Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Clin Infect Dis ; 76(8): 1476-1482, 2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-36445792

RESUMEN

BACKGROUND: Sotrovimab is an anti-spike neutralization monoclonal antibody developed to reduce the risk of coronavirus disease 2019 (COVID-19) progression and advancement to hospitalization in high-risk patients. Currently, there is limited research describing the association of sotrovimab treatment in patients with hematologic malignancy and the predictive factors of hospitalization. METHODS: We performed an observational study of 156 consecutive cancer patients who received sotrovimab at Memorial Sloan Kettering Cancer Center in New York City during the BA.1 Omicron surge. We evaluated the demographic, clinical, and laboratory characteristics of the patients who had subsequent COVID-19-related hospitalization(s) compared to those who did not. RESULTS: Among the 156 study patients, 17 (11%) were hospitalized, of whom 4 were readmitted for COVID-19-related complications; 3 deaths were attributed to COVID-19. Results from multivariable logistic regression show that significant factors associated with hospitalization include patients on anti-CD20 therapy (adjusted odds ratio [aOR], 5.59 [95% confidence interval {CI}, 1.73-18.12]; P = .004) and with relapse/refractory disease (aOR, 5.69 [95% CI, 1.69-19.16]; P = .005). Additionally, whole genome sequencing of severe acute respiratory syndrome coronavirus 2 detected high occurrences of mutations in the spike gene associated with treatment-related resistance longitudinal samples from 11 patients treated with sotrovimab. CONCLUSIONS: While sotrovimab is effective at reducing COVID-19 hospitalization and disease severity in patients with hematologic malignancy when administered early, patients who received anti-CD20 antibodies showed substantial morbidity. Due to the high potential for resistance mutation to sotrovimab and increased morbidity in patients on anti-CD20 therapy, combination treatment should be explored to determine whether it provides added benefits compared to monotherapy.


Asunto(s)
COVID-19 , Neoplasias Hematológicas , Humanos , Recurrencia Local de Neoplasia , Neoplasias Hematológicas/complicaciones , Neoplasias Hematológicas/tratamiento farmacológico , Anticuerpos Neutralizantes , Hospitalización
2.
Haematologica ; 108(11): 3058-3067, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37345467

RESUMEN

AZD7442 (tixagevimab-cilgavimab) is a combination of two human monoclonal antibodies for pre-exposure prophylaxis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection among high-risk patients who do not mount a reliable vaccine response. Foremost among these are hematologic malignancy patients with limited clinical trial or realworld experience to assess the effectiveness of this combination treatment since the emergence of Omicron and its subvariants. We performed a retrospective study of 892 high-risk hematologic malignancy patients who received AZD7442 at Memorial Sloan Kettering Cancer Center in New York City from January 1, 2022 to July 31, 2022. We evaluated demographic, clinical, and laboratory characteristics and performed regression analyses to evaluate risk factors for breakthrough infection. We also evaluated the impact of updated AZD7442 dosing regimens on the risk of breakthrough infection. Among 892 patients, 98 (10.9%) had a breakthrough infection during the study period. A majority received early outpatient treatment (82%) and eventually eight (8.2%) required hospitalization for management of Coronavirus Disease 2019 (COVID-19), with a single instance of severe COVID-19 and death. Patients who received a repeat dose or a higher firsttime dose of AZD7442 had a lower incidence of breakthrough infection. Univariate analyses did not reveal any significant predictors of breakthrough infection. While AZD7442 is effective at reducing SARS-CoV-2 breakthrough infection in patients with hematologic malignancies, no risk factors reliably predicted risk of infection. Patients who received updated dosing regimens as per Food and Drug Administration guidelines had better protection against breakthrough infection.


Asunto(s)
COVID-19 , Neoplasias Hematológicas , Profilaxis Pre-Exposición , Humanos , COVID-19/prevención & control , SARS-CoV-2 , Infección Irruptiva , Estudios Retrospectivos , Anticuerpos Monoclonales , Neoplasias Hematológicas/complicaciones , Neoplasias Hematológicas/tratamiento farmacológico
3.
Br J Anaesth ; 127(1): 153-163, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34006377

RESUMEN

BACKGROUND: Nerve damage is consistently demonstrated after subepineural injection in animal studies, but not after purposeful injection in patients participating in clinical studies. There is a need to better visualise nerves in order to understand the structural changes that occur during subepineural injection. METHODS: We scanned the brachial plexuses of three anaesthetised pigs using micro-ultrasound imaging (55-22 MHz probe), inserted 21 gauge block needles into the radial, median, and axillary nerves, and injected two 0.5 ml boluses of saline into nerves at a rate of 12 ml min-1. Our objectives were to measure the area and diameter of nerves and fascicles, and to describe changes in nerve anatomy, comparing our findings with histology. RESULTS: Images were acquired at 42 sites across 18 nerves in three pigs and compared dimensions (geometric ratio; 95% confidence interval; P value). As expected, the nerve cross-sectional area was greater in the proximal brachial plexus compared with the mid-plexus (2.10; 1.07-4.11; P<0.001) and the distal plexus (2.64; 1.42-4.87; P<0.001). Nerve area expanded after 0.5 ml injection (2.13; 1.48-3.08; P<0.001). Using microultrasound, subepineural injection was characterised by nerve and fascicle rotation, uniform, or localised swelling and epineural rupture. Micro-ultrasound revealed a unique pattern suggestive of subperineural injection after a median nerve injection, and good face validity with histology. Histology showed epineural trauma and inflammation to the perineurium. CONCLUSION: We accurately identified fascicles and real-time structural changes to peripheral nerves using micro-ultrasound. This is the first study to visualise in vivo and in real-time the motion of nerves and fascicles in response to anaesthetic needle insertion and fluid injection.


Asunto(s)
Bloqueo del Plexo Braquial/métodos , Plexo Braquial/diagnóstico por imagen , Sistemas de Computación , Transductores , Ultrasonografía Intervencional/métodos , Adyuvantes Anestésicos/administración & dosificación , Anestésicos Disociativos/administración & dosificación , Animales , Plexo Braquial/efectos de los fármacos , Masculino , Nervios Periféricos/diagnóstico por imagen , Nervios Periféricos/efectos de los fármacos , Porcinos
4.
Biochim Biophys Acta ; 1862(4): 556-565, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26692170

RESUMEN

Cervical cancer is caused by infections with human papillomaviruses (HPV) and genetic alternations in the cervical epithelium. While the former is well studied, the latter remains unclear. We report here that CYB5D2/Neuferricin possesses tumor suppressing activity towards cervical tumorigenesis. Ectopic expression of CYB5D2 did not affect HeLa cell proliferation and the cell's ability to form xenograft tumors, but significantly inhibited HeLa cell invasion in vitro and the cell-produced lung metastasis in NOD/SCID mice. Knockdown of CYB5D2 enhanced HeLa cell invasion. Two mutations in CYB5D2, the substitutions of arginine (R) 7 with either proline (P) or glycine (G), were reported in colon cancer. Both CYB5D2(R7P) and CYB5D2(R7G) were incapable of inhibiting HeLa cell invasion. CYB5D2 binds heme, in which aspartate (D) 86 is required. While CYB5D2(D86G) is heme-binding defective, it inhibited HeLa cell invasion. On the other hand, CYB5D2(R7P) and CYB5D2(R7G) bound heme but did not inhibit HeLa cell invasion. Collectively, CYB5D2 inhibits HeLa cell invasion independently of its heme binding. Furthermore, immunohistochemistry examination of CYB5D2 expression in 20 normal cervical tissues and 40 cervical squamous cell carcinomas (SCC) revealed a CYB5D2 reduction in 87.5% (35/40) of SCC. Analysis of CYB5D2 gene expression and genomic alteration data available from Oncomeine™ detected significant reductions of CYB5D2 mRNA in 40 SCCs and CYB5D2 gene copy number in 107 SCCs. Collectively, we provide evidence that CYB5D2 is a candidate tumor suppressor of cervical tumorigenesis.


Asunto(s)
Citocromos b5/biosíntesis , Regulación Neoplásica de la Expresión Génica , Proteínas Supresoras de Tumor/biosíntesis , Neoplasias del Cuello Uterino/enzimología , Animales , Citocromos b5/genética , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Invasividad Neoplásica , Proteínas Supresoras de Tumor/genética , Neoplasias del Cuello Uterino/genética
5.
Exp Cell Res ; 328(1): 132-142, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25149681

RESUMEN

Despite the development of chemoresistance as a major concern in prostate cancer therapy, the underlying mechanisms remain elusive. In this report, we demonstrate that DU145-derived prostate cancer stem cells (PCSCs) progress slowly with more cells accumulating in the G1 phase in comparison to DU145 non-PCSCs. Consistent with the important role of the AKT pathway in promoting G1 progression, DU145 PCSCs were less sensitive to growth factor-induced activation of AKT in comparison to non-PCSCs. In response to etoposide (one of the most commonly used chemotherapeutic drugs), DU145 PCSCs survived significantly better than non-PCSCs. In addition to etoposide, PCSCs demonstrated increased resistance to docetaxel, a taxane drug that is commonly used to treat castration-resistant prostate cancer. Etoposide produced elevated levels of γH2AX and triggered a robust G2/M arrest along with a coordinated reduction of the G1 population in PCSCs compared to non-PCSCs, suggesting that elevated γH2AX plays a role in the resistance of PCSCs to etoposide-induced cytotoxicity. We have generated xenograft tumors from DU145 PCSCs and non-PCSCs. Consistent with the knowledge that PCSCs produce xenograft tumors with more advanced features, we were able to demonstrate that PCSC-derived xenograft tumors displayed higher levels of γH2AX and p-CHK1 compared to non-PCSC-produced xenograft tumors. Collectively, our research suggests that the elevation of DNA damage response contributes to PCSC-associated resistance to genotoxic reagents.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Etopósido/farmacología , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/patología , Animales , Western Blotting , Ensayo de Unidades Formadoras de Colonias , Daño del ADN/genética , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Células Tumorales Cultivadas
6.
Biochim Biophys Acta ; 1833(1): 90-100, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23098854

RESUMEN

Non-homologous end joining (NHEJ) is one of the major pathways that repairs double-stranded DNA breaks (DSBs). Activation of DNA-PK is required for NHEJ. However, the mechanism leading to DNA-PKcs activation remains incompletely understood. We provide evidence here that the MEK-ERK pathway plays a role in DNA-PKcs-mediated NHEJ. In comparison to the vehicle control (DMSO), etoposide (ETOP)-induced DSBs in MCF7 cells were more rapidly repaired in the presence of U0126, a specific MEK inhibitor, based on the reduction of γH2AX and tail moments. Additionally, U0126 increased reactivation of luciferase activity, which resulted from the repair of restriction enzyme-cleaved DSBs. Furthermore, while inhibition of ERK activation using the dominant-negative MEK1K97M accelerated the repair of DSBs, enforcing ERK activation with the constitutively active MEK1Q56P reduced DSB repair. In line with MEK activating ERK1 and ERK2 kinases, knockdown of either ERK1 or ERK2 increased DSB repair. Consistent with the activation of DNA-PKcs being required for NHEJ, we demonstrated that inhibition of ERK activation using U0126, MEK1K97M, and knockdown of ERK1 or ERK2 enhanced ETOP-induced activation of DNA-PKcs. Conversely, enforcing ERK activation by MEK1Q56P reduced ETOP-initiated DNA-PKcs activation. Taken together, we demonstrate that ERK reduces NHEJ-mediated repair of DSBs via attenuation of DNA-PKcs activation.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Proteínas Nucleares/agonistas , ARN Interferente Pequeño/farmacología , Antineoplásicos Fitogénicos/farmacología , Proteína Quinasa Activada por ADN/metabolismo , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Etopósido/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Nucleares/metabolismo , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos
7.
Cancer Invest ; 32(7): 330-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24884829

RESUMEN

Pyruvate kinase M2 (PKM2) is essential for aerobic glycolysis, the dominant metabolic pathway utilized by cancer cells. To determine the association of PKM2 with prostate cancer (PC), we examined 29 primary PC and three lymph node metastatic tumors; elevation of PKM2 was observed in Gleason 8-10 tumors compared to Gleason 6-7 carcinomas. High PKM2 was detected by immunohistochemistry in more aggressive xenograft tumors derived from PC stem-like cells (PCSCs) compared to those produced from non-PCSCs. While PCSCs and non-PCSCs expressed comparable levels of PKM2, distinct posttranslational modifications were observed. Collectively, upregulation and specific modification to PKM2 associate with PC progression.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de la Membrana/genética , Neoplasias de la Próstata/patología , Hormonas Tiroideas/genética , Anciano , Animales , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proliferación Celular , Predisposición Genética a la Enfermedad , Glucólisis/genética , Humanos , Metástasis Linfática , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Clasificación del Tumor , Trasplante de Neoplasias , Neoplasias de la Próstata/genética , Procesamiento Proteico-Postraduccional , Hormonas Tiroideas/metabolismo , Proteínas de Unión a Hormona Tiroide
8.
Sci Rep ; 14(1): 10276, 2024 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704483

RESUMEN

Fuchs endothelial corneal dystrophy (FECD) is a complex corneal disease characterized by the progressive decline and morphological changes of corneal endothelial cells (CECs) that leads to corneal edema and vision loss. The most common mutation in FECD is an intronic CTG repeat expansion in transcription factor 4 (TCF4) that leads to its altered expression. Corneal endothelial wound healing occurs primarily through cell enlargement and migration, and FECD CECs have been shown to display increased migration speeds. In this study, we aim to determine whether TCF4 can promote cellular migration in FECD CECs. We generated stable CEC lines derived from FECD patients that overexpressed different TCF4 isoforms and investigated epithelial-to-mesenchymal (EMT) expression, morphological analysis and cellular migration speeds. We found that full length TCF4-B isoform overexpression promotes cellular migration in FECD CECs in an EMT-independent manner. RNA-sequencing identified several pathways including the negative regulation of microtubules, with TUBB4A (tubulin beta 4A class IVa) as the top upregulated gene. TUBB4A expression was increased in FECD ex vivo specimens, and there was altered expression of cytoskeleton proteins, tubulin and actin, compared to normal healthy donor ex vivo specimens. Additionally, there was increased acetylation and detyrosination of microtubules in FECD supporting that microtubule stability is altered in FECD and could promote cellular migration. Future studies could be aimed at investigating if targeting the cytoskeleton and microtubules would have therapeutic potential for FECD by promoting cellular migration and regeneration.


Asunto(s)
Movimiento Celular , Endotelio Corneal , Distrofia Endotelial de Fuchs , Microtúbulos , Factor de Transcripción 4 , Humanos , Distrofia Endotelial de Fuchs/genética , Distrofia Endotelial de Fuchs/metabolismo , Distrofia Endotelial de Fuchs/patología , Movimiento Celular/genética , Microtúbulos/metabolismo , Factor de Transcripción 4/metabolismo , Factor de Transcripción 4/genética , Endotelio Corneal/metabolismo , Endotelio Corneal/patología , Masculino , Femenino , Transición Epitelial-Mesenquimal/genética , Anciano , Células Endoteliales/metabolismo , Células Endoteliales/patología , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/genética , Persona de Mediana Edad , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética
9.
Can J Ophthalmol ; 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38513716

RESUMEN

OBJECTIVE: To investigate endothelial cell loss (ECL) associated with Descemet membrane endothelial keratoplasty (DMEK) donor tissues preloaded in the DMEK RAPID transport system after 1 and 5 days and to compare prestamping with 2 different F-mark inks. METHODS: DMEK donor tissues were stripped, marked with gentian violet dye applied as an F-mark, trephined, stained with trypan blue, and then preloaded into the DMEK RAPID transport system by an eye bank technician. Preloaded DMEK tissues were then unfolded and stained with calcein AM after 1 or 5 days of storage. Tissues were imaged, analyzed for total tissue ECL, and immunostained for corneal endothelium markers zonular occludens-1 and xCD166. Additionally, ECL and the intensity of an F-mark caused by 2 different inks were quantified. RESULTS: Preloaded DMEK tissues displayed an average ECL of 11.9% ± 4.5% (n = 8) at 1 day and 9.9% ± 4.2% (n = 9) at 5 days. No difference was found between the 2 groups. Zonular occludens-1 and activated leukocyte cell adhesion molecule (ALCAM; also know as CD166) staining showed that the corneal endothelial monolayer remained intact on preloaded tissues. On 5-day preloaded DMEK tissues, the average ECL and mean grayscale caused by the Keir Surgical ink F-mark and the Cardinal Health ink F-mark were 4.3% ± 0.8% and 158.5 ± 13.9% and 5.0% ± 1.1% and 142.9% ± 20.0%, respectively. No difference was found between the F-mark inks. CONCLUSION: Preloaded DMEK donor tissues resulted in an acceptable ECL range after 1 and 5 days of storage and were deemed suitable for transplantation. Both F-mark inks are acceptable for prestamping preloaded DMEK tissues prior to surgical transplantation with comparable ECL and intensities.

10.
Biochim Biophys Acta ; 1822(6): 875-84, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22406297

RESUMEN

Loss of IQGAP2 contributes to the tumorigenesis of hepatocellular carcinoma and gastric cancer. However, whether IQGAP2 also suppresses prostate tumorigenesis remains unclear. We report here that IQGAP2 is a candidate tumour suppressor of prostate cancer (PC). Elevated IQGAP2 was detected in prostatic intraepithelial neoplasia (PIN), early stages of PCs (Gleason score ≤3), and androgen-dependent LNCaP PC cells. However, IQGAP2 was expressed at substantially reduced levels not only in prostate glands and non-tumorigenic BPH-1 prostate epithelial cells but also in advanced (Gleason score 4 or 5) and androgen-independent PCs. Furthermore, xenograft tumours that were derived from stem-like DU145 cells displayed advanced features and lower levels of IQGAP2 in comparison to xenograft tumours that were produced from non stem-like DU145 cells. Collectively, these results suggest that IQGAP2 functions in the surveillance of prostate tumorigenesis. Consistent with this concept, ectopic IQGAP2 reduced the proliferation of DU145, PC3, and 293T cells as well as the invasion ability of DU145 cells. While ectopic IQGAP2 up-regulated E-cadherin in DU145 and PC3 cells, knockdown of IQGAP2 reduced E-cadherin expression. In primary PC and DU145 cells-derived xenograft tumours, the majority of tumours with high levels of IQGAP2 were strongly-positive for E-cadherin. Therefore, IQGAP2 may suppress PC tumorigenesis, at least in part, by up-regulation of E-cadherin. Mechanistically, overexpression of IQGAP2 significantly reduced AKT activation in DU145 cells and inhibition of AKT activation upregulated E-cadherin, suggesting that IQGAP2 increases E-cadherin expression by inhibiting AKT activation. Taken together, we demonstrate here that IQGAP2 is a candidate tumour suppressor of PC.


Asunto(s)
Transformación Celular Neoplásica , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Cadherinas/biosíntesis , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Próstata/metabolismo , Próstata/patología , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasia Intraepitelial Prostática/patología , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Trasplante Heterólogo , Proteínas Supresoras de Tumor/genética
11.
Infect Control Hosp Epidemiol ; 44(5): 821-823, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-35506167

RESUMEN

We used a self-reporting system to compare symptom frequency of hospital personnel with coronavirus disease 2019 before and after the emergence of the Omicron variant. Omicron was more likely to result in asymptomatic carriage (7% vs 12%; P = .009), and fewer symptoms were observed in those with booster vaccination.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Personal de Hospital , Atención a la Salud , Prueba de COVID-19
12.
Artículo en Inglés | MEDLINE | ID: mdl-33053737

RESUMEN

Triclosan was introduced into the market in the 1970s and has since been used as an antimicrobial agent in a diverse array of consumer and personal care products. Although it has been widely used over a number of years, there is growing concern and debate over its safety and efficacy and its potential as an endocrine disruptor. Although prior animal toxicology studies have shown an association between triclosan and decreased testosterone levels, human studies have been limited, particularly for adult men. Using the National Health and Nutrition Examination Survey data (NHANES, 2011-2012), we examined the association of urinary triclosan on testosterone levels in adult men 18-65 years of age. Multivariable linear regression analysis failed to show an association between triclosan and serum testosterone (ß = 0.0003, p = 0.98, 95% CI = -0.024, 0.025). The results suggest there is no association or that triclosan concentrations are too low to cause a significant impact on testosterone levels. Additionally, longitudinal studies would provide a more comprehensive understanding of the direction of change and magnitude of causal relationships over time.


Asunto(s)
Disruptores Endocrinos , Testosterona , Triclosán , Adolescente , Adulto , Anciano , Disruptores Endocrinos/toxicidad , Disruptores Endocrinos/orina , Humanos , Modelos Lineales , Masculino , Persona de Mediana Edad , Encuestas Nutricionales , Testosterona/sangre , Triclosán/toxicidad , Triclosán/orina , Adulto Joven
13.
Ultrasound Med Biol ; 46(2): 359-368, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31708270

RESUMEN

Gas vesicles (GVs) are nanosized structures (45-800 nm) and have been reported to produce non-linear contrast signals, making them an attractive agent for molecular targeting of tumors. One barrier to their use for pre-clinical oncology studies is rapid uptake into the reticuloendothelial system (RES) and consequent rapid decrease in contrast signal after infusion ends and low signal on reperfusion after a bubble burst sequence. The purpose of this study was to examine suppression of the RES and surface modification of GVs to prolong contrast circulation in tumors for ultrasound imaging. Ultrasound imaging to measure dynamics of contrast signal intensity in tumor models was carried out using a 21-MHz high-frequency array transducer with the Vevo 2100 ultrasound system. The non-linear contrast signal from intravenously injected GVs compared with peak enhancement was measured during contrast wash-out and on reperfusion after a contrast burst sequence. Disrupting the RES by saturating the macrophage population or chemically inhibiting the Kupffer cell population with gadolinium or Intralipid preserves 62%-88% of GVs' contrast enhancement relative to peak during the wash-out phase and 32%-56% on reperfusion compared with 38% and 14%, respectively, for no disruption of the RES, indicating longer circulation of GVs in the tumor. Additionally, coating the GVs with 2-, 5- or 10-kDa polyethylene glycol (PEG) chains resulted in >70% contrast signal retention in the tumors during wash-out and, for 5- or 10-kDa PEG chains, a return to >45% of peak contrast signal on reperfusion. These findings indicate that GVs can be used as a contrast agent for tumor imaging and that disruption of the RES improved recirculation and maintained contrast enhancement caused by GVs in the tumors.


Asunto(s)
Medios de Contraste/administración & dosificación , Medios de Contraste/farmacocinética , Gases , Microburbujas , Sistema Mononuclear Fagocítico/metabolismo , Neoplasias/diagnóstico por imagen , Línea Celular Tumoral , Humanos , Neoplasias/patología , Polietilenglicoles , Ultrasonografía/métodos
15.
Transl Oncol ; 11(4): 1012-1022, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29966864

RESUMEN

Butyrylcholinesterase (BChE) is a plasma enzyme that hydrolyzes ghrelin and bioactive esters, suggesting a role in modulating metabolism. Serum BChE is reduced in cancer patients. In prostate cancer (PC), the down-regulation is associated with disease recurrence. Nonetheless, how BChE is expressed in PC and its impact on PC remain unclear. We report here the biphasic changes of BChE expression in PC. In vitro, BChE expression was decreased in more tumorigenic PC stem-like cells (PCSLCs), DU145, and PC3 cells compared to less tumorigenic non-stem PCs and LNCaP cells. On the other hand, BChE was expressed at a higher level in LNCaP cells than immortalized but non-tumorigenic prostate epithelial BPH-1 cells. In vivo, BChE expression was up-regulated in DU145 xenografts compared to LNCaP xenografts; DU145 cell-derived lung metastases displayed comparable levels of BChE as subcutaneous tumors. Furthermore, LNCaP xenografts produced in castrated mice exhibited a significant increase of BChE expression compared to xenografts generated in intact mice. In patients, BChE expression was down-regulated in PCs (n = 340) compared to prostate tissues (n = 86). In two independent PC populations MSKCC (n = 130) and TCGA Provisional (n = 490), BChE mRNA levels were reduced from World Health Organization grade group 1 (WHOGG 1) PCs to WHOGG 3 PCs, followed by a significant increase in WHOGG 5 PCs. The up-regulation was associated with a reduction in disease-free survival (P = .008). Collectively, we demonstrated for the first time a biphasic alteration of BChE, its down-regulation at early stage of PC and its up-regulation at advanced PC.

16.
Nat Protoc ; 12(10): 2050-2080, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28880278

RESUMEN

Gas vesicles (GVs) are a unique class of gas-filled protein nanostructures that are detectable at subnanomolar concentrations and whose physical properties allow them to serve as highly sensitive imaging agents for ultrasound and MRI. Here we provide a protocol for isolating GVs from native and heterologous host organisms, functionalizing these nanostructures with moieties for targeting and fluorescence, characterizing their biophysical properties and imaging them using ultrasound and MRI. GVs can be isolated from natural cyanobacterial and haloarchaeal host organisms or from Escherichia coli expressing a heterologous GV gene cluster and purified using buoyancy-assisted techniques. They can then be modified by replacing surface-bound proteins with engineered, heterologously expressed variants or through chemical conjugation, resulting in altered mechanical, surface and targeting properties. Pressurized absorbance spectroscopy is used to characterize their mechanical properties, whereas dynamic light scattering (DLS)and transmission electron microscopy (TEM) are used to determine nanoparticle size and morphology, respectively. GVs can then be imaged with ultrasound in vitro and in vivo using pulse sequences optimized for their detection versus background. They can also be imaged with hyperpolarized xenon MRI using chemical exchange saturation transfer between GV-bound and dissolved xenon-a technique currently implemented in vitro. Taking 3-8 d to prepare, these genetically encodable nanostructures enable multimodal, noninvasive biological imaging with high sensitivity and potential for molecular targeting.


Asunto(s)
Medios de Contraste/química , Imagen por Resonancia Magnética/métodos , Nanoestructuras/química , Ultrasonografía/métodos , Escherichia coli , Microscopía Electrónica de Transmisión
17.
Oncotarget ; 8(12): 19218-19235, 2017 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-28186973

RESUMEN

Although the FAM84B gene lies within chromosome 8q24, a locus frequently altered in prostate cancer (PC), its alteration during prostate tumorigenesis has not been well studied. We report here FAM84B upregulation in DU145 cell-derived prostate cancer stem-like cells (PCSLCs) and DU145 cell-produced lung metastases compared to subcutaneous xenograft tumors. FAM84B protein was detected in bone metastases and primary PCs. Nanostring examination of 7 pairs of tumor adjacent normal and PC tissues revealed elevations in FAM84B mRNA levels in all carcinomas. Furthermore, through analysis of FAM84B expression using large datasets within the Gene Expression Omnibus and OncomineTM database, we demonstrate significant increases in FAM84B mRNA in 343 primary PCs versus 181 normal tissues, and elevations in the FAM84B gene copy number (GCN) in 171 primary PCs versus 61 normal tissues. While FAM84B was not detected at higher levels via immunohistochemistry in high grade (Gleason score/GS 8-10) tumors compared to GS6-7 PCs, analyses of FAM84B mRNA and GCN using datasets within the cBioPortal database demonstrated FAM84B upregulation in 12% (67/549) of primary PCs and 18% (73/412) of metastatic castration resistant PCs (mCRPCs), and GCN increases in 4.8% (26/546) of primary PCs and 26% (121/467) of mCRPCs, revealing an association of the aforementioned changes with CRPC development. Of note, an increase in FAM84B expression was observed in xenograft CRPCs produced by LNCaP cells. Furthermore, FAM84B upregulation and GCN increases correlate with decreases in disease free survival and overall survival. Collectively, we demonstrate a novel association of FAM84B with PC tumorigenesis and CRPC progression.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/secundario , Transformación Celular Neoplásica/patología , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/patología , Neoplasias de la Próstata Resistentes a la Castración/patología , Neoplasias de la Próstata/patología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Progresión de la Enfermedad , Humanos , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos NOD , Ratones SCID , Clasificación del Tumor , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Pronóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Data Brief ; 6: 811-6, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26937452

RESUMEN

We have recently reported that CYB5D2 plays a role in suppression of cervical cancer tumorigenesis, "CYB5D2 displays tumor suppression activities towards cervical cancer" [1]. We provide the accompany data here describing the effects of CYB5D2 overexpression and addition of recombinant CYB5D2 on HeLa cell cycle distribution. Furthermore, we will present the conditions used to specifically determine CYB5D2 expression in primary cervical and cervical cancer tissues using immunohistochemistry (IHC) and the patient cohort involved in assessing the CYB5D2 protein levels in primary cervical and cervical cancer tissues.

19.
Oncotarget ; 7(50): 83115-83133, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27825118

RESUMEN

Evidence supports the upregulation of MUC1 in prostate cancer (PC). However, this has not been thoroughly investigated. We report here an association of MUC1 upregulation with PC metastasis and the development of castration resistant PC (CRPC). MUC1 expression was specifically increased in DU145 cell-derived PC stem-like cells (PCSLCs) in comparison to their non-PCSLCs counterparts. While immunohistochemistry staining of 34 primary PCs revealed variability in MUC1 expression, Nanostring technology demonstrated elevated MUC1 mRNA levels in 4 of 7 PCs compared to their normal matched tissues. By analyzing MUC1 mRNA levels and gene copy number (GCN) using the OncomineTM database, elevations in MUC1 mRNA in 82 metastases versus 280 primary PCs and in MUC1 GCN in 37 metastases over 181 primary tumors were demonstrated. Analysis of genomic datasets within cBioPortal revealed increases in MUC1 GCN in 2% (6/333) of primary PCs, 6% (9/150) of metastatic PCs, and 33% (27/82) of CRPCs; in comparison, the respective increase in androgen receptor (AR) GCN was 1%, 63%, and 56%, revealing a specific increase in MUC1 GCN for CRPC. Furthermore, a 25-gene MUC1 network was amplified in 52% of CRPCs compared to 69% of CRPCs displaying increases in an AR co-regulator group. While genomic alterations in the MUC1 network largely overlap with those in the AR group, 18 CRPCs (66.7% being neuroendocrine PC) showed genomic alterations only in the MUC1 network. Moreover, genomic alterations in the MUC1 network correlated with PC relapse. Collectively, our observations suggest a combination therapy involving MUC1-based immunotherapy and androgen deprivation.


Asunto(s)
Adenocarcinoma/genética , Biomarcadores de Tumor/genética , Amplificación de Genes , Mucina-1/genética , Tumores Neuroendocrinos/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/secundario , Adenocarcinoma/terapia , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos Fitogénicos/farmacología , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Biología Computacional , Bases de Datos Genéticas , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Docetaxel , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Mucina-1/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/secundario , Tumores Neuroendocrinos/terapia , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Neoplasias de la Próstata Resistentes a la Castración/terapia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal , Análisis de Supervivencia , Taxoides/farmacología , Factores de Tiempo , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Res ; 76(6): 1603-14, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26795349

RESUMEN

Prostate cancer metastasis is the main cause of disease-related mortality. Elucidating the mechanisms underlying prostate cancer metastasis is critical for effective therapeutic intervention. In this study, we performed gene-expression profiling of prostate cancer stem-like cells (PCSC) derived from DU145 human prostate cancer cells to identify factors involved in metastatic progression. Our studies revealed contactin 1 (CNTN1), a neural cell adhesion protein, to be a prostate cancer-promoting factor. CNTN1 knockdown reduced PCSC-mediated tumor initiation, whereas CNTN1 overexpression enhanced prostate cancer cell invasion in vitro and promoted xenograft tumor formation and lung metastasis in vivo. In addition, CNTN1 overexpression in DU145 cells and corresponding xenograft tumors resulted in elevated AKT activation and reduced E-cadherin (CDH1) expression. CNTN1 expression was not readily detected in normal prostate glands, but was clearly evident on prostate cancer cells in primary tumors and lymph node and bone metastases. Tumors from 637 patients expressing CNTN1 were associated with prostate cancer progression and worse biochemical recurrence-free survival following radical prostatectomy (P < 0.05). Collectively, our findings demonstrate that CNTN1 promotes prostate cancer progression and metastasis, prompting further investigation into the mechanisms that enable neural proteins to become aberrantly expressed in non-neural malignancies.


Asunto(s)
Adhesión Celular/genética , Contactina 1/genética , Metástasis de la Neoplasia/genética , Moléculas de Adhesión de Célula Nerviosa/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Animales , Neoplasias Óseas/genética , Cadherinas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia/patología , Células Madre Neoplásicas/patología , Próstata/patología , Proteínas Proto-Oncogénicas c-akt/genética , Transcriptoma/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda