Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 148(1)2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33318148

RESUMEN

Androgens/androgen receptor (AR)-mediated signaling pathways are essential for prostate development, morphogenesis and regeneration. Specifically, stromal AR signaling has been shown to be essential for prostatic initiation. However, the molecular mechanisms underlying AR-initiated mesenchymal-epithelial interactions in prostate development remain unclear. Here, using a newly generated mouse model, we have directly addressed the fate and role of genetically marked AR-expressing cells during embryonic prostate development. Androgen signaling-initiated signaling pathways were identified in mesenchymal niche populations at single-cell transcriptomic resolution. The dynamic cell-signaling networks regulated by stromal AR were additionally characterized in relation to prostatic epithelial bud formation. Pseudotime analyses further revealed the differentiation trajectory and fate of AR-expressing cells in both prostatic mesenchymal and epithelial cell populations. Specifically, the cellular properties of Zeb1-expressing progenitors were assessed. Selective deletion of AR signaling in a subpopulation of mesenchymal rather than epithelial cells dysregulated the expression of the master regulators and significantly impaired prostatic bud formation. These data provide novel, high-resolution evidence demonstrating the important role of mesenchymal androgen signaling in the cellular niche controlling prostate early development by initiating dynamic mesenchyme-epithelia cell interactions.


Asunto(s)
Andrógenos/farmacología , Comunicación Celular , Linaje de la Célula , Próstata/citología , Análisis de la Célula Individual , Animales , Comunicación Celular/efectos de los fármacos , Comunicación Celular/genética , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes del Desarrollo , Masculino , Mesodermo/citología , Ratones , Próstata/efectos de los fármacos , RNA-Seq , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
2.
Development ; 148(19)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34427305

RESUMEN

Stromal androgen-receptor (AR) action is essential for prostate development, morphogenesis and regeneration. However, mechanisms underlying how stromal AR maintains the cell niche in support of pubertal prostatic epithelial growth are unknown. Here, using advanced mouse genetic tools, we demonstrate that selective deletion of stromal AR expression in prepubescent Shh-responsive Gli1-expressing cells significantly impedes pubertal prostate epithelial growth and development. Single-cell transcriptomic analyses showed that AR loss in these prepubescent Gli1-expressing cells dysregulates androgen signaling-initiated stromal-epithelial paracrine interactions, leading to growth retardation of pubertal prostate epithelia and significant development defects. Specifically, AR loss elevates Shh-signaling activation in both prostatic stromal and adjacent epithelial cells, directly inhibiting prostatic epithelial growth. Single-cell trajectory analyses further identified aberrant differentiation fates of prostatic epithelial cells directly altered by stromal AR deletion. In vivo recombination of AR-deficient stromal Gli1-lineage cells with wild-type prostatic epithelial cells failed to develop normal prostatic epithelia. These data demonstrate previously unidentified mechanisms underlying how stromal AR-signaling facilitates Shh-mediated cell niches in pubertal prostatic epithelial growth and development.


Asunto(s)
Andrógenos/metabolismo , Proteínas Hedgehog/metabolismo , Próstata/crecimiento & desarrollo , Nicho de Células Madre , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas Hedgehog/genética , Masculino , Ratones , Próstata/citología , Próstata/metabolismo , RNA-Seq , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Transcriptoma , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
3.
PLoS Genet ; 16(1): e1008588, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31929563

RESUMEN

Prostate embryonic development, pubertal and adult growth, maintenance, and regeneration are regulated through androgen signaling-mediated mesenchymal-epithelial interactions. Specifically, the essential role of mesenchymal androgen signaling in the development of prostate epithelium has been observed for over 30 years. However, the identity of the mesenchymal cells responsible for this paracrine regulation and related mechanisms are still unknown. Here, we provide the first demonstration of an indispensable role of the androgen receptor (AR) in sonic hedgehog (SHH) responsive Gli1-expressing cells, in regulating prostate development, growth, and regeneration. Selective deletion of AR expression in Gli1-expressing cells during embryogenesis disrupts prostatic budding and impairs prostate development and formation. Tissue recombination assays showed that urogenital mesenchyme (UGM) containing AR-deficient mesenchymal Gli1-expressing cells combined with wildtype urogenital epithelium (UGE) failed to develop normal prostate tissue in the presence of androgens, revealing the decisive role of AR in mesenchymal SHH responsive cells in prostate development. Prepubescent deletion of AR expression in Gli1-expressing cells resulted in severe impairment of androgen-induced prostate growth and regeneration. RNA-sequencing analysis showed significant alterations in signaling pathways related to prostate development, stem cells, and organ morphogenesis in AR-deficient Gli1-expressing cells. Among these altered pathways, the transforming growth factor ß1 (TGFß1) pathway was up-regulated in AR-deficient Gli1-expressing cells. We further demonstrated the activation of TGFß1 signaling in AR-deleted prostatic Gli1-expressing cells, which inhibits prostate epithelium growth through paracrine regulation. These data demonstrate a novel role of the AR in the Gli1-expressing cellular niche for regulating prostatic cell fate, morphogenesis, and renewal, and elucidate the mechanism by which mesenchymal androgen-signaling through SHH-responsive cells elicits the growth and regeneration of prostate epithelium.


Asunto(s)
Proteínas Hedgehog/metabolismo , Morfogénesis , Próstata/metabolismo , Receptores Androgénicos/metabolismo , Regeneración , Transducción de Señal , Animales , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Próstata/citología , Próstata/crecimiento & desarrollo , Próstata/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
4.
PLoS Genet ; 15(10): e1008451, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31658259

RESUMEN

E-cadherin complexes with the actin cytoskeleton via cytoplasmic catenins and maintains the functional characteristics and integrity of the epithelia in normal epithelial tissues. Lost expression of E-cadherin disrupts this complex resulting in loss of cell polarity, epithelial denudation and increased epithelial permeability in a variety of tissues. Decreased expression of E-cadherin has also been observed in invasive and metastatic human tumors. In this study, we investigated the effect of E-cadherin loss in prostatic epithelium using newly developed genetically engineered mouse models. Deletion of E-cadherin in prostatic luminal epithelial cells with modified probasin promoter driven Cre (PB-Cre4) induced the development of mouse prostatic intraepithelial neoplasia (PIN). An increase in levels of cytoplasmic and nuclear ß-catenin appeared in E-cadherin deleted atypical cells within PIN lesions. Using various experimental approaches, we further demonstrated that the knockdown of E-cadherin expression elevated free cytoplasmic and nuclear ß-catenin and enhanced androgen-induced transcription and cell growth. Intriguingly, pathological changes representing prostatic epithelial cell denudation and increased apoptosis accompanied the above PIN lesions. The essential role of E-cadherin in maintaining prostatic epithelial integrity and organization was further demonstrated using organoid culture approaches. To directly assess the role of loss of E-cadherin in prostate tumor progression, we generated a new mouse model with bigenic Cdh1 and Pten deletion in prostate epithelium. Early onset, aggressive tumor phenotypes presented in the compound mice. Strikingly, goblet cell metaplasia was observed, intermixed within prostatic tumor lesions of the compound mice. This study provides multiple lines of novel evidence demonstrating a comprehensive role of E-cadherin in maintaining epithelial integrity during the course of prostate oncogenic transformation, tumor initiation and progression.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Transformación Celular Neoplásica/patología , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/patología , Animales , Antígenos CD/genética , Cadherinas/genética , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Epiteliales , Epitelio , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Cultivo Primario de Células , Próstata/citología , Próstata/patología , Neoplasia Intraepitelial Prostática/genética , Neoplasias de la Próstata/genética , ARN Interferente Pequeño , beta Catenina/genética , beta Catenina/metabolismo
5.
J Biol Chem ; 295(2): 631-644, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31819003

RESUMEN

Co-occurrence of aberrant hepatocyte growth factor (HGF)/MET proto-oncogene receptor tyrosine kinase (MET) and Wnt/ß-catenin signaling pathways has been observed in advanced and metastatic prostate cancers. This co-occurrence positively correlates with prostate cancer progression and castration-resistant prostate cancer development. However, the biological consequences of these abnormalities in these disease processes remain largely unknown. Here, we investigated the aberrant activation of HGF/MET and Wnt/ß-catenin cascades in prostate tumorigenesis by using a newly generated mouse model in which both murine Met transgene and stabilized ß-catenin are conditionally co-expressed in prostatic epithelial cells. These compound mice displayed accelerated prostate tumor formation and invasion compared with their littermates that expressed only stabilized ß-catenin. RNA-Seq and quantitative RT-PCR analyses revealed increased expression of genes associated with tumor cell proliferation, progression, and metastasis. Moreover, Wnt signaling pathways were robustly enriched in prostate tumor samples from the compound mice. ChIP-qPCR experiments revealed increased ß-catenin recruitment within the regulatory regions of the Myc gene in tumor cells of the compound mice. Interestingly, the occupancy of MET on the Myc promoter also appeared in the compound mouse tumor samples, implicating a novel role of MET in ß-catenin-mediated transcription. Results from implanting prostate graft tissues derived from the compound mice and controls into HGF-transgenic mice further uncovered that HGF induces prostatic oncogenic transformation and cell growth. These results indicate a role of HGF/MET in ß-catenin-mediated prostate cancer cell growth and progression and implicate a molecular mechanism whereby nuclear MET promotes aberrant Wnt/ß-catenin signaling-mediated prostate tumorigenesis.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones SCID , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/patología , Proto-Oncogenes Mas
6.
Ann Surg Oncol ; 28(7): 3779-3788, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33231769

RESUMEN

BACKGROUND: Neoadjuvant therapy is increasingly used for patients with pancreatic ductal adenocarcinoma (PDAC). It is unknown whether neoadjuvant chemoradiotherapy is more effective than chemotherapy (NCRT vs. NAC). We aim to compare pathological and survival outcomes of NCRT and NAC in patients with PDAC. PATIENTS AND METHODS: Single-center analysis of PDAC patients treated with NCRT or NAC followed by resection between December 2008 and December 2018 was performed. Average treatment effect (ATE) was estimated after case-control matching using Mahalanobis distance nearest-neighbor matching. Inverse probability weighted estimates (IPWE)-based ATE was estimated for disease-free survival (DFS) and overall survival (OS). RESULTS: Among the 418 patients (mean age 66.8 years, 51% female) included in the study, 327 received NAC and 91 received NCRT. NCRT patients had higher rates of locally advanced disease, number of neoadjuvant chemotherapy cycles, more chemotherapy regimen crossover (gemcitabine and 5-FU based), and were more likely to undergo open surgical procedures and/or vascular resection (all p < 0.05). After matched analysis, NCRT was associated with a significant reduction in lymph node positive disease [ATE = (-)0.24, p = 0.007] and lymphovascular invasion [ATE = (-)0.20, p = 0.02]. While NCRT was associated with significantly improved DFS by 9.5 months (p = 0.006), it did not affect OS by IPWE-based ATE after adjusting for adjuvant therapy (ATE = 5.5 months; p = 0.32). CONCLUSION: Compared with NAC alone, NCRT is associated with improved pathologic surrogates and disease-free survival, but not overall survival in patients with PDAC.


Asunto(s)
Terapia Neoadyuvante , Neoplasias Pancreáticas , Anciano , Quimioradioterapia , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Estadificación de Neoplasias , Neoplasias Pancreáticas/terapia , Estudios Retrospectivos
7.
Ann Surg Oncol ; 28(5): 2438-2446, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33523364

RESUMEN

AIMS: National studies have demonstrated disparities in the treatment and survival of pancreatic cancer patients based on socioeconomic status (SES). This study aimed to identify specific differences in perioperative management and outcomes based on patient SES and to study the role of a multidisciplinary clinic (MDC) in mitigating any variations. METHODS: The study analyzed patients undergoing pancreaticoduodenectomy for pancreatic ductal adenocarcinoma in a large hospital system. The patients were categorized into groups of high and low SES and whether they were managed by the authors' pancreatic cancer MDC or not. The study compared differences in disease characteristics, receipt of multimodality therapy, perioperative outcomes, and recurrence-free and overall survival. RESULTS: Of the 162 low-SES patients and 119 high-SES patients, 54% were managed in the MDC. Outside the MDC, low-SES patients were less likely to receive neoadjuvant chemotherapy and had less minimally invasive surgery, a longer OR time, less enhanced recovery participation, and more major complications (p < 0.05). No SES disparities were observed among the MDC patients. Despite similar tumor characteristics, the low-SES patients had inferior median overall survival (21 vs 32 months; p = 0.005), but the MDC appeared to eliminate this disparity. Low SES correlated with inferior survival for the non-MDC patients (17 vs 32 months; p < 0.001), but not for the MDC patients (24 vs 25 months; p = 0.33). These findings persisted in the multivariable analysis. CONCLUSION: A pancreatic cancer MDC standardizes treatment decisions, eliminates disparities in surgical outcomes, and improves survival for low-SES patients.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/cirugía , Disparidades en Atención de Salud , Humanos , Recurrencia Local de Neoplasia , Pancreatectomía , Neoplasias Pancreáticas/cirugía , Clase Social
8.
Pediatr Blood Cancer ; 68 Suppl 2: e28597, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33818886

RESUMEN

Radiation therapy (RT) is an integral part of the management of many pediatric tumors; however, it is associated with both acute and permanent adverse events that can significantly impact a child's quality of life, lead to treatment delays, and potentially affect outcomes of cancer therapy. Prevention, early detection, and optimal management of these adverse effects will help reduce their impact on the patients' quality of life and overall well-being. Unfortunately, there has not been a coordinated effort to study the etiology, evaluate risk factors, and explore novel treatments for these conditions. Studies of supportive care for children undergoing RT are often small and uncontrolled. This review will focus on the impact of irradiation on the different organ systems and their current management. Further studies are required to improve our understanding of the contributing factors and explore novel treatment options for these adverse effects and to enable children and their families to better cope with some of the unavoidable toxicities following multimodality therapy.


Asunto(s)
Neoplasias/radioterapia , Cuidados Paliativos/métodos , Traumatismos por Radiación/terapia , Tolerancia a Radiación , Radioterapia/efectos adversos , Niño , Humanos , Traumatismos por Radiación/etiología
9.
Differentiation ; 107: 1-10, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30927641

RESUMEN

Androgen signaling is essential for prostate development, morphogenesis, and regeneration. Emerging evidence also indicates a regulatory role of Notch signaling in prostate development, differentiation, and growth. However, the collaborative regulatory mechanisms of androgen and Notch signaling during prostate development, growth, and regeneration are largely unknown. Hairy and Enhancer of Split 1 (Hes1) is a transcriptional regulator of Notch signaling pathways, and its expression is responsive to Notch signaling. Hes1-expressing cells have been shown to possess the regenerative capability to repopulate a variety of adult tissues. In this study, we developed new mouse models to directly assess the role of the androgen receptor in prostatic Hes1-expressing cells. Selective deletion of AR expression in embryonic Hes1-expressing cells impeded early prostate development both in vivo and in tissue xenograft experiments. Prepubescent deletion of AR expression in Hes1-expressing cells resulted in prostate glands containing abnormalities in cell morphology and gland architecture. A population of castration-resistant Hes1-expressing cells was revealed in the adult prostate, with the ability to repopulate prostate epithelium following androgen supplementation. Deletion of AR in Hes1-expressing cells diminishes their regenerative ability. These lines of evidence demonstrate a critical role for the AR in Notch-responsive cells during the course of prostate development, morphogenesis, and regeneration, and implicate a mechanism underlying interaction between the androgen and Notch signaling pathways in the mouse prostate.


Asunto(s)
Próstata/fisiología , Receptores Notch/metabolismo , Regeneración , Factor de Transcripción HES-1 , Andrógenos/metabolismo , Animales , Masculino , Ratones , Modelos Animales , Próstata/embriología , Receptores Androgénicos/metabolismo , Transducción de Señal , Factor de Transcripción HES-1/biosíntesis , Factor de Transcripción HES-1/genética
10.
J Gen Intern Med ; 34(5): 769-772, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30993629

RESUMEN

BACKGROUND: The Accreditation Council for Graduate Medical Education (ACGME) demands that physicians should be trained to engage in clinical activities with other health profession providers. Incorporation of advanced practice providers (APPs) into medicine ward teams has not yet been described. AIM: To describe a pilot and feasibility evaluation of an interprofessional general medicine ward team with internal medicine residents and APPs to encourage resident leadership development, enhance service to education balance, and promote interprofessional collaboration. SETTING: University of Colorado, Internal Medicine Residency Program. PARTICIPANTS: Sixteen internal medicine residents, 16 interns, 19 Department of Medicine faculty members, and 8 advanced practice provider fellows in hospital medicine. PROGRAM DESCRIPTION: The authors describe an interprofessional general medicine ward team including team structure, and roles and responsibilities of each team member. PROGRAM EVALUATION: Each team member completed an electronic survey following the rotation and the majority agreed that the pilot team allowed for an enhanced resident leadership role, and helped to restore the service to education balance and interprofessional collaboration. DISCUSSION: An interprofessional general medicine ward team is feasible, has the potential to optimize service to education balance, and exposes learners to a collaborative interprofessional clinical environment.


Asunto(s)
Enfermería de Práctica Avanzada/organización & administración , Medicina Interna/educación , Grupo de Atención al Paciente/organización & administración , Educación de Postgrado en Medicina/organización & administración , Estudios de Factibilidad , Humanos , Internado y Residencia , Relaciones Interprofesionales , Proyectos Piloto , Evaluación de Programas y Proyectos de Salud , Encuestas y Cuestionarios
11.
Int J Syst Evol Microbiol ; 66(8): 2803-2812, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27045762

RESUMEN

Strains of members of the genus Corynebacterium derived from ophthalmologic patients in Japan, Belgium and Switzerland and found to be closely related to-, but distinguishable from Corynebacterium mastitidis by 16S rRNA gene sequencing, were characterized using biochemical, chemotaxonomic, MALDI-TOF mass spectrometry and antimicrobial susceptibility methods and DNA-DNA hybridization as well as by whole-genome sequencing (WGS). Based on this investigation, we describe Corynebacterium lowii sp. nov. and Corynebacterium oculi sp. nov., derived from human ocular specimens, as well as emend the description of Corynebacterium mastitidis. Type strains for these species are: C. lowii R-50085T (=LMG 28276T =CCUG 65815T) and C. oculi R-50187T (=LMG 28277T =CCUG 65816T). DNA G+C content was found to be 62.2 % (by HPLC) and 62.8 % (by WGS) for C. lowii R-50085T, 64.1 % (HPLC) and 64.8 % (WGS) for C. oculi R-50187T and 67.8 % (HPLC) for C. mastitidis LMG 19040T [=S-8T =CCUG 38654T =CECT 4843T =CIP 105509T =DSM 44356T =IFO (NBRC)16160T =JCM 12269T].


Asunto(s)
Corynebacterium/clasificación , Ojo/microbiología , Filogenia , Técnicas de Tipificación Bacteriana , Composición de Base , Bélgica , Corynebacterium/genética , Corynebacterium/aislamiento & purificación , ADN Bacteriano/genética , Ácidos Grasos/química , Humanos , Japón , Hibridación de Ácido Nucleico , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Suiza
13.
J Cell Biol ; 223(9)2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-38990714

RESUMEN

Dermal fibroblasts deposit type I collagen, the dominant extracellular matrix molecule found in skin, during early postnatal development. Coincident with this biosynthetic program, fibroblasts proteolytically remodel pericellular collagen fibrils by mobilizing the membrane-anchored matrix metalloproteinase, Mmp14. Unexpectedly, dermal fibroblasts in Mmp14-/- mice commit to a large-scale apoptotic program that leaves skin tissues replete with dying cells. A requirement for Mmp14 in dermal fibroblast survival is recapitulated in vitro when cells are embedded within, but not cultured atop, three-dimensional hydrogels of crosslinked type I collagen. In the absence of Mmp14-dependent pericellular proteolysis, dermal fibroblasts fail to trigger ß1 integrin activation and instead actuate a TGF-ß1/phospho-JNK stress response that leads to apoptotic cell death in vitro as well as in vivo. Taken together, these studies identify Mmp14 as a requisite cell survival factor that maintains dermal fibroblast viability in postnatal dermal tissues.


Asunto(s)
Apoptosis , Supervivencia Celular , Fibroblastos , Metaloproteinasa 14 de la Matriz , Animales , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 14 de la Matriz/genética , Fibroblastos/metabolismo , Ratones , Ratones Noqueados , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Integrina beta1/metabolismo , Integrina beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Dermis/metabolismo , Dermis/citología , Células Cultivadas , Matriz Extracelular/metabolismo , Ratones Endogámicos C57BL , Piel/metabolismo
14.
Heart Rhythm ; 21(7): 1154-1160, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38395245

RESUMEN

BACKGROUND: Ventricular arrhythmia (VA) is the primary mechanism of sudden death in patients with structural heart disease. Cardiac stereotactic body radiation therapy (SBRT) delivered to the scar in the left ventricle significantly reduces the burden of VA. OBJECTIVE: The goal of this study was to investigate the impact of SBRT on scar morphology and VA inducibility in a porcine infarct model. METHODS: Myocardial infarction (MI) was created in 10 Yorkshire pigs involving the left anterior descending artery territory. Cardiac positron emission tomography and computed tomography were performed for targeted SBRT. Alternative pigs received SBRT at 25 Gy in a single fraction. The terminal experiment included endocardial mapping, programmed ventricular stimulation, and tissue harvesting. RESULTS: Of the 10 pigs infarcted, 2 died prematurely after MI and 8 (4 MI and 4 MI+SBRT) survived. Mean time from MI to SBRT was 48 ± 12 days, and mean time from SBRT to harvest was 32 ± 12 days. Scar was localized on intracardiac mapping in all pigs, and the scar was denser in the MI+SBRT compared with the MI-only group (33% ± 20% vs 14% ± 11%; P = .07). All 4 MI pigs had inducible VA during programmed stimulation, whereas only 1 of 4 pigs had inducible VA in the MI+SBRT arm (100% vs 25%; P = .07). No myocardial fibrosis was seen in the remote areas in either group. CONCLUSION: SBRT reduced VA inducibility in pigs with scarring after MI. Endocardial mapping revealed denser scar in pigs receiving SBRT compared with those that did not, suggesting that SBRT suppresses VA inducibility through better scar homogenization.


Asunto(s)
Modelos Animales de Enfermedad , Infarto del Miocardio , Radiocirugia , Animales , Radiocirugia/métodos , Radiocirugia/efectos adversos , Porcinos , Infarto del Miocardio/complicaciones , Taquicardia Ventricular/etiología , Taquicardia Ventricular/fisiopatología , Cicatriz/etiología , Ventrículos Cardíacos/fisiopatología , Ventrículos Cardíacos/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos
15.
bioRxiv ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38766091

RESUMEN

Ewing sarcoma (ES) is an aggressive cancer diagnosed in adolescents and young adults. The fusion oncoprotein (EWSR1::FLI1) that drives Ewing sarcoma is known to downregulate TGFBR2 expression (part of the TGFß receptor). Because TGFBR2 is downregulated, it was thought that TGFß likely plays an inconsequential role in Ewing biology. However, the expression of TGFß in the Ewing tumor immune microenvironment (TIME) and functional impact of TGFß in the TIME remains largely unknown given the historical lack of immunocompetent preclinical models. Here, we use single-cell RNAseq analysis of human Ewing tumors to show that immune cells, such as NK cells, are the largest source of TGFß production in human Ewing tumors. We develop a humanized (immunocompetent) mouse model of ES and demonstrate distinct TME signatures and metastatic potential in these models as compared to tumors developed in immunodeficient mice. Using this humanized model, we study the effect of TGFß inhibition on the Ewing TME during radiation therapy, a treatment that both enhances TGFß activation and is used to treat aggressive ES. Utilizing a trivalent ligand TGFß TRAP to inhibit TGFß, we demonstrate that in combination with radiation, TGFß inhibition both increases ES immune cell infiltration and decreases lung metastatic burden in vivo . The culmination of these data demonstrates the value of humanized models to address immunobiologic preclinical questions in Ewing sarcoma and suggests TGFß inhibition as a promising intervention during radiation therapy to promote metastatic tumor control.

16.
Nat Commun ; 15(1): 1231, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38336745

RESUMEN

Androgen deprivation therapy (ADT) targeting androgen/androgen receptor (AR)- signaling pathways is the main therapy for advanced prostate cancer (PCa). However, ADT eventually fails in most patients who consequently develop castration-resistant prostate cancer (CRPC). While more potent AR antagonists and blockers for androgen synthesis were developed to improve clinical outcomes, they also show to induce more diverse CRPC phenotypes. Specifically, the AR- and neuroendocrine-null PCa, DNPC, occurs in abiraterone and enzalutamide-treated patients. Here, we uncover that current ADT induces aberrant HGF/MET signaling activation that further elevates Wnt/ß-catenin signaling in human DNPC samples. Co-activation of HGF/MET and Wnt/ß-catenin axes in mouse prostates induces DNPC-like lesions. Single-cell RNA sequencing analyses identify increased expression and activity of XPO1 and ribosomal proteins in mouse DNPC-like cells. Elevated expression of XPO1 and ribosomal proteins is also identified in clinical DNPC specimens. Inhibition of XPO1 and ribosomal pathways represses DNPC growth in both in vivo and ex vivo conditions, evidencing future therapeutic targets.


Asunto(s)
Andrógenos , Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Ratones , Animales , Andrógenos/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Antagonistas de Andrógenos/farmacología , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular , Vía de Señalización Wnt , Proteínas Ribosómicas/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Factor de Crecimiento de Hepatocito/metabolismo
17.
BMC Genomics ; 14: 895, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24341328

RESUMEN

BACKGROUND: The Streptococcus Anginosus Group (SAG) represents three closely related species of the viridans group streptococci recognized as commensal bacteria of the oral, gastrointestinal and urogenital tracts. The SAG also cause severe invasive infections, and are pathogens during cystic fibrosis (CF) pulmonary exacerbation. Little genomic information or description of virulence mechanisms is currently available for SAG. We conducted intra and inter species whole-genome comparative analyses with 59 publically available Streptococcus genomes and seven in-house closed high quality finished SAG genomes; S. constellatus (3), S. intermedius (2), and S. anginosus (2). For each SAG species, we sequenced at least one numerically dominant strain from CF airways recovered during acute exacerbation and an invasive, non-lung isolate. We also evaluated microevolution that occurred within two isolates that were cultured from one individual one year apart. RESULTS: The SAG genomes were most closely related to S. gordonii and S. sanguinis, based on shared orthologs and harbor a similar number of proteins within each COG category as other Streptococcus species. Numerous characterized streptococcus virulence factor homologs were identified within the SAG genomes including; adherence, invasion, spreading factors, LPxTG cell wall proteins, and two component histidine kinases known to be involved in virulence gene regulation. Mobile elements, primarily integrative conjugative elements and bacteriophage, account for greater than 10% of the SAG genomes. S. anginosus was the most variable species sequenced in this study, yielding both the smallest and the largest SAG genomes containing multiple genomic rearrangements, insertions and deletions. In contrast, within the S. constellatus and S. intermedius species, there was extensive continuous synteny, with only slight differences in genome size between strains. Within S. constellatus we were able to determine important SNPs and changes in VNTR numbers that occurred over the course of one year. CONCLUSIONS: The comparative genomic analysis of the SAG clarifies the phylogenetics of these bacteria and supports the distinct species classification. Numerous potential virulence determinants were identified and provide a foundation for further studies into SAG pathogenesis. Furthermore, the data may be used to enable the development of rapid diagnostic assays and therapeutics for these pathogens.


Asunto(s)
Genoma Bacteriano , Filogenia , Streptococcus anginosus/clasificación , Streptococcus anginosus/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Orden Génico , Transferencia de Gen Horizontal , Genes Bacterianos , Sitios Genéticos , Genómica , Histidina Quinasa , Repeticiones de Minisatélite , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Proteínas Quinasas/genética , Secuencias Repetitivas de Ácidos Nucleicos , Streptococcus anginosus/patogenicidad , Virulencia/genética , Factores de Virulencia/genética
18.
Proc (Bayl Univ Med Cent) ; 36(2): 178-180, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36876260

RESUMEN

At our hospital, direct and video laryngoscopy are used in airway management for cesarean deliveries performed with general anesthesia. We hypothesized that video laryngoscopy would have a higher success rate of endotracheal intubation on the first attempt compared to direct laryngoscopy. We used our electronic medical record system to search for patients who had cesarean deliveries with general anesthesia with endotracheal intubation performed in the operating room from July 1, 2017, through June 30, 2021. Totals of 186 and 176 patients had direct and video laryngoscopy for the first intubation attempts, respectively; 177 (95%) and 163 (93%) patients, respectively, had a successful intubation on the first attempt with each method. The odds ratio of successful intubation on the first attempt for video laryngoscopy was 0.64 (95% CI 0.27, 1.53; P = 0.31) compared to patients who had direct laryngoscopy. There was no statistically significant difference in Cormack-Lehane grade views of the glottis between direct and video laryngoscopy on the first attempt. In conclusion, there was no statistically significant improvement in the success rate of intubation on the first attempt when video laryngoscopy was used for patients undergoing general anesthesia for cesarean delivery.

19.
J Natl Cancer Inst ; 115(11): 1278-1293, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37348864

RESUMEN

Immunotherapy combinations are being investigated to expand the benefit of immune checkpoint blockade across many cancer types. Radiation combinations, in particular using stereotactic body radiotherapy, are of keen interest because of underlying mechanistic rationale, safety, and availability as a standard of care in certain cancers. In addition to direct tumor cytotoxicity, radiation therapy has immunomodulatory effects such as induction of immunogenic cell death, enhancement of antigen presentation, and expansion of the T-cell receptor repertoire as well as recruitment and increased activity of tumor-specific effector CD8+ cells. Combinations of radiation with cytokines and/or chemokines and anti-programmed death 1 and anticytotoxic T-lymphocyte antigen 4 therapies have demonstrated safety and feasibility, as well as the potential to improve long-term outcomes and possibly induce out of irradiated field or abscopal responses. Novel immunoradiotherapy combinations represent a promising therapeutic approach to overcome radioresistance and further enhance systemic immunotherapy. Potential benefits include reversing CD8+ T-cell exhaustion, inhibiting myeloid-derived suppressor cells, and reversing M2 macrophage polarization as well as decreasing levels of colony-stimulating factor-1 and transforming growth factor-ß. Here, we discuss current data and mechanistic rationale for combining novel immunotherapy agents with radiation therapy.


Asunto(s)
Neoplasias , Radioinmunoterapia , Humanos , Terapia Combinada , Neoplasias/radioterapia , Neoplasias/tratamiento farmacológico , Inmunoterapia , Dosis de Radiación
20.
J Clin Invest ; 133(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37183819

RESUMEN

BACKGROUNDWe previously demonstrated the safety of stereotactic body radiotherapy followed by pembrolizumab (SBRT+P) in patients with advanced solid tumors. This phase I clinical trial was expanded to study the safety of partial tumor irradiation (partial-Rx). We assessed irradiated local failure (LF) and clinical outcomes with correlations to biomarkers including CD8+ T cell radiomics score (RS) and circulating cytokines.METHODSPatients received SBRT to 2-4 metastases and pembrolizumab for up to 7 days after SBRT. Tumors measuring up to 65 cc received the full radiation dose (complete-Rx), whereas tumors measuring more than 65 cc received partial-Rx. Landmark analysis was used to assess the relationship between tumor response and overall survival (OS). Multivariable analysis was performed for RS and circulating cytokines.RESULTSIn the combined (expansion plus original) cohort, 97 patients (219 metastases) were analyzed and received SBRT+P. Forty-six (47%) patients received at least 1 partial-Rx treatment. There were 7 (7.2%)dose-limiting toxicities (DLTs). 1-year LF was 7.6% overall, and 13.3% and 5.4% for partial-Rx and complete-Rx tumors, respectively (HR 2.32, 95% CI 0.90-5.97, P = 0.08). The overall, unirradiated, and irradiated objective response rates were 22%, 12%, and 34%, respectively. Irradiated tumor response to SBRT+P was associated with prolonged OS; 1-year OS was 71% (responders), 42% (mixed-responders), and 0% (nonresponders) (P < 0.01). High-RS was significantly associated with improved LF, progression-free survival (PFS), and OS. Elevated circulating IL-8 was independently associated with inferior PFS and OS.CONCLUSIONSBRT+P is safe in patients with large, advanced solid tumors. Additional studies are warranted to assess noninferiority of complete versus partial irradiation of tumors in the setting of immunotherapy.TRIAL REGISTRATIONClinicaltrials.gov NCT02608385FUNDINGMerck Investigator Studies Program; Hillman Fellows for Innovative Cancer Research Program; NIH grants UM1CA186690-06, P50CA254865-01A1, P30CA047904-32, and R01DE031729-01A1.


Asunto(s)
Neoplasias , Radiocirugia , Humanos , Anticuerpos Monoclonales Humanizados/efectos adversos , Citocinas , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Radiocirugia/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA