killing by clindamycin was described well by a mechanism-based model that generated a maximal killing rate constant of clindamycin against the susceptible NP MRSA subpopulation of 0.267 h-1 in monoculture and 0.0395 h-1 in the presence of P. aeruginosa. During exposure to gentamicin, P. aeruginosa was the dominant organism in co-culture experiments regardless of the drug concentration or S. aureus phenotype; however, the SCVP MRSA was able to dominate the joint population beginning at a levofloxacin concentration of 1.5 mg/L. CONCLUSIONS: The anti-staphylococcal activity of clindamycin was attenuated by the presence of P. aeruginosa.


Asunto(s)
Antibacterianos/farmacología , Interacciones Microbianas , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/fisiología , Antibacterianos/uso terapéutico , Relación Dosis-Respuesta a Droga , Monitoreo de Drogas , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Modelos Biológicos , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/aislamiento & purificación , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/aislamiento & purificación
16.
Autophagy ; 15(10): 1843-1844, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31242070

RESUMEN

Although alterations of the macroautophagy/autophagy-lysosome pathway have been observed in cancer for many years, the mechanisms underlying these changes and the importance of autophagic and lysosomal reprogramming by cancer have yet to be well identified. Our recent study demonstrates that oncogenic BRAF signaling promotes melanoma growth and resistance to BRAF-targeted therapy through phosphorylation and functional inactivation of TFEB (transcription factor EB) and consequent suppression of the autophagy-lysosome gene network. This is by no means the first time that this pathway has been directly linked to oncogenic BRAF-driven melanoma. The key observations revealed in this study also leads to a complex but growing convergence of our understanding of the biology of the autophagy-lysosome pathway and the mechanisms underlying cancer prevention and treatment.


Asunto(s)
Autofagia/fisiología , Melanoma/patología , Neoplasias Cutáneas/patología , Sustitución de Aminoácidos/genética , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Ácido Glutámico/genética , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Melanoma/genética , Proteínas Proto-Oncogénicas B-raf/genética , Transducción de Señal/genética , Neoplasias Cutáneas/genética , Valina/genética
17.
Nat Commun ; 10(1): 1693, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30979895

RESUMEN

Autophagy maintains homeostasis and is induced upon stress. Yet, its mechanistic interaction with oncogenic signaling remains elusive. Here, we show that in BRAFV600E-melanoma, autophagy is induced by BRAF inhibitor (BRAFi), as part of a transcriptional program coordinating lysosome biogenesis/function, mediated by the TFEB transcription factor. TFEB is phosphorylated and thus inactivated by BRAFV600E via its downstream ERK independently of mTORC1. BRAFi disrupts TFEB phosphorylation, allowing its nuclear translocation, which is synergized by increased phosphorylation/inactivation of the ZKSCAN3 transcriptional repressor by JNK2/p38-MAPK. Blockade of BRAFi-induced transcriptional activation of autophagy-lysosomal function in melanoma xenografts causes enhanced tumor progression, EMT-transdifferentiation, metastatic dissemination, and chemoresistance, which is associated with elevated TGF-ß levels and enhanced TGF-ß signaling. Inhibition of TGF-ß signaling restores tumor differentiation and drug responsiveness in melanoma cells. Thus, the "BRAF-TFEB-autophagy-lysosome" axis represents an intrinsic regulatory pathway in BRAF-mutant melanoma, coupling BRAF signaling with TGF-ß signaling to drive tumor progression and chemoresistance.


Asunto(s)
Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Autofagia , Línea Celular Tumoral , Progresión de la Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Células HEK293 , Humanos , Lisosomas/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Microscopía Confocal , Metástasis de la Neoplasia , Trasplante de Neoplasias , Oncogenes , Fosforilación , ARN Interferente Pequeño , Transducción de Señal , Neoplasias Cutáneas/patología , Fracciones Subcelulares , Factor de Crecimiento Transformador beta/metabolismo
18.
Autophagy ; 15(2): 366-367, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30209981

RESUMEN

Ultraviolet radiation (UVR)-induced skin pigmentation, afforded by the dark organelles termed melanosomes, accounts for the first-line protection against environmental UVR that increases the risk of developing skin cancers including melanoma. We have recently discovered that UVRAG, originally identified as a BECN1-binding macroautophagy/autophagy protein, appears to have a specialized function in melanosome biogenesis beyond autophagy through its interaction with the biogenesis of lysosome-related organelles complex 1 (BLOC-1). This melanogenic function of UVRAG is controlled by the melanocyte-specific transcription factor MITF as a downstream effector of the α-melanocyte-stimulating hormone (α-MSH)-cAMP signaling in the suntan response, which is compromised in BRAF mutant melanoma. Thus we propose a new mode of UVRAG activity and regulation in melanocyte biology that may affect melanoma predisposition.


Asunto(s)
Pigmentación de la Piel , Proteínas Supresoras de Tumor/metabolismo , Beclina-1 , Humanos , Melaninas/metabolismo , Melanocitos/metabolismo , Melanocitos/efectos de la radiación , Melanosomas/metabolismo , Melanosomas/efectos de la radiación , Pigmentación de la Piel/efectos de la radiación , Rayos Ultravioleta
19.
Proc Natl Acad Sci U S A ; 115(33): E7728-E7737, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30061422

RESUMEN

UV-induced cell pigmentation represents an important mechanism against skin cancers. Sun-exposed skin secretes α-MSH, which induces the lineage-specific transcriptional factor MITF and activates melanogenesis in melanocytes. Here, we show that the autophagic tumor suppressor UVRAG plays an integral role in melanogenesis by interaction with the biogenesis of lysosome-related organelles complex 1 (BLOC-1). This interaction is required for BLOC-1 stability and for BLOC-1-mediated cargo sorting and delivery to melanosomes. Absence of UVRAG dispersed BLOC-1 distribution and activity, resulting in impaired melanogenesis in vitro and defective melanocyte development in zebrafish in vivo. Furthermore, our results establish UVRAG as an important effector for melanocytes' response to α-MSH signaling as a direct target of MITF and reveal the molecular basis underlying the association between oncogenic BRAF and compromised UV protection in melanoma.


Asunto(s)
Melaninas/biosíntesis , Melanosomas/metabolismo , Pigmentación de la Piel/efectos de la radiación , Proteínas Supresoras de Tumor/metabolismo , Rayos Ultravioleta , Animales , Células HEK293 , Humanos , Melaninas/genética , Melanoma/genética , Melanoma/metabolismo , Melanosomas/genética , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Supresoras de Tumor/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
20.
Autophagy ; 12(9): 1677-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27439570

RESUMEN

Ultraviolet (UV)-induced DNA damage is a major risk factor for skin cancers including melanoma. UVRAG, originally identified to complement UV sensitivity in xeroderma pigmentosum (XP), has since been implicated in modulating macroautophagy/autophagy, in coordinating different intracellular trafficking pathways, and in maintaining chromosomal stability. Intriguingly, our recent study has demonstrated that UVRAG plays an essential role in protecting cells from UV-induced DNA damage by activating the nucleotide excision repair (NER) pathway. Since NER is the major mechanism by which cells maintain DNA integrity against UV insult, the inactivation of UVRAG seen in some melanoma may impart these cells with an ability to accumulate high-load UV mutagenesis, leading to cancer progression. Thus, this property of UVRAG has untapped potential to be of fundamental importance in understanding the genetics and pathogenesis of human skin cancer.


Asunto(s)
Autofagia , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Proteínas Supresoras de Tumor/fisiología , Rayos Ultravioleta , Xerodermia Pigmentosa/metabolismo , Animales , Proteínas Cullin/metabolismo , Daño del ADN , Reparación del ADN , Humanos , Mutagénesis , Mutación , Fotoquímica , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA