Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Sci Rep ; 14(1): 8127, 2024 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-38584156

RESUMEN

The traditional lecture-based learning (LBL) method is facing great challenges due to its low efficiency and single proceeding form. We designed a PRI-E learning mode that combined and modified problem-based, case-based, and evidence-based learning with a step-by-step approach. We evaluated the practical learning outcomes of using the PRI-E mode by comparing it with traditional lecture-based learning in oral and maxillofacial oncology education. "PRI-E" consists of the first letters of the English words Passion, Research, Innovation, and Education, and it means "the best Education". This prospective randomized controlled trial included 40 participants. We evenly divided the participants into the PRI-E (n = 20) and LBL group (n = 20) based on the entrance test scores. The same staff group designed and then taught the learning content with different group measures. The evaluation included the final test scores and questionnaire assessments. Without affecting the examination results, the PRI-E teaching method was more satisfactory and popular with participants in terms of ability development and classroom participation. Enacting the PRI-E teaching method required more time, but this did not affect its popularity among the participants. Compared with the LBL learning mode, the PRI-E learning mode was more organized and efficient in oral and maxillofacial oncology education without affecting academic performance. This model has a high degree of satisfaction, which is conducive to training students' comprehensive ability.


Asunto(s)
Aprendizaje , Aprendizaje Basado en Problemas , Humanos , Aprendizaje Basado en Problemas/métodos , Estudios Prospectivos , Estudiantes , Evaluación Educacional
2.
Artículo en Inglés | MEDLINE | ID: mdl-38388332

RESUMEN

BACKGROUND: Thalidomide has anti-inflammatory properties and has been used off-label for multiple mucocutaneous disorders, but its application in managing refractory oral mucosal diseases is unclear. This study aimed to review the efficacy and safety of thalidomide in treating various oral mucosal disorders refractory to conventional therapies. METHODS: The medical records of patients who were prescribed thalidomide from 2002 through 2021 for oral mucosal disorders were reviewed. Data collected included demographic characteristics, oral mucosal disease diagnosis, treatment courses, and thalidomide dose, duration, response, and side effects. RESULTS: Thalidomide was prescribed for 28 patients with diagnoses of recurrent aphthous stomatitis (n = 14), inflammatory oral lichenoid lesions (n = 6), traumatic ulcerative granuloma with stroma eosinophilia (n = 5), chronic radiation-induced mucositis (n = 2), and orofacial granulomatosis (n = 1). Patients were treated for a median duration of 84 days (range 2-1,582). Clinical improvement was observed in 19 of 22 patients who completed at least 1 cycle of thalidomide (86.4%), with complete resolution in 12 patients (54.5%). Adverse events occurred in 75% of patients (n = 21), with 8 requiring thalidomide discontinuation. The most common adverse events included peripheral neuropathy (42.9%), drowsiness (28.6%), and constipation (21.4%). CONCLUSIONS: Thalidomide may be considered for the management of refractory oral mucosal disorders. Drug side effects are common and need monitoring closely during use.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Enfermedades de la Boca , Estomatitis Aftosa , Humanos , Talidomida/efectos adversos , Estomatitis Aftosa/tratamiento farmacológico , Estomatitis Aftosa/inducido químicamente , Enfermedades de la Boca/inducido químicamente , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/tratamiento farmacológico , Granuloma
3.
PLoS One ; 19(2): e0298240, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38315680

RESUMEN

PF-07209960 is a novel bispecific fusion protein composed of an anti-PD-1 antibody and engineered IL-15 cytokine mutein with reduced binding affinity to its receptors. The pharmacokinetics (PK), pharmacodynamics (PD), and toxicity of PF-07209960 were evaluated following once every other week subcutaneous (SC) or intravenous (IV) administration to cynomolgus monkeys in a repeat-dose PKPD (0.01-0.3 mg/kg/dose) and GLP toxicity study (0.1-3 mg/kg/dose). PF-07209960 showed dose dependent pharmacokinetics with a terminal T1/2 of 8 and 13 hours following IV administration at 0.03 and 0.1 mg/kg, respectively. The clearance is faster than a typical IgG1 antibody. Slightly faster clearance was also observed following the second dose, likely due to increased target pool and formation of anti-drug antibodies (ADA). Despite a high incidence rate of ADA (92%) observed in GLP toxicity study, PD-1 receptor occupancy, IL-15 signaling (STAT5 phosphorylation) and T cell expansion were comparable following the first and second doses. Activation and proliferation of T cells were observed with largest increase in cell numbers found in gamma delta T cells, followed by CD4+ and CD8+ T cells, and then NK cells. Release of cytokines IL-6, IFNγ, and IL-10 were detected, which peaked at 72 hours postdose. There was PF-07209960-related mortality at ≥1 mg/kg. At scheduled necropsy, microscopic findings were generalized mononuclear infiltration in various tissues. Both the no observed adverse effect level (NOAEL) and the highest non severely toxic dose (HNSTD) were determined to be 0.3 mg/kg/dose, which corresponded to mean Cmax and AUC48 values of 1.15 µg/mL and 37.9 µg*h/mL, respectively.


Asunto(s)
Anticuerpos Monoclonales , Receptor de Muerte Celular Programada 1 , Animales , Macaca fascicularis , Interleucina-15 , Administración Intravenosa , Citocinas , Inhibidores de Puntos de Control Inmunológico
4.
Oncologist ; 29(3): e382-e391, 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-37874927

RESUMEN

OBJECTIVES: Immune-related adverse events (irAEs) are common. Oral irAEs tend to cluster in patients who experience concurrent toxicities. We aimed to characterize the frequency and trajectory of non-oral irAEs in patients who developed oral irAEs, assess their relationship with non-oral irAEs, and compare those characteristics with patients without oral irAEs. METHODS: A retrospective chart review was conducted to identify patients who started ICIT between December 11, 2011, and September 15, 2019 (n = 4683) in the Mass General Brigham Registered Patient Data Registry. Demographic information, cancer diagnosis, ICIT regimen, treatment duration, and time and number of infusions to irAE onset were recorded. Non-oral irAEs were categorized into 13 groups. Patients with melanoma, pulmonary cancer, or head and neck cancer who had oral irAEs were then matched with those without oral irAEs to compare the prevalence of concomitant non-oral irAEs. RESULTS: Three hundred and fourteen patients with oral irAEs with a mean age of 65.9 ±â€…12.6 years (43.3% females) were included. Patients with multiple oral irAEs were more likely to have non-oral irAEs (OR: 2.7, 95% CI, 1.3-3.5), including cutaneous (OR: 1.7, 95% CI, 1.1-3.0), rheumatological (OR: 2.2, 95% CI, 1.1-4.2), thyroid (OR: 2.4, 95% CI, 1.2-4.9), and neurological irAEs (OR: 2.5, 95% CI, 1.0-6.3). Compared to matched patients with non-oral irAEs, patients with oral irAEs were more likely to have cutaneous (OR: 1.7, 95% CI, 1.0-2.8) and thyroid (OR: 2.86, 95% CI, 1.1-7.5) irAEs. The development of oral and non-oral irAEs is often coincidental. CONCLUSION: Patients who have non-oral irAEs should be monitored for development of oral irAEs for prompt management.


Asunto(s)
Antineoplásicos Inmunológicos , Neoplasias Pulmonares , Melanoma , Femenino , Humanos , Persona de Mediana Edad , Anciano , Masculino , Nivolumab/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Estudios Retrospectivos , Melanoma/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico
5.
Cancers (Basel) ; 15(18)2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37760517

RESUMEN

Post-oropharyngeal cancer treatment complications include a multitude of oral side effects that impact overall survival and quality of life. These include acute and chronic conditions affecting the oral cavity and head and neck, such as mucositis, infection, xerostomia, dysgeusia, radiation caries, osteonecrosis, and trismus. This review will summarize the most common oral complications from oropharyngeal cancer therapy. The authors would like to point out that the literature cited frequently combines oropharyngeal and head and neck cancer results. If recommendations are made strictly related to oropharyngeal cancers, this will be highlighted.

6.
Nat Cancer ; 4(1): 43-61, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36646856

RESUMEN

Prolonged interferon (IFN) signaling in cancer cells can promote resistance to immune checkpoint blockade (ICB). How cancer cells retain effects of prolonged IFN stimulation to coordinate resistance is unclear. We show that, across human and/or mouse tumors, immune dysfunction is associated with cancer cells acquiring epigenetic features of inflammatory memory. Here, inflammatory memory domains, many of which are initiated by chronic IFN-γ, are maintained by signal transducer and activator of transcription (STAT)1 and IFN regulatory factor (IRF)3 and link histone 3 lysine 4 monomethylation (H3K4me1)-marked chromatin accessibility to increased expression of a subset of IFN-stimulated genes (ISGs). These ISGs include the RNA sensor OAS1 that amplifies type I IFN (IFN-I) and immune inhibitory genes. Abrogating cancer cell IFN-I signaling restores anti-programmed cell death protein 1 (PD1) response by increasing IFN-γ in immune cells, promoting dendritic cell and CD8+ T cell interactions, and expanding T cells toward effector-like states rather than exhausted states. Thus, cancer cells acquire inflammatory memory to augment a subset of ISGs that promote and predict IFN-driven immune dysfunction.


Asunto(s)
Interferón Tipo I , Neoplasias , Animales , Humanos , Ratones , Memoria Epigenética , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Interferón Tipo I/metabolismo , Interferón Tipo I/farmacología , Interferón gamma/genética , Interferón gamma/metabolismo , Interferón gamma/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Transducción de Señal , Linfocitos T/inmunología
7.
Oral Dis ; 29(7): 2938-2943, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36165852

RESUMEN

OBJECTIVE: To evaluate the prevalence of medial vascular calcifications in the oral and maxillofacial region and their association with systemic diseases. MATERIALS AND METHODS: The study included 211 consecutive patients with systemic diseases (January 2015-May 2016). Medical history and radiographic images were evaluated. Univariate analysis (t-test) was performed for continuous variables (age). The Chi square test was applied for the categorical variables (Mönckeberg medial arteriosclerosis [MMA], gender). RESULTS: There was a 6.2% prevalence of MMA. The mean age of patients with MMA was 65.46 ± 13.38. The prevalence of kidney disease in patients with MMA was significantly higher than in those without MMA (p < 0.001). This finding was maintained even after adjusting for other systemic diseases (OR = 31.84 [8.63-136.78]). CONCLUSION: A significant prevalence of MMA in kidney disease patients was observed in this pilot study.


Asunto(s)
Arteriosclerosis , Esclerosis Calcificante de la Media de Monckeberg , Humanos , Proyectos Piloto , Esclerosis Calcificante de la Media de Monckeberg/complicaciones , Arteriosclerosis/diagnóstico por imagen , Arteriosclerosis/epidemiología , Arteriosclerosis/complicaciones
8.
BMC Oral Health ; 22(1): 161, 2022 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-35524296

RESUMEN

OBJECTIVE: To compare the reported efficacy and costs of available interventions used for the management of oral lichen planus (OLP). MATERIALS AND METHODS: A systematic literature search was performed from database inception until March 2021 in MEDLINE via PubMed and the Cochrane library following PRISMA guidelines. Only randomized controlled trials (RCT) comparing an active intervention with placebo or different active interventions for OLP management were considered. RESULTS: Seventy (70) RCTs were included. The majority of evidence suggested efficacy of topical steroids (dexamethasone, clobetasol, fluocinonide, triamcinolone), topical calcineurin inhibitors (tacrolimus, pimecrolimus, cyclosporine), topical retinoids, intra-lesional triamcinolone, aloe-vera gel, photodynamic therapy, and low-level laser therapies for OLP management. Based on the estimated cost per month and evidence for efficacy and side-effects, topical steroids (fluocinonide > dexamethasone > clobetasol > triamcinolone) appear to be more cost-effective than topical calcineurin inhibitors (tacrolimus > pimecrolimus > cyclosporine) followed by intra-lesional triamcinolone. CONCLUSION: Of common treatment regimens for OLP, topical steroids appear to be the most economical and efficacious option followed by topical calcineurin inhibitors. Large-scale multi-modality, prospective trials in which head-to-head comparisons interventions are compared are required to definitely assess the cost-effectiveness of OLP treatments.


Asunto(s)
Ciclosporinas , Liquen Plano Oral , Administración Tópica , Inhibidores de la Calcineurina/uso terapéutico , Clobetasol/uso terapéutico , Ciclosporinas/uso terapéutico , Dexametasona/uso terapéutico , Fluocinonida/uso terapéutico , Costos de la Atención en Salud , Humanos , Liquen Plano Oral/tratamiento farmacológico , Esteroides/uso terapéutico , Tacrolimus/uso terapéutico , Resultado del Tratamiento , Triamcinolona/uso terapéutico
9.
Cancer Immunol Res ; 9(10): 1141-1157, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376502

RESUMEN

The use of cytokines for immunotherapy shows clinical efficacy but is frequently accompanied by severe adverse events caused by excessive and systemic immune activation. Here, we set out to address these challenges by engineering a fusion protein of a single, potency-reduced, IL15 mutein and a PD1-specific antibody (anti-PD1-IL15m). This immunocytokine was designed to deliver PD1-mediated, avidity-driven IL2/15 receptor stimulation to PD1+ tumor-infiltrating lymphocytes (TIL) while minimally affecting circulating peripheral natural killer (NK) cells and T cells. Treatment of tumor-bearing mice with a mouse cross-reactive fusion, anti-mPD1-IL15m, demonstrated potent antitumor efficacy without exacerbating body weight loss in B16 and MC38 syngeneic tumor models. Moreover, anti-mPD1-IL15m was more efficacious than an IL15 superagonist, an anti-mPD-1, or the combination thereof in the B16 melanoma model. Mechanistically, anti-PD1-IL15m preferentially targeted CD8+ TILs and single-cell RNA-sequencing analyses revealed that anti-mPD1-IL15m treatment induced the expansion of an exhausted CD8+ TIL cluster with high proliferative capacity and effector-like signatures. Antitumor efficacy of anti-mPD1-IL15m was dependent on CD8+ T cells, as depletion of CD8+ cells resulted in the loss of antitumor activity, whereas depletion of NK cells had little impact on efficacy. The impact of anti-hPD1-IL15m on primary human TILs from patients with cancer was also evaluated. Anti-hPD1-IL15m robustly enhanced the proliferation, activation, and cytotoxicity of CD8+ and CD4+ TILs from human primary cancers in vitro, whereas tumor-derived regulatory T cells were largely unaffected. Taken together, our findings showed that anti-PD1-IL15m exhibits a high translational promise with improved efficacy and safety of IL15 for cancer immunotherapy via targeting PD1+ TILs.See related Spotlight by Felices and Miller, p. 1110.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias del Colon/terapia , Inmunoterapia , Interleucina-15/uso terapéutico , Melanoma Experimental/terapia , Animales , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Modelos Animales de Enfermedad , Humanos , Interleucina-15/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/inmunología , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico
10.
Materials (Basel) ; 14(14)2021 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-34300937

RESUMEN

From the perspective of damage mechanics, the damage parameters were introduced as the characterizing quantity of the decrease in the mechanical properties of powder superalloy material FGH96 under fatigue loading. By deriving a damage evolution equation, a fatigue life prediction model of powder superalloy containing inclusions was constructed based on damage mechanics. The specimens containing elliptical subsurface inclusions and semielliptical surface inclusions were considered. The CONTA172 and TARGE169 elements of finite element software (ANSYS) were used to simulate the interfacial debonding between the inclusions and matrix, and the interface crack initiation life was calculated. Through finite element modeling, the stress field evolution during the interface debonding was traced by simulation. Finally, the effect of the position and shape size of inclusions on interface debonding was explored.

11.
Cancer ; 127(11): 1796-1804, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33595843

RESUMEN

BACKGROUND: Immune checkpoint inhibitors (ICIs) are increasingly accepted as a treatment option for several cancers. Although various systemic immune-related adverse events (irAEs) have been characterized, the effect of ICIs on the oral cavity and contiguous structures is still poorly understood. METHODS: Electronic medical records of 4683 patients in the Mass General Brigham Registered Patient Data Registry who received ICI therapy (ICIT) between December 2011 and September 2019 were reviewed. Reports of oral conditions were categorized into oral mucosal disorders, xerostomia, and dysgeusia. After applying exclusion criteria, demographic characteristics and clinical features were summarized for the patients who had oral irAEs. RESULTS: In total, 317 patients developed oral conditions that were associated with ICIT (incidence, 6.8%; 317 of 4683 patients). These conditions included xerostomia (68.5%), oral mucosal disorders (33.4%), and dysgeusia (24.0%). In patients with oral irAEs, respiratory cancer (28.4%) was the most common primary cancer, followed by melanoma (26.2%), and head and neck cancer (14.8%). Oral mucosal disorders developed after the initiation of ICIT between 2 and 851 days (between 1 and 1332 days in patients with xerostomia and between 1 and 1455 days in patients with dysgeusia). Of all oral irAEs, 50.9% developed within 3 months, and 85.5% developed within 12 months. CONCLUSIONS: Oral side effects appear to be more common among patients who receive ICIT than has been previously reported. Concomitant cytotoxic regimens may exacerbate the risk of oral adverse events, perhaps representing the sum of the effects of different, but simultaneous or sequential, pathogenic mechanisms. Additional studies are warranted to better characterize oral irAEs and their biologic basis.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Administración Oral , Instituciones Oncológicas , Femenino , Hospitales Generales , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Massachusetts , Neoplasias/tratamiento farmacológico
13.
Nature ; 582(7812): 416-420, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32499641

RESUMEN

Regulatory T (Treg) cells are required to control immune responses and maintain homeostasis, but are a significant barrier to antitumour immunity1. Conversely, Treg instability, characterized by loss of the master transcription factor Foxp3 and acquisition of proinflammatory properties2, can promote autoimmunity and/or facilitate more effective tumour immunity3,4. A comprehensive understanding of the pathways that regulate Foxp3 could lead to more effective Treg therapies for autoimmune disease and cancer. The availability of new functional genetic tools has enabled the possibility of systematic dissection of the gene regulatory programs that modulate Foxp3 expression. Here we developed a CRISPR-based pooled screening platform for phenotypes in primary mouse Treg cells and applied this technology to perform a targeted loss-of-function screen of around 500 nuclear factors to identify gene regulatory programs that promote or disrupt Foxp3 expression. We identified several modulators of Foxp3 expression, including ubiquitin-specific peptidase 22 (Usp22) and ring finger protein 20 (Rnf20). Usp22, a member of the deubiquitination module of the SAGA chromatin-modifying complex, was revealed to be a positive regulator that stabilized Foxp3 expression; whereas the screen suggested that Rnf20, an E3 ubiquitin ligase, can serve as a negative regulator of Foxp3. Treg-specific ablation of Usp22 in mice reduced Foxp3 protein levels and caused defects in their suppressive function that led to spontaneous autoimmunity but protected against tumour growth in multiple cancer models. Foxp3 destabilization in Usp22-deficient Treg cells could be rescued by ablation of Rnf20, revealing a reciprocal ubiquitin switch in Treg cells. These results reveal previously unknown modulators of Foxp3 and demonstrate a screening method that can be broadly applied to discover new targets for Treg immunotherapies for cancer and autoimmune disease.


Asunto(s)
Sistemas CRISPR-Cas , Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Autoinmunidad/inmunología , Células Cultivadas , Factores de Transcripción Forkhead/biosíntesis , Edición Génica , Regulación de la Expresión Génica , Humanos , Inmunoterapia , Masculino , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/prevención & control , Estabilidad Proteica , Reproducibilidad de los Resultados , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/metabolismo
14.
J Autoimmun ; 109: 102423, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32057541

RESUMEN

Endoplasmic reticulum (ER)-associated degradation (ERAD) is a mechanism against ER stress, wherein unfolded/misfolded proteins accumulated in the ER are transported to the cytosol for degradation by the ubiquitin-proteasome system. The ER resident E3 ubiquitin ligase HRD1 has been identified as a key ERAD factor that directly catalyzes ubiquitin conjugation onto the unfolded or misfolded proteins for proteasomal degradation. The abnormally increased HRD1 expression was discovered in rheumatoid synovial cells, providing the first evidence for HRD1 dysregulation involved in human inflammatory pathogenesis. Further studies shown that inflammatory cytokines involved in rheumatoid pathogenesis including IL-1ß, TNF-α, IL-17 and IL-26 induce HRD1 expression. Recent studies using mice with tissue-specific targeted deletion of HRD1 gene have revealed important functions of HRD1 in immune regulation and inflammatory diseases. HRD1 has been shown critical for dendritic cell expression of antigens to both CD4 and CD8 T cells. Both TCR and costimulatory receptor CD28 signaling induces HRD1 expression, which promotes T cell clonal expansion and IL-2 production. Together with the fact that HRD1 is required for maintaining the stability of regulatory T cell (Treg) stability, HRD1 appears to fine tone T cell immunity. In addition, HRD1 is involved in humoral immune response by regulating early B cell development and maintaining B cell survival upon recognition of specific antigen. HRD1 appears to target its substrates for ubiquitination through, either ERAD-dependent or -independent, at least two distinct molecular mechanisms in a cell or tissue specific manner to achieve its physiological functions. Dysregulation of HRD1 expression and/or it functions are involved in autoimmune inflammatory diseases in particular rheumatoid arthritis and lupus. Here, we review current findings on the mechanism of HRD1 protein in immune regulation and the involvement of HRD1 in the pathogenesis of autoimmune inflammatory diseases.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Autoinmunidad , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Humanos , Inmunomodulación , Procesamiento Proteico-Postraduccional , Proteolisis , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
15.
Sensors (Basel) ; 19(23)2019 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-31757063

RESUMEN

Piezoelectric (PZT) ceramic elements are often subjected to complex loads during in- service lifetime in structural health monitoring (SHM) systems, and debonding of both excitation actuators and receiving sensors have a negative effect on the monitoring signals. A first systematic investigation of debonding behaviors by considering actuators and sensors simultaneously was performed in this paper. The debonding areas of actuators were set in different percentage range from 0% to 70%, and sensors in 0%, 20%, 40% and 60%. The signal-based monitoring method was used to extract the characteristic parameters of both the amplitudes and phases of received signals. Experimental results revealed that as the debonding areas of the actuators increase, the normalized amplitude appears a quick decrease before 35% debonding area of actuators and then a slow rise until 60% of debonding reached. This may be explained that the 35% debonding turning point correspond to the coincidence of the excitation frequencies of peripheral actuators with the inherent frequency of the central piezoelectric sensor, and the 60% be the result of the maximum ability of piezoelectric sensor. The degrees of debonding of actuators and sensors also have significant influence on the phase angle offset, with large debonding of actuators increases the phase offset sharply. The research work may provide useful information for practical monitoring of SHM systems.

16.
Cell ; 178(4): 933-948.e14, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31398344

RESUMEN

Interferon-gamma (IFNG) augments immune function yet promotes T cell exhaustion through PDL1. How these opposing effects are integrated to impact immune checkpoint blockade (ICB) is unclear. We show that while inhibiting tumor IFNG signaling decreases interferon-stimulated genes (ISGs) in cancer cells, it increases ISGs in immune cells by enhancing IFNG produced by exhausted T cells (TEX). In tumors with favorable antigenicity, these TEX mediate rejection. In tumors with neoantigen or MHC-I loss, TEX instead utilize IFNG to drive maturation of innate immune cells, including a PD1+TRAIL+ ILC1 population. By disabling an inhibitory circuit impacting PD1 and TRAIL, blocking tumor IFNG signaling promotes innate immune killing. Thus, interferon signaling in cancer cells and immune cells oppose each other to establish a regulatory relationship that limits both adaptive and innate immune killing. In melanoma and lung cancer patients, perturbation of this relationship is associated with ICB response independent of tumor mutational burden.


Asunto(s)
Inmunidad Adaptativa/inmunología , Inmunidad Innata/inmunología , Interferón gamma/genética , Interferón gamma/metabolismo , Neoplasias Pulmonares/inmunología , Melanoma/inmunología , Traslado Adoptivo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Línea Celular Tumoral , Estudios de Cohortes , Femenino , Técnicas de Inactivación de Genes , Humanos , Interferón gamma/antagonistas & inhibidores , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Supervivencia sin Progresión , RNA-Seq , Transfección
17.
JCI Insight ; 4(5)2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30843874

RESUMEN

Treg differentiation, maintenance, and function are controlled by the transcription factor FoxP3, which can be destabilized under inflammatory or other pathological conditions. Tregs can be destabilized under inflammatory or other pathological conditions, but the underlying mechanisms are not fully defined. Herein, we show that inflammatory cytokines induce ER stress response, which destabilizes Tregs by suppressing FoxP3 expression, suggesting a critical role of the ER stress response in maintaining Treg stability. Indeed, genetic deletion of Hrd1, an E3 ligase critical in suppressing the ER stress response, leads to elevated expression of ER stress-responsive genes in Treg and largely diminishes Treg suppressive functions under inflammatory condition. Mice with Treg-specific ablation of Hrd1 displayed massive multiorgan lymphocyte infiltration, body weight loss, and the development of severe small intestine inflammation with aging. At the molecular level, the deletion of Hrd1 led to the activation of both the ER stress sensor IRE1α and its downstream MAPK p38. Pharmacological suppression of IRE1α kinase, but not its endoribonuclease activity, diminished the elevated p38 activation and fully rescued the stability of Hrd1-null Tregs. Taken together, our studies reveal ER stress response as a previously unappreciated mechanism underlying Treg instability and that Hrd1 is crucial for maintaining Treg stability and functions through suppressing the IRE1α-mediated ER stress response.


Asunto(s)
Citocinas/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/metabolismo , Activación de Linfocitos/inmunología , Linfocitos T Reguladores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis , Colitis/inmunología , Colitis/patología , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Endorribonucleasas , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Homeostasis , Inflamación/inmunología , Inflamación/patología , Linfocitos Nulos , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Transcriptoma , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética
18.
EMBO J ; 37(22)2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30389664

RESUMEN

The endoplasmic reticulum-associated protein degradation (ERAD) is responsible for recognizing and retro-translocating protein substrates, misfolded or not, from the ER for cytosolic proteasomal degradation. HMG-CoA Reductase (HMGCR) Degradation protein-HRD1-was initially identified as an E3 ligase critical for ERAD. However, its physiological functions remain largely undefined. Herein, we discovered that hepatic HRD1 expression is induced in the postprandial condition upon mouse refeeding. Mice with liver-specific HRD1 deletion failed to repress FGF21 production in serum and liver even in the refeeding condition and phenocopy the FGF21 gain-of-function mice showing growth retardation, female infertility, and diurnal circadian behavior disruption. HRD1-ERAD facilitates the degradation of the liver-specific ER-tethered transcription factor CREBH to downregulate FGF21 expression. HRD1-ERAD catalyzes polyubiquitin conjugation onto CREBH at lysine 294 for its proteasomal degradation, bridging a multi-organ crosstalk in regulating growth, circadian behavior, and female fertility through regulating the CREBH-FGF21 regulatory axis.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Factores de Crecimiento de Fibroblastos/biosíntesis , Hígado/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Femenino , Fertilidad/genética , Factores de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Hígado/patología , Masculino , Ratones , Ratones Transgénicos , Poliubiquitina/genética , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Ubiquitina-Proteína Ligasas/genética
19.
Nat Commun ; 9(1): 3659, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30201971

RESUMEN

The HMG-CoA reductase degradation protein 1 (HRD1) has been identified as a key enzyme for endoplasmic reticulum-associated degradation of misfolded proteins, but its organ-specific physiological functions remain largely undefined. Here we show that mice with HRD1 deletion specifically in the liver display increased energy expenditure and are resistant to HFD-induced obesity and liver steatosis and insulin resistance. Proteomic analysis identifies a HRD1 interactome, a large portion of which includes metabolic regulators. Loss of HRD1 results in elevated ENTPD5, CPT2, RMND1, and HSD17B4 protein levels and a consequent hyperactivation of both AMPK and AKT pathways. Genome-wide mRNA sequencing revealed that HRD1-deficiency reprograms liver metabolic gene expression profiles, including suppressing genes involved in glycogenesis and lipogenesis and upregulating genes involved in glycolysis and fatty acid oxidation. We propose HRD1 as a liver metabolic regulator and a potential drug target for obesity, fatty liver disease, and insulin resistance associated with the metabolic syndrome.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Hígado/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adenilato Quinasa/metabolismo , Animales , Peso Corporal , Dieta Alta en Grasa , Activación Enzimática , Ácidos Grasos/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Glucólisis , Células HEK293 , Células Hep G2 , Humanos , Lipogénesis , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Proteoma , Proteómica , Triglicéridos/metabolismo , Ubiquitinación
20.
J Biol Chem ; 293(28): 11067-11075, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29789426

RESUMEN

The type III NAD-dependent histone deacetylase Sirt1 plays important roles in a variety of pathobiological functions through targeting either the acetylated histones or transcription factors. However, the molecular mechanisms underlying how the Sirt1 functions are regulated remain vague. Herein we identified that the Janus kinase 1 (JAK1) interacts with Sirt1 and catalyzes its phosphorylation at the tyrosine residues of 280 and 301, both of which are highly conserved and located in the histone deacetylase catalytic domain of Sirt1. IL-6 stimulation enhanced Sirt1 interaction with JAK1 and JAK1-mediated Sirt1 phosphorylation. Interestingly, JAK1-mediated Sirt1 phosphorylation did not alter Sirt1 deacetylase catalytic activity, but instead it is required for Sirt1 interaction with the downstream transcription factor STAT3. JAK1-mediated phosphorylation enhanced Sirt1 suppression of STAT3 acetylation and transcriptional activity. As a consequence, Sirt1 activation attenuates IL-6 activity in protecting cancer cells from chemotherapeutic drug-induced apoptosis. Our studies identify JAK1 as a previously unappreciated tyrosine kinase of Sirt1 and reveal a novel negative feedback of the JAK1-STAT3 pathway.


Asunto(s)
Retroalimentación Fisiológica , Janus Quinasa 1/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo , Activación Enzimática , Células HEK293 , Humanos , Janus Quinasa 1/genética , Fosforilación , Factor de Transcripción STAT3/genética , Sirtuina 1/genética , Tirosina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...