Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Behav Sleep Med ; 22(2): 217-233, 2024 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-37401160

RESUMEN

OBJECTIVES: Both sleep and cognition are partially modulated by the endocannabinoid (ECB) system. Cannabis has been reported to have effects on sleep and cognition. This review aims to summarize the recent literature on the ECB system, the role of cannabis and the ECB system on sleep regulation and cognition. Further, this review will identify existing gaps in knowledge and suggest potential targets for future research. METHODS: We performed this review in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Reports were identified by searching PubMed/MEDLINE, Embase, CINAHL, Web of Science, and PsycINFO for articles published through September 2021 for studies with data available on aspects of cognition, cannabis, or the ECB system, and sleep or circadian rhythms (CRs). RESULTS: We identified 6 human and 6 animal studies to be eligible for inclusion in this review. Several human studies found that cannabis use is not associated with changes in sleep quality or cognitive function. However, individual cannabinoids appeared to have independent effects on cognition and sleep; THC alone decreased cognitive performance and increased daytime sleepiness, whereas CBD alone had no effect on sleep or cognition. Animal studies demonstrated that manipulation of the ECB system altered activity and cognitive function, some of which appeared to be dependent on the light/dark cycle. CONCLUSION: The sleep-wake cycle and CRs are both likely modulated by the ECB system, potentially resulting in effects on cognition, however this area is critically understudied.


Asunto(s)
Cannabinoides , Cannabis , Animales , Humanos , Endocannabinoides , Sueño , Cognición
3.
Front Digit Health ; 4: 920706, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36082232

RESUMEN

Digital surveys, such as mobile phone ecological momentary assessment (EMA), bear the potential to assess and target individual wellbeing in a personalized, real-time approach and allow for interaction in situations when in-person contact is not possible, such as during the coronavirus pandemic. While the use of digital technology might especially benefit research in older adults who find themselves in circumstances of reduced mobility, little is known about their barriers to adherence. We investigated baseline and structural factors that predict study withdrawal and adherence from daily smartphone EMA self-report surveys in the StayWELL Study. The StayWELL study is a longitudinal, observational study on the relationship between social restrictions during the coronavirus pandemic and mental well-being in 95 community-dwelling older aged adults (67-87 years) who were participants in a randomized clinical trial using EMA. Withdrawal was associated with less research staff changes and less likely in participants that reached the study mid-point. No baseline characteristics predicted withdrawal. Main reasons for withdrawal were communication issues, i.e. staff not being able to contact participants. We found an adherence rate of 82% and no fatigue effects. Adherence was predicted by education status, study participation duration, reaching the study midpoint and time between study start and enrollment. COVID infections or supporting people in the household was not related to adherence. To conclude, it is feasible to conduct an EMA study in older people without impacting engagement during a pandemic. Furthermore, personal characteristics and smartphone operating system (Android vs. iOS) used did not relate to engagement, allowing for a broad distribution of digital health technologies. Our study adds information on single predictive variables relevant for adherence and withdrawal from EMA smartphone surveys in older people that can inform the design of future digital EMA research to maximize engagement and reliability of study results.

4.
Clin Cancer Res ; 22(21): 5349-5361, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27189164

RESUMEN

PURPOSE: The transcriptional regulation mediating cancer cell differentiation into distinct molecular subtypes and modulating sensitivity to existing treatments is an enticing therapeutic target. Our objective was to characterize the ability of the forkhead/winged transcription factor FOXP3 to modulate the differentiation of bladder cancer. EXPERIMENTAL DESIGN: Expression of FOXP3 was analyzed by immunohistochemistry in a tumor microarray of 587 samples and overall survival in a subset of 187 patients following radical cystectomy. Functional assays were performed in SW780 and HT1376 cell lines in vitro and in vivo and gene expression profiling performed by RNA-Seq. Validation was undertaken using gene expression profiles of 131 patients from The Cancer Genome Atlas (TCGA) consortium in bladder cancer. RESULTS: FOXP3 expression correlates with bladder cancer stage and inversely with overall survival, with biased expression of the FOXP3Δ3 isoform. Functional assays of FOXP3Δ3 demonstrated resistance to chemotherapy in vitro, whereas subcutaneous xenografts overexpressing FOXP3Δ3 developed larger and more poorly differentiated bladder cancers. RNA expression profiling revealed a unique FOXP3Δ3 gene signature supporting a role in chemotherapy resistance. Accordingly, knockdown of Foxp3 by siRNA in HT1376 cells conferred sensitivity to cisplatin- and gemcitabine-induced cytotoxicity. Validation in TCGA dataset demonstrated increased expression of FOXP3 in subtypes II to IV and skewing of molecular subtypes based on FOXP3Δ3-specific gene expression. CONCLUSIONS: (i) Biased expression of the FOXP3Δ3 isoform in bladder cancer inversely correlates with overall survival, (ii) FOXP3Δ3 induces a unique gene program that mediates cancer differentiation, and (iii) FOXP3Δ3 may augment chemotherapy resistance. Clin Cancer Res; 22(21); 5349-61. ©2016 AACR.


Asunto(s)
Diferenciación Celular/genética , Resistencia a Antineoplásicos/genética , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica/genética , Isoformas de Proteínas/metabolismo , Neoplasias de la Vejiga Urinaria/genética , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Cisplatino/farmacología , Cistectomía/métodos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Inmunohistoquímica/métodos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , ARN Interferente Pequeño/genética , Vejiga Urinaria/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Gemcitabina
5.
J Urol ; 194(2): 547-55, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25748177

RESUMEN

PURPOSE: Cisplatin based chemotherapy regimens form the basis of systemic bladder cancer treatment, although they show limited response rates and efficacy. Recent molecular analysis of bladder cancer revealed a high incidence of mutations in chromatin regulatory genes, suggesting a therapeutic avenue for histone deacetylase inhibitors. We investigated the ability of the novel histone deacetylase inhibitor AR-42 to synergize with cisplatin in preclinical models of bladder cancer. MATERIALS AND METHODS: We assessed the ability of the pan-histone deacetylase inhibitor AR-42 with and without cisplatin to destroy bladder cancer cells by survival and apoptosis assays in vitro, and by growth and differentiation in an in vivo xenograft model. We also assessed the response to the bladder cancer stem cell population by examining the effect of AR-42 on the CD44(+)CD49f(+) population with and without cisplatin. Synergy was calculated using combination indexes. RESULTS: The AR-42 and cisplatin combination synergistically destroyed bladder cancer cells via apoptosis and it influenced tumor growth and differentiation in vivo. When tested in the CD44(+)CD49f(+) bladder cancer stem cell population, AR-42 showed greater efficacy with and without cisplatin. CONCLUSIONS: AR-42 may be an attractive novel histone deacetylase inhibitor with activity against bladder cancer. Its efficacy in bladder cancer stem cells and synergy with cisplatin warrant further clinical investigation. Our in vitro and animal model studies provide preclinical evidence that AR-42 may be administered in conjunction with cisplatin based chemotherapy to improve the treatment of bladder cancer in patients.


Asunto(s)
Cisplatino/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Fenilbutiratos/farmacología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Histona Desacetilasas , Humanos , Neoplasias de la Vejiga Urinaria/patología
6.
Prostate ; 75(5): 463-73, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25597486

RESUMEN

BACKGROUND: The influence of pattern recognition receptor (PRR) signaling in the prostate tumor microenvironment remains unclear. Although there may be a role for PRR agonists as adjuvants to therapy, prior evidence suggests tumor promoting as well as tumor inhibiting mechanisms. The purpose of this study is to examine the role of the key Toll-like receptor (TLR) signaling adaptor protein myeloid differentiation primary response gene 88 (MyD88) in prostate cancer development. METHODS: MyD88(-/-) mice in a C57Bl6 background were crossed with transgenic adenocarcinomas of the mouse prostate (TRAMP) mice to create MyD88(-/-) TRAMP(Tg+/-) animals, which were compared to MyD88(+/+) TRAMP(Tg+/-) animals and their non-transgenic counterparts at 30 weeks. Prostates were examined histologically, by immunohistochemistry and immunofluorescence staining, and by qPCR, to characterize tumor-infiltrating immune populations as well as activation of the downstream NF-κB pathway and androgen receptor (AR) expression. Splenocytes were examined for development of distinct immune cell populations. RESULTS: Absence of MyD88 led to increased prostatic intraepithelial neoplasm (PIN) and areas of well-differentiated adenocarcinoma in TRAMP transgenic mice. Analysis of infiltrating immune populations revealed an increase in CD11b(+) Gr1(+) myeloid-derived suppressor cells (MDSCs), as evidenced by increased expression of prostatic arginase-1 and iNOS as well as the cytokine IL-10, and a deficiency in NK cells in prostates from MyD88(-/-) TRAMP(Tg+/-) compared to MyD88(+/+) TRAMP(Tg+/-) mice, whereas a decrease in splenocytic NK cell differentiation was observed in MyD88(-/-) mice. Prostate tumors revealed no significant differences in NF-κB or AR expression in MyD88(+/+) TRAMP(Tg+/-) compared to MyD88(-/-) TRAMP(Tg+/-) mice. CONCLUSIONS: During prostate cancer development in the TRAMP model, MyD88 may play a role in limiting prostate tumorigenesis by altering tumor-infiltrating immune populations. This suggests that in the context of specific cancers, distinct PRRs and signaling pathways of innate immune signaling may influence the tumor microenvironment and represent a novel therapeutic strategy.


Asunto(s)
Adenocarcinoma/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Factor 88 de Diferenciación Mieloide/fisiología , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/patología , Animales , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Linfocitos Infiltrantes de Tumor/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Neoplasias de la Próstata/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Androgénicos/metabolismo , Transducción de Señal
7.
Am J Cancer Res ; 2(6): 745-51, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226620

RESUMEN

The development of new cancer therapeutics would benefit from incorporating efficient tumor models that mimic human disease. We have developed a subcutaneous bladder tumor regeneration system that recapitulates primary human bladder tumor architecture by recombining benign human fetal bladder stromal cells with SW780 bladder carcinoma cells. As a first step, SW780 cells were seeded in ultra low attachment cultures in order to select for sphere-forming cells, the putative cancer stem cell (CSC) phenotype. Spheroids were combined with primary human fetal stromal cells or vehicle control and injected subcutaneously with Matrigel into NSG mice. SW780 bladder tumors that formed in the presence of stroma showed accelerated growth, muscle invasion, epithelial to mesenchymal transition (EMT), decreased differentiation, and greater activation of growth pathways compared to tumors formed in the absence of fetal stroma. Tumors grown with stroma also demonstrated a greater similarity to typical malignant bladder architecture, including the formation of papillary structures. In an effort to determine if cancer cells from primary tumors could form similar structures in vivo using this recombinatorial approach, putative CSCs, sorted based on the CD44(+)CD49f(+) antigenic profile, were collected and recombined with fetal bladder stromal cells and Matrigel prior to subcutaneous implantation. Retrieved grafts contained tumors that exhibited the same structure as the original primary human tumor. Primary bladder tumor regeneration using human fetal bladder stroma may help elucidate the influences of stroma on tumor growth and development, as well as provide an efficient and accessible system for therapeutic testing.

8.
J Immunol ; 187(6): 3438-47, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21841127

RESUMEN

We previously reported that 1 h after infusion of CD20 mAb rituximab in patients with chronic lymphocytic leukemia (CLL), >80% of CD20 was removed from circulating B cells, and we replicated this finding, based on in vitro models. This reaction occurs via an endocytic process called shaving/trogocytosis, mediated by FcγR on acceptor cells including monocytes/macrophages, which remove and internalize rituximab-CD20 immune complexes from B cells. Beers et al. reported that CD20 mAb-induced antigenic modulation occurs as a result of internalization of B cell-bound mAb-CD20 complexes by the B cells themselves, with internalization of ∼40% observed after 2 h at 37°C. These findings raise fundamental questions regarding the relative importance of shaving versus internalization in promoting CD20 loss and have substantial implications for the design of mAb-based cancer therapies. Therefore, we performed direct comparisons, based on flow cytometry, to determine the relative rates and extent of shaving versus internalization. B cells, from cell lines, from patients with CLL, and from normal donors, were opsonized with CD20 mAbs rituximab or ofatumumab and incubated for varying times and then reacted with acceptor THP-1 monocytes to promote shaving. We find that shaving induces considerably greater loss of CD20 and bound mAb from opsonized B cells in much shorter time periods (75-90% in <45 min) than is observed for internalization. Both shaving/trogocytosis and internalization could contribute to CD20 loss when CLL patients receive rituximab therapy, but shaving should occur more rapidly and is most likely to be the key mechanism of CD20 loss.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Antígenos CD20/metabolismo , Antineoplásicos/farmacología , Linfocitos B/metabolismo , Endocitosis/fisiología , Monocitos/metabolismo , Animales , Linfocitos B/efectos de los fármacos , Línea Celular , Separación Celular , Endocitosis/efectos de los fármacos , Citometría de Flujo , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Ratones , Receptores de IgG/metabolismo , Rituximab , Transfección
9.
Eur J Immunol ; 41(8): 2436-46, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21674476

RESUMEN

We have reported that during complement-mediated cytolysis of B cells promoted by the CD20 mAbs rituximab or ofatumumab (OFA), long, thin structures that we call streamers (≥ 3 cell diameters) are rapidly generated and grow out from the cell surface. Streamers appear before cells are killed and contain opsonizing mAbs and membrane lipids. By exploiting the differential Ca(2+) requirements of discrete steps in the complement cascade, we determined that mAb-opsonized cells first tagged with C3b using C5-depleted serum are killed on addition of serum and EDTA, but the cells do not produce streamers. Also, cells first opsonized with OFA are lysed in serum containing Mg-EGTA by the alternative complement pathway but streamers are not produced. These findings indicate that Ca(2+) influx is necessary for streamer formation. Other mAbs that promote complement-mediated cytolysis also induce streamers on target cells. Streamer-like structures called nanotubes have been reported in several cellular systems, and are thought to promote intercellular communication/signaling. We tested whether this signaling could influence the susceptibility of neighboring cells contacted by streamers to complement attack and found that complement-mediated cytolysis of OFA-opsonized cells increases the resistance of unopsonized indicator cell populations to subsequent lysis when these cells are exposed to OFA and complement.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Calcio/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Citotoxicidad Inmunológica/inmunología , Anticuerpos Monoclonales Humanizados , Anticuerpos Monoclonales de Origen Murino/inmunología , Antígenos CD20/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígenos CD55/inmunología , Antígenos CD55/metabolismo , Calcio/antagonistas & inhibidores , Calcio/metabolismo , Línea Celular , Línea Celular Tumoral , Quelantes/farmacología , Activación de Complemento/inmunología , Complemento C3b/inmunología , Complemento C3b/metabolismo , Complemento C5/inmunología , Complemento C5/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Vía Alternativa del Complemento/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Ácido Egtácico/farmacología , Citometría de Flujo , Humanos , Antígenos Comunes de Leucocito/inmunología , Antígenos Comunes de Leucocito/metabolismo , Meliteno/farmacología , Microscopía Fluorescente , Fosfatidilserinas/inmunología , Fosfatidilserinas/metabolismo , Rituximab , Suero/inmunología , Suero/metabolismo
10.
Blood ; 115(11): 2283-91, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-20068220

RESUMEN

The clinical hallmark of paroxysmal nocturnal hemoglobinuria (PNH) is chronic intravascular hemolysis that is a consequence of unregulated activation of the alternative pathway of complement (APC). Intravascular hemolysis can be inhibited in patients by treatment with eculizumab, a monoclonal antibody that binds complement C5 thereby preventing formation of the cytolytic membrane attack complex of complement. However, in essentially all patients treated with eculizumab, persistent anemia, reticulocytosis, and biochemical evidence of hemolysis are observed; and in a significant proportion, their PNH erythrocytes become opsonized with complement C3. These observations suggest that PNH patients treated with eculizumab are left with clinically significant immune-mediated hemolytic anemia because the antibody does not block APC activation. With a goal of improving PNH therapy, we characterized the activity of anti-C3b/iC3b monoclonal antibody 3E7 in an in vitro model of APC-mediated hemolysis. We show that 3E7 and its chimeric-deimmunized derivative H17 block both hemolysis and C3 deposition on PNH erythrocytes. The antibody is specific for the APC C3/C5 convertase because classical pathway-mediated hemolysis is unaffected by 3E7/H17. These findings suggest an approach to PNH treatment in which both intravascular and extravascular hemolysis can be inhibited while preserving important immune functions of the classical pathway of complement.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Especificidad de Anticuerpos/efectos de los fármacos , Complemento C3/inmunología , Vía Alternativa del Complemento/efectos de los fármacos , Hemoglobinuria Paroxística/inmunología , Hemoglobinuria Paroxística/prevención & control , Hemólisis/efectos de los fármacos , Ácidos , Anticuerpos Monoclonales/uso terapéutico , Complemento C5/deficiencia , Citotoxicidad Inmunológica/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Eritrocitos/inmunología , Eritrocitos/patología , Hemoglobinuria Paroxística/tratamiento farmacológico , Humanos , Modelos Inmunológicos , Unión Proteica/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...