Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Anat ; 37(1): 147-152, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38057962

RESUMEN

The embryological origin of the trapezius and sternocleidomastoid muscles has been debated for over a century. To shed light on this issue, the present anatomical study was performed. Five fresh frozen human cadavers, three males and two females, were used for this study. Samples from each specimen's trapezius and sternocleidomastoid were fixed in 10% formalin and placed in paraffin blocks. As Paired like homeodomain 2 (Pitx2) and T-box factor 1(Tbx1) have been implicated in the region and muscle type regulation, we performed Tbx1 and Pitx2 Immunohistochemistry (IHC) on these muscle tissue samples to identify the origin of the trapezius and sternocleidomastoid muscles. We have used the latest version of QuPath, v0.4.3, software to quantify the Tbx and Pitx2 staining. For the sternocleidomastoid muscle, for evaluated samples, the average amount of positively stained Tbx1 and Pitx2 was 25% (range 16%-30%) and 18% (range 12%-23%), respectively. For the trapezius muscles, for evaluated samples, the average amount of positively stained Tbx1 and Pitx2 parts of the samples was 17% (range 15%-20%) and 15% (14%-17%), respectively. Our anatomical findings suggest dual origins of both the trapezius and sternocleidomastoid muscles. Additionally, as neither Pitx2 nor Tbx1 made up all the staining observed for each muscle, other contributions to these structures are likely. Future studies with larger samples are now necessary to confirm these findings.


Asunto(s)
Músculos Superficiales de la Espalda , Factores de Transcripción , Masculino , Femenino , Humanos , Factores de Transcripción/fisiología , Músculos del Cuello
2.
Int J Mol Sci ; 25(5)2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38473978

RESUMEN

Female breast cancer accounts for 15.2% of all new cancer cases in the United States, with a continuing increase in incidence despite efforts to discover new targeted therapies. With an approximate failure rate of 85% for therapies in the early phases of clinical trials, there is a need for more translatable, new preclinical in vitro models that include cellular heterogeneity, extracellular matrix, and human-derived biomaterials. Specifically, adipose tissue and its resident cell populations have been identified as necessary attributes for current preclinical models. Adipose-derived stromal/stem cells (ASCs) and mature adipocytes are a normal part of the breast tissue composition and not only contribute to normal breast physiology but also play a significant role in breast cancer pathophysiology. Given the recognized pro-tumorigenic role of adipocytes in tumor progression, there remains a need to enhance the complexity of current models and account for the contribution of the components that exist within the adipose stromal environment to breast tumorigenesis. This review article captures the current landscape of preclinical breast cancer models with a focus on breast cancer microphysiological system (MPS) models and their counterpart patient-derived xenograft (PDX) models to capture patient diversity as they relate to adipose tissue.


Asunto(s)
Neoplasias de la Mama , Animales , Humanos , Femenino , Neoplasias de la Mama/patología , Tejido Adiposo/patología , Adipocitos/patología , Obesidad/patología , Células del Estroma/patología , Modelos Animales de Enfermedad
3.
J Craniofac Surg ; 34(8): 2317-2320, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37665070

RESUMEN

INTRODUCTION: The purpose of the study was to provide an overview of our initial experience utilizing urinary bladder matrix (UBM) for reconstructing avulsed injuries resulting from trauma. MATERIALS AND METHODS: This retrospective case series evaluated patients presented with avulsed soft tissue injuries to the head and neck who underwent reconstruction with UBM. Patients were treated by Oral and Maxillofacial Surgery Service in Louisiana State University Health Sciences Center (Baton Rouge, LA). Descriptive variables were collected. Descriptive statistics were calculated. RESULTS: Eight patients (mean age 55.8 y) met our inclusion criteria. Wounds were located in the scalp (n=2, 25%), mandible (n=2, 25%), upper eyelid (n=1, 12.5%), cheek (n=1, 12.5%), nose (n=1, 12.5%), or neck (n=1, 12.5%). The depth of the wound extended from the skin to the subcutaneous tissue (n=1, 12.5%), muscle (n=2, 25%), bone (n=3, 37.5%), and/or cartilage (n=1, 12.5%). The mean wound diameter was 47.9 cm 2 (range 17-85 cm 2 ). Wounds were classified as acute (n=6, 75%) or chronic wounds (n=2, 25%). At 6 months, all patients had achieved complete healing with no need for additional surgical procedures (n=8, 100%) with a mean healing time of 36.5 days (range 14-90 d). CONCLUSION: Urinary bladder matrix minimize donor-side morbidity, eliminates contraction, and offers a wide range of product sizes to cover a wide range of maxillofacial soft tissue defects in a single-stage manner.


Asunto(s)
Procedimientos de Cirugía Plástica , Traumatismos de los Tejidos Blandos , Humanos , Persona de Mediana Edad , Vejiga Urinaria/cirugía , Estudios Retrospectivos , Traumatismos de los Tejidos Blandos/cirugía , Cicatrización de Heridas , Trasplante de Piel , Resultado del Tratamiento
4.
J Cell Biochem ; 122(8): 835-850, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33876843

RESUMEN

Triple-negative breast cancer (TNBC) presents a clinical challenge due to the aggressive nature of the disease and a lack of targeted therapies. Constitutive activation of the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway has been linked to chemoresistance and metastatic progression through distinct mechanisms, including activation of epithelial-to-mesenchymal transition (EMT) when cells adopt a motile and invasive phenotype through loss of epithelial markers (CDH1), and acquisition of mesenchymal markers (VIM, CDH2). Although MAPK/ERK1/2 kinase inhibitors (MEKi) are useful antitumor agents in a clinical setting, including the Food and Drug Administration (FDA)-approved MEK1,2 dual inhibitors cobimetinib and trametinib, there are limitations to their clinical utility, primarily adaptation of the BRAF pathway and ocular toxicities. The MEK5 (HGNC: MAP2K5) pathway has important roles in metastatic progression of various cancer types, including those of the prostate, colon, bone and breast, and elevated levels of ERK5 expression in breast carcinomas are linked to a worse prognoses in TNBC patients. The purpose of this study is to explore MEK5 regulation of the EMT axis and to evaluate a novel pan-MEK inhibitor on clinically aggressive TNBC cells. Our results show a distinction between the MEK1/2 and MEK5 cascades in maintenance of the mesenchymal phenotype, suggesting that the MEK5 pathway may be necessary and sufficient in EMT regulation while MEK1/2 signaling further sustains the mesenchymal state of TNBC cells. Furthermore, additive effects on MET induction are evident through the inhibition of both MEK1/2 and MEK5. Taken together, these data demonstrate the need for a better understanding of the individual roles of MEK1/2 and MEK5 signaling in breast cancer and provide a rationale for the combined targeting of these pathways to circumvent compensatory signaling and subsequent therapeutic resistance.


Asunto(s)
Movimiento Celular , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , MAP Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Neoplasias de la Mama Triple Negativas/metabolismo , Femenino , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 5/antagonistas & inhibidores , MAP Quinasa Quinasa 5/genética , Células MCF-7 , Proteínas Proto-Oncogénicas c-fos/genética , Neoplasias de la Mama Triple Negativas/genética
5.
Anticancer Drugs ; 32(4): 365-375, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33595947

RESUMEN

Patients with advanced breast cancer often develop bone metastases. Treatment is limited to palliative care. Parathyroid hormone (PTH)/parathyroid hormone-related peptide (PTHrP) antagonists for bone metastases failed clinically due to short half-life and inadequate concentration in bone. We synthesized two novel PTHrP antagonists fused to an inert bacterial collagen binding domain (CBD) that directs drugs to bone. PTH(7-33)-CBD is an N-terminal truncated PTHrP antagonist. [W2]PTH(1-33)-CBD is an PTHrP inverse-agonist. The aim of this study was to assess PTH(7-33)-CBD to reduce breast cancer bone metastases and prevent osteolytic destruction in mice and to assess both drugs for apoptosis of breast cancer cells in vitro and inhibition of PTH receptor (PTHR1). PTH(7-33)-CBD (1000 µg/kg, subcutaneous) or vehicle was administered 24 h prior to MDA-MB-231 breast cancer cell inoculation into the tibia marrow. Weekly tumor burden and bone density were measured. Pharmacokinetic analysis of PTH(7-33)-CBD in rat serum was evaluated. Drug effect on cAMP accumulation in SaOS-2 osteosarcoma cells and apoptosis of MDA-MB-231 cells was assessed. PTH(7-33)-CBD reduced MDA-MB-231 tumor burden and osteolytic destruction in mice 4-5 weeks post-treatment. PTH(7-33)-CBD (1000 µg/kg i.v. and subcutaneous) in rats was rapidly absorbed with peak concentration 5-min and terminal half-life 3-h. Bioavailability by the subcutaneous route was 43% relative to the i.v. route. PTH(7-33)-CBD was detected only on rat periosteal bone surfaces that stained positive for collagen-1. PTH(7-33)-CBD and [W2]PTH(1-33)-CBD (10-8M) blocked basal and PTH agonist-induced cAMP accumulation in SaOS-2 osteosarcoma cells. Both drugs induced PTHR1-dependent apoptosis of MDA-MB-231 cells in vitro. Novel bone-targeted PTHrP antagonists represent a new paradigm for treatment of breast cancer bone metastases.


Asunto(s)
Neoplasias Óseas/prevención & control , Neoplasias Óseas/secundario , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Hormona Paratiroidea/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Línea Celular Tumoral , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Relacionada con la Hormona Paratiroidea/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Prostate ; 80(10): 764-776, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32356608

RESUMEN

BACKGROUND: Aging is the most important risk factor for prostate cancer (PCa), but how age contributes to PCa is poorly understood. Aging is characterized by low-grade systemic inflammation (i.e., inflammaging) that is often attributed to the progressive activation of immune cells over time, which may play an important role in prostate carcinogenesis. Th17 response is elevated in aging humans and mice, but it remains unknown whether it is increased in prostate tissue or contributes to prostate carcinogenesis during aging. In this study, we aimed to determine the role of age-related Th17 response in PCa cell growth, migration, and invasion. METHODS: C57BL/6J (B6) mouse was used as an aging animal model and the prostate histopathology during aging was analyzed. Splenic CD4+ T cells were isolated from young (16-20 weeks old) and aged (96-104 weeks old) mice, and cultured in the presence of plate-bound anti-CD3/anti-CD28, with or without Th17 differentiation conditions. The cells were collected and used for subsequent flow cytometry or quantitative reverse transcription polymerase chain reaction. The supernatant was collected and used to treat PCa cell lines. The treated PCa cells were analyzed for cell viability, migration, invasion, and nuclear factor kappa B (NF-κB) signaling. RESULTS: Aged mice had enlarged prostate glands and increased morphological alterations, with not only increased inflammatory cell infiltration but also increased Th17 cytokines in prostate tissue, compared to young mice. Naïve CD4+ T cells from aged mice differentiated increased interleukin (IL)-17-expressing cells. CD4+ T cells from aged mice spleen had increased Th17 cells, Th17 cytokines and Th17/Treg ratio compared to young mice. Factors secreted from aged CD4+ T cells, especially from ex vivo differentiated Th17 cells, not only promoted PCa cell viability, migration, and invasion but also activated the NF-κB signaling in PCa cells compared to young mice. CONCLUSIONS: These results indicate that age-related CD4+ T cells, especially Th17 cells-secreted factors have the potential to contribute to prostate carcinogenesis. Our work could prompt further research using autochthonous PCa mouse models at different ages to elucidate the functional role of Th17 response in prostate carcinogenesis during aging.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Neoplasias de la Próstata/inmunología , Células Th17/inmunología , Envejecimiento/inmunología , Animales , Linfocitos T CD4-Positivos/patología , Diferenciación Celular/inmunología , Línea Celular Tumoral , Movimiento Celular/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , FN-kappa B/inmunología , Invasividad Neoplásica , Células PC-3 , Neoplasias de la Próstata/patología , Células Th17/patología
7.
J Oral Maxillofac Surg ; 78(9): 1461-1466, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32653307

RESUMEN

The surgeon needs to have an inexpensive, available, nontoxic, and practical disinfectant that is effective in sanitizing against the COVID-19 (Coronavirus Disease 2019) virus. The purpose of this article was to review the evidence for using hypochlorous acid in the office setting on a daily basis. The method used to assemble recommendations was a review of the literature including evidence for this solution when used in different locations and industries other than the oral-maxillofacial clinic facility. The results indicate that this material can be used with a high predictability for disinfecting against the COVID-19 (Coronavirus Disease 2019) virus.


Asunto(s)
Infecciones por Coronavirus/prevención & control , Desinfectantes/química , Ácido Hipocloroso/química , Pandemias/prevención & control , Neumonía Viral/prevención & control , Betacoronavirus , COVID-19 , Consultorios Odontológicos , Humanos , SARS-CoV-2 , Cirugía Bucal
8.
BMC Cancer ; 19(1): 205, 2019 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-30845999

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) represents an aggressive subtype with limited therapeutic options. Experimental preclinical models that recapitulate their tumors of origin can accelerate target identification, thereby potentially improving therapeutic efficacy. Patient-derived xenografts (PDXs), due to their genomic and transcriptomic fidelity to the tumors from which they are derived, are poised to improve the preclinical testing of drug-target combinations in translational models. Despite the previous development of breast and TNBC PDX models, those derived from patients with demonstrated health-disparities are lacking. METHODS: We use an aggressive TNBC PDX model propagated in SCID/Beige mice that was established from an African-American woman, TU-BcX-2 K1, and assess its metastatic potential and drug sensitivities under distinct in vitro conditions. Cellular derivatives of the primary tumor or the PDX were grown in 2D culture conditions or grown in mammospheres 3D culture. Flow cytometry and fluorescence staining was used to quantify cancer stem cell-like populations. qRT-PCR was used to describe the mesenchymal gene signature of the tumor. The sensitivity of TU-BcX-2 K1-derived cells to anti-neoplastic oncology drugs was compared in adherent cells and mammospheres. Drug response was evaluated using a live/dead staining kit and crystal violet staining. RESULTS: TU-BcX-2 K1 has a low propensity for metastasis, reflects a mesenchymal state, and contains a large burden of cancer stem cells. We show that TU-BcX-2 K1 cells have differential responses to cytotoxic and targeted therapies in 2D compared to 3D culture conditions insofar as several drug classes conferred sensitivity in 2D but not in 3D culture, or cells grown as mammospheres. CONCLUSIONS: Here we introduce a new TNBC PDX model and demonstrate the differences in evaluating drug sensitivity in adherent cells compared to mammosphere, or suspension, culture.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Neoplasias de la Mama Triple Negativas/genética , Animales , Biomarcadores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Inmunohistoquímica , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Aesthet Surg J ; 36(1): 93-104, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26063833

RESUMEN

BACKGROUND: Fat grafting has become popular for repair of postsurgical/postradiation defects after head/neck cancers resection. Fat graft supplementation with adipose tissue-derived stromal/stem cells (ASCs) is proposed to improve graft viability/efficacy, although the impact of ASCs on head/neck cancer cells is unknown. OBJECTIVES: To determine whether ASCs affect growth, migration, and metastasis of human head/neck cancer. METHODS: Human Cal-27 and SCC-4 head/neck cancer cells were co-cultured human ASCs, or treated with ASC conditioned medium (CM), and cancer cell growth/migration was assessed by MTT, cell count, and scratch/wound healing assays in vitro. Co-injection of 3 × 10(6) Cal-27/green fluorescent protein (GFP) cells and ASCs into the flank of NUDE mice assessed ASC effect on tumor growth/morphology. Quantitation of human chromosome 17 DNA in mouse organs assessed ASC effects on micrometastasis. Primary tumors were evaluated for markers of epithelial-to-mesenchymal transition, matrix metalloproteinases, and angiogenesis by immunohistochemistry. RESULTS: Co-culture of Cal-27 or SCC-4 cells with ASCs from 2 different donors or ASC CM had no effect on cell growth in vitro. However, ASC CM stimulated Cal-27 and SCC-4 migration. Co-injection of ASCs from 2 different donors with Cal-27 cells did not affect tumor volume at 6 weeks, but increased Cal-27 micrometastasis to the brain. Evaluation of tumors sections from 1 ASC donor co-injection revealed that ASCs were viable and well integrated with Cal-27/GFP cells. These tumors exhibited increased MMP2, MMP9, IL-8, and microvessel density. CONCLUSIONS: Human ASCs did not alter growth of human head/neck cancer cells or tumor xenografts, but stimulated migration and early micrometastasis to mouse brain.


Asunto(s)
Tejido Adiposo/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias de Cabeza y Cuello/patología , Xenoinjertos/metabolismo , Células Madre/metabolismo , Células del Estroma/metabolismo , Tejido Adiposo/citología , Animales , Neoplasias Encefálicas/metabolismo , Carcinoma de Células Escamosas , Proliferación Celular , Células Cultivadas , Femenino , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Ratones , Ratones Desnudos , Trasplante Heterólogo
10.
Genesis ; 52(1): 29-38, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24178943

RESUMEN

The NR4A nuclear receptor NHR-6 is an essential regulator of spermatheca organogenesis in C. elegans. In this study, we perform a focused, RNAi-based screen to identify modifiers of partial nhr-6 loss of function. Ninety-eight genes that encode signaling proteins expressed in the spermatheca were screened for enhancement of the nhr-6 RNAi phenotype. We identify the C. elegans gene jun-1, which encodes the homolog of the Jun transcription factor, as a strong enhancer of nhr-6 partial loss of function. We show that nhr-6 and jun-1 function together to regulate development of the spermatheca and are necessary for generating an organ with the normal number of cells. jun-1 is expressed in all cells of the developing spermatheca. We also provide evidence that NHR-6 and JUN-1 can physically interact in a yeast two-hybrid assay. Our results provide in vivo evidence that NR4A nuclear receptor and Jun transcription factor interactions are essential in regulating developmental processes in metazoans.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Femenino , Genes de Helminto , Gónadas/crecimiento & desarrollo , Gónadas/metabolismo , Organogénesis , Interferencia de ARN , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos
11.
Breast Cancer Res Treat ; 145(3): 593-604, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24810497

RESUMEN

Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype that lacks effective targeted therapies. The epithelial-to-mesenchymal transition (EMT) is a key contributor in the metastatic process. We previously showed the pan-deacetylase inhibitor LBH589 induces CDH1 expression in TNBC cells, suggesting regulation of EMT. The purpose of this study was to examine the effects of LBH589 on the metastatic qualities of TNBC cells and the role of EMT in this process. A panel of breast cancer cell lines (MCF-7, MDA-MB-231, and BT-549), drugged with LBH589, was examined for changes in cell morphology, migration, and invasion in vitro. The effect on in vivo metastasis was examined using immunofluorescent staining of lung sections. EMT gene expression profiling was used to determine LBH589-induced changes in TNBC cells. ZEB overexpression studies were conducted to validate requirement of ZEB in LBH589-mediated proliferation and tumorigenesis. Our results indicate a reversal of EMT by LBH589 as demonstrated by altered morphology and altered gene expression in TNBC. LBH589 was shown to be a more potent inhibitor of EMT than other HDAC inhibitors, SAHA and TMP269. Additionally, we found that LBH589 inhibits metastasis of MDA-MB-231 cells in vivo. These effects of LBH589 were mediated in part by inhibition of ZEB, as overexpression of ZEB1 or ZEB2 mitigated the effects of LBH589 on MDA-MB-231 EMT-associated gene expression, migration, invasion, CDH1 expression, and tumorigenesis. These data indicate therapeutic potential of LBH589 in targeting EMT and metastasis of TNBC.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Proteínas de Homeodominio/antagonistas & inhibidores , Ácidos Hidroxámicos/farmacología , Indoles/farmacología , Proteínas Represoras/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Proteínas de Homeodominio/biosíntesis , Humanos , Células MCF-7 , Ratones , Ratones SCID , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/tratamiento farmacológico , Panobinostat , Proteínas Represoras/biosíntesis , Factores de Transcripción/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
12.
Ann Plast Surg ; 73 Suppl 1: S104-7, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25003407

RESUMEN

Found in most mesenchymally derived organs, mesenchymal stem cells are undifferentiated cells capable of developing into many cell types. Adipose stem cells are a type of mesenchymal stem cell easily extracted from lipoaspirate, often readily available, and are conformable to the tissue defect. Their ability for self-renewal, unlimited proliferation and proangiogenic, and immunomodulatory properties have made them attractive adjuncts in plastic surgery. Since the discovery of pluripotent cells in adipose tissue, plastic surgeons have applied the technology toward improving wound healing, soft tissue augmentation, and tissue engineering. More recently, some surgeons have used adipose stem cells in cancer reconstruction. By mixing lipoaspirate with concentrated fractions of adipose stem cells through a technique termed cell-assisted lipotransfer, plastic surgeons have claimed improved aesthetic results. Promising early results have been tempered by in vitro and animal studies demonstrating increased tumor proliferation and metastasis rates with the use of adipose and other mesenchymal stem cells. This review provides a succinct yet comprehensive overview of the current literature evaluating the oncologic risks associated with adipose stem cell use in cancer.


Asunto(s)
Tejido Adiposo/citología , Neoplasias/cirugía , Trasplante de Células Madre , Humanos , Trasplante de Células Madre Mesenquimatosas , Procedimientos de Cirugía Plástica/métodos
13.
J Natl Cancer Inst ; 116(10): 1598-1611, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38833676

RESUMEN

BACKGROUND: The role of Th17 cells in prostate cancer is not fully understood. The transcription factor BATF controls the differentiation of Th17 cells. Mice deficient in Batf do not produce Th17 cells. METHODS: In this study, we aimed to characterize the role of Batf-dependent Th17 cells in prostate cancer by crossbreeding Batf knockout mice with mice conditionally mutant for Pten. RESULTS: We found that Batf knockout mice had changes in the morphology of prostate epithelial cells compared with normal mice, and Batf knockout mice deficient in Pten (called Batf-) had smaller prostate size and developed fewer invasive prostate adenocarcinomas than Pten-deficient mice with Batf expression (called Batf+). The prostate tumors in Batf- mice showed reduced proliferation, increased apoptosis, decreased angiogenesis and inflammatory cell infiltration, and activation of nuclear factor-κB signaling. Moreover, Batf- mice showed significantly reduced interleukin 23 (IL-23)-IL-23R signaling. In the prostate stroma of Batf- mice, IL-23R-positive cells were decreased considerably compared with Batf+ mice. Splenocytes and prostate tissues from Batf- mice cultured under Th17 differentiation conditions expressed reduced IL-23/IL-23R than cultured cells from Batf+ mice. Anti-IL-23p19 antibody treatment of Pten-deficient mice reduced prostate tumors and angiogenesis compared with control immunoglobulin G-treated mice. In human prostate tumors, BATF messenger RNA level was positively correlated with IL-23A and IL-23R but not RORC. CONCLUSION: Our novel findings underscore the crucial role of IL-23-IL-23R signaling in mediating the function of Batf-dependent Th17 cells, thereby promoting prostate cancer initiation and progression. This finding highlights the BATF-IL-23R axis as a promising target for the development of innovative strategies for prostate cancer prevention and treatment.


Asunto(s)
Adenocarcinoma , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Progresión de la Enfermedad , Ratones Noqueados , Fosfohidrolasa PTEN , Neoplasias de la Próstata , Receptores de Interleucina , Transducción de Señal , Células Th17 , Animales , Masculino , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/genética , Células Th17/inmunología , Células Th17/metabolismo , Ratones , Fosfohidrolasa PTEN/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/inmunología , Adenocarcinoma/metabolismo , Adenocarcinoma/genética , Receptores de Interleucina/metabolismo , Interleucina-23/metabolismo , Humanos , Apoptosis , Proliferación Celular , FN-kappa B/metabolismo
14.
BMC Cell Biol ; 14: 34, 2013 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-23924189

RESUMEN

BACKGROUND: Obesity is associated with a higher risk of developing cancer and co-morbidities that are part of the metabolic syndrome. Adipose tissue is recognized as an endocrine organ, as it affects a number of physiological functions, and contains adipose tissue-derived stem cells (ASCs). ASCs can differentiate into cells of multiple lineages, and as such are applicable to tissue engineering and regenerative medicine. Yet the question of whether ASC functionality is affected by the donor's body mass index (BMI) still exists. RESULTS: ASCs were isolated from patients having different BMIs (BMI-ASCs), within the ranges of 18.5-32.8. It was hypothesized that overweight BMI-ASCs would be more compromised in early adipogenic and osteogenic potential, and ability to form colonies in vitro. BMI was inversely correlated with ASC proliferation and colony forming potential as assessed by CyQUANT proliferation assay (fluorescence- based measurement of cellular DNA content), and colony forming assays. BMI was positively correlated with early time point (day 7) but not later time point (day 15) intracytoplasmic lipid accumulation as assessed by Oil-Red-O staining. Alizarin red staining and RT-PCR for alkaline phosphatase demonstrated that elevated BMI resulted in compromised ASC mineralization of extracellular matrix and decreased alkaline phosphatase mRNA expression. CONCLUSIONS: These data demonstrate that elevated BMI resulted in reduced ASC proliferation, and potentially compromised osteogenic capacity in vitro; thus BMI is an important criterion to consider in selecting ASC donors for clinical applications.


Asunto(s)
Células Madre Adultas/citología , Índice de Masa Corporal , Diferenciación Celular/fisiología , Proliferación Celular , Osteogénesis/fisiología , Grasa Subcutánea/citología , Adulto , Células Madre Adultas/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Medios de Cultivo/farmacología , Femenino , Humanos , Técnicas In Vitro , Persona de Mediana Edad , Obesidad/patología , Osteogénesis/efectos de los fármacos , Sobrepeso/patología , Grasa Subcutánea/efectos de los fármacos
15.
BMC Cancer ; 13: 139, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23517088

RESUMEN

BACKGROUND: NR4A orphan nuclear receptors are involved in multiple biological processes which are important in tumorigenesis such as cell proliferation, apoptosis, differentiation, and glucose utilization. The significance of NR4A family member NURR1 (NR4A2) in breast cancer etiology has not been elucidated. The purpose of this study was to ascertain the impact of NURR1 expression on breast transformation, tumor growth, and breast cancer patient survival. METHODS: We determined the expression of NURR1 in normal breast versus breast carcinoma in tissue microarrays (immunohistochemistry), tissue lysates (immunoblot), and at the mRNA level (publically available breast microarrays). In addition NURR1 expression was compared among breast cancer patients in cohorts based on p53 expression, estrogen receptor α expression, tumor grade, and lymph node metastases. Kaplan-Meier survival plots were used to determine the correlation between NURR1 expression and relapse free survival (RFS). Using shRNA-mediated silencing, we determined the effect of NURR1 expression on tumor growth in mouse xenografts. RESULTS: Results from breast cancer tissue arrays demonstrate a higher NURR1 expression in the normal breast epithelium compared to breast carcinoma cells (p ≤ 0.05). Among cases of breast cancer, NURR1 expression in the primary tumors was inversely correlated with lymph node metastases (p ≤ 0.05) and p53 expression (p ≤ 0.05). Clinical stage and histological grade were not associated with variation in NURR1 expression. In gene microarrays, 4 of 5 datasets showed stronger mean expression of NURR1 in normal breast as compared to transformed breast. Additionally, NURR1 expression was strongly correlated with increase relapse free survival (HR = 0.7) in a cohort of all breast cancer patients, but showed no significant difference in survival when compared among patients whom have not been treated systemically (HR = 0.91). Paradoxically, NURR1 silenced breast xenografts showed significantly decreased growth in comparison to control, underscoring a biphasic role for NURR1 in breast cancer progression. CONCLUSIONS: NURR1 function presents a dichotomy in breast cancer etiology, in which NURR1 expression is associated with normal breast epithelial differentiation and efficacy of systemic cancer therapy, but silencing of which attenuates tumor growth. This provides a strong rationale for the potential implementation of NURR1 as a pharmacologic target and biomarker for therapeutic efficacy in breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Animales , Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Minería de Datos , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Ratones , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Pronóstico , Trasplante Heterólogo , Carga Tumoral/genética
16.
J Pineal Res ; 55(4): 377-87, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24033914

RESUMEN

Obesity is a chronic inflammation with increased serum levels of insulin, insulin-like growth factor 1 (IGF1), and interleukin-17 (IL-17). The objective of this study was to test a hypothesis that insulin and IGF1 enhance IL-17-induced expression of inflammatory chemokines/cytokines through a glycogen synthase kinase 3ß (GSK3B)-dependent mechanism, which can be inhibited by melatonin. We found that insulin/IGF1 and lithium chloride enhanced IL-17-induced expression of C-X-C motif ligand 1 (Cxcl1) and C-C motif ligand 20 (Ccl20) in the Gsk3b(+/+) , but not in Gsk3b(-/-) mouse embryonic fibroblast (MEF) cells. IL-17 induced higher levels of Cxcl1 and Ccl20 in the Gsk3b(-/-) MEF cells, compared with the Gsk3b(+/+) MEF cells. Insulin and IGF1 activated Akt to phosphorylate GSK3B at serine 9, thus inhibiting GSK3B activity. Melatonin inhibited Akt activation, thus decreasing P-GSK3B at serine 9 (i.e., increasing GSK3B activity) and subsequently inhibiting expression of Cxcl1 and Ccl20 that was induced either by IL-17 alone or by a combination of insulin and IL-17. Melatonin's inhibitory effects were only observed in the Gsk3b(+/+) , but in not Gsk3b(-/-) MEF cells. Melatonin also inhibited expression of Cxcl1, Ccl20, and Il-6 that was induced by a combination of insulin and IL-17 in the mouse prostatic tissues. Further, nighttime human blood, which contained high physiologic levels of melatonin, decreased expression of Cxcl1, Ccl20, and Il-6 in the PC3 human prostate cancer xenograft tumors. Our data support our hypothesis and suggest that melatonin may be used to dampen IL-17-mediated inflammation that is enhanced by the increased levels of insulin and IGF1 in obesity.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Insulina/farmacología , Interleucina-17/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Cloruro de Litio/farmacología , Melatonina/metabolismo , Melatonina/farmacología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
17.
Breast Cancer Res Treat ; 132(2): 391-409, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21509526

RESUMEN

KX-01 is the first clinical Src inhibitor of the novel peptidomimetic class that targets the peptide substrate site of Src providing more specificity toward Src kinase. The present study was designed to evaluate the effects of KX-01 as a single agent and in combination with tamoxifen (TAM) on cell growth and apoptosis of ERα positive breast cancer in vitro and in vivo. Flow cytometry demonstrated that KX-01 induced cell cycle arrest in G2/M phase. Immunofluorescent staining for mitotic phase markers and TUNEL staining indicated that cells had arrested in the mitotic phase and mitotic arrested cells were undergoing apoptosis. KX-01 induced nuclear accumulation of cyclin B1, and activation of CDK1, MPM2, and Cdc25C that is required for progression past the G2/M checkpoint. Apoptosis resulted from activation of caspases 6, 7, 8, and 9. Combinational index analysis revealed that combinations of KX-01 with TAM resulted in synergistic growth inhibition of breast cancer cell lines. KX-01 combined with TAM resulted in decreased ERα phosphorylation at Src-regulated phosphorylation sites serines 118 and 167 that were associated with reduced ERα transcriptional activity. Orally administered KX-01 resulted in a dose dependent growth inhibition of MCF-7 tumor xenografts, and in combination with TAM exhibited synergistic growth inhibition. Immunohistochemical analysis revealed that combinational treatment reduced angiogenesis, and ERα signaling in tumors compared to either drug alone that may underlie the synergistic tumor growth inhibition. Combinations of KX-01 with endocrine therapy present a promising new strategy for clinical management of ERα positive breast cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Receptor alfa de Estrógeno/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Dominio Catalítico , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Moduladores de los Receptores de Estrógeno/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Etiquetado Corte-Fin in Situ , Ratones , Ratones Desnudos , Mitosis/efectos de los fármacos , Neovascularización Patológica/enzimología , Neovascularización Patológica/prevención & control , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Tamoxifeno/farmacología , Factores de Tiempo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/metabolismo
18.
BMC Cancer ; 12: 10, 2012 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22233382

RESUMEN

BACKGROUND: The study of breast cancer metastasis depends on the use of established breast cancer cell lines that do not accurately represent the heterogeneity and complexity of human breast tumors. A tumor model was developed using primary breast tumor-initiating cells isolated from patient core biopsies that would more accurately reflect human breast cancer metastasis. METHODS: Tumorspheres were isolated under serum-free culture conditions from core biopsies collected from five patients with clinical diagnosis of invasive ductal carcinoma (IDC). Isolated tumorspheres were transplanted into the mammary fat pad of NUDE mice to establish tumorigenicity in vivo. Tumors and metastatic lesions were analyzed by hematoxylin and eosin (H+E) staining and immunohistochemistry (IHC). RESULTS: Tumorspheres were successfully isolated from all patient core biopsies, independent of the estrogen receptor α (ERα)/progesterone receptor (PR)/Her2/neu status or tumor grade. Each tumorsphere was estimated to contain 50-100 cells. Transplantation of 50 tumorspheres (1-5 × 103 cells) in combination with Matrigel into the mammary fat pad of NUDE mice resulted in small, palpable tumors that were sustained up to 12 months post-injection. Tumors were serially transplanted three times by re-isolation of tumorspheres from the tumors and injection into the mammary fat pad of NUDE mice. At 3 months post-injection, micrometastases to the lung, liver, kidneys, brain and femur were detected by measuring content of human chromosome 17. Visible macrometastases were detected in the lung, liver and kidneys by 6 months post-injection. Primary tumors variably expressed cytokeratins, Her2/neu, cytoplasmic E-cadherin, nuclear ß catenin and fibronectin but were negative for ERα and vimentin. In lung and liver metastases, variable redistribution of E-cadherin and ß catenin to the membrane of tumor cells was observed. ERα was re-expressed in lung metastatic cells in two of five samples. CONCLUSIONS: Tumorspheres isolated under defined culture conditions from patient core biopsies were tumorigenic when transplanted into the mammary fat pad of NUDE mice, and metastasized to multiple mouse organs. Micrometastases in mouse organs demonstrated a dormancy period prior to outgrowth of macrometastases. The development of macrometastases with organ-specific phenotypic distinctions provides a superior model for the investigation of organ-specific effects on metastatic cancer cell survival and growth.


Asunto(s)
Mama/patología , Carcinoma Ductal de Mama/secundario , Neoplasias Mamarias Experimentales/patología , Animales , Biomarcadores de Tumor/metabolismo , Biopsia , Carcinoma Ductal de Mama/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Desnudos
19.
Carcinogenesis ; 32(7): 964-72, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21317300

RESUMEN

Recent studies have implicated multipotential mesenchymal stem cells (MSCs) as an aid to breast cancer cell proliferation and metastasis, partly as a result of the MSCs secretome. As the tumor gets beyond 2 mm in diameter, the stromal cells could undergo starvation due to the lack of sufficient nutrients in solid tumor microenvironment. In this study, we investigated the survival mechanisms used by stressed stromal cells in breast cancers. We used serum-deprived mesenchymal stem cells (SD-MSCs) and MCF-7 breast cancer cells as model system with a hypothesis that stromal cells in the nutrient-deprived core utilize survival mechanisms for supporting surrounding cells. We tested this hypothesis using in vivo tumor xenografts in immunodeficient mice, which indicated that SD-MSCs supported MCF-7 tumor growth by protection from apoptosis. Histochemical assays showed that SD-MSCs-injected tumors exhibited higher cellularity, decreased apoptosis and decreased differentiation. Beclin-1 staining indicated autophagic areas surrounded by actively proliferating cells. Furthermore, in vitro studies demonstrate that SD-MSCs survive using autophagy and secrete paracrine factors that support tumor cells following nutrient/serum deprivation. Western blot and immunocytochemistry analysis of SD-MSCs demonstrated upregulation and perinuclear relocation of autophagy key regulators such as beclin-1, ATG10, ATG12, MAP-LC3 and lysosomes. Electron microscopic analysis detected a time-dependent increase in autophagosome formation and HDAC6 activity assays indicated the upregulation of autophagy. Taken together, these data suggest that under nutrient-deprived conditions that can occur in solid tumors, stromal cells utilize autophagy for survival and also secrete anti-apoptotic factors that can facilitate solid tumor survival and growth.


Asunto(s)
Autofagia , Células Madre Mesenquimatosas/inmunología , Neoplasias/patología , Células del Estroma/patología , Animales , Apoptosis/inmunología , Línea Celular Tumoral , Supervivencia Celular/inmunología , Células Cultivadas , Medio de Cultivo Libre de Suero , Femenino , Humanos , Ratones , Ratones SCID , Neoplasias/inmunología
20.
Front Mol Biosci ; 8: 696537, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34150854

RESUMEN

Prostate cancer (PCa) is associated with advanced age, but how age contributes to prostate carcinogenesis remains unknown. The prostate-specific Pten conditional knockout mouse model closely imitates human PCa initiation and progression. To better understand how age impacts PCa in an experimental model, we have generated a spatially and temporally controlled Pten-null PCa murine model at different ages (aged vs. non-aged) of adult mice. Here, we present a protocol to inject the Cre-expressing adenovirus with luciferin tag, intraductally, into the prostate anterior lobes of Pten-floxed mice; Pten-loss will be triggered post-Cre expression at different ages. In vivo imaging of luciferin signal following viral infection confirmed successful delivery of the virus and Cre activity. Immunohistochemical staining confirmed prostate epithelial-specific expression of Cre recombinase and the loss of Pten and activation of P-Akt, P-S6, and P-4E-BP1. The Cre-expression, Pten ablation, and activated PI3K/AKT/mTOR pathways were limited to the prostate epithelium. All mice developed prostatic epithelial hyperplasia within 4 weeks after Pten ablation and prostatic intraepithelial neoplasia (PIN) within 8 weeks post-Pten ablation. Some PINs had progressed to invasive adenocarcinoma at 8-16 weeks post-Pten ablation. Aged mice exhibited significantly accelerated PI3K/AKT/mTOR signaling and increased PCa onset and progression compared to young mice. The viral infection success rate is ∼80%. This model will be beneficial for investigations of cancer-related to aging.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA