Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Blood ; 123(17): 2691-702, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24599548

RESUMEN

Leukemia cells are protected from chemotherapy-induced apoptosis by their interactions with bone marrow mesenchymal stromal cells (BM-MSCs). Yet the underlying mechanisms associated with this protective effect remain unclear. Genome-wide gene expression profiling of BM-MSCs revealed that coculture with leukemia cells upregulated the transcription of genes associated with nuclear factor (NF)-κB signaling. Moreover, primary BM-MSCs from leukemia patients expressed NF-κB target genes at higher levels than their normal BM-MSC counterparts. The blockade of NF-κB activation via chemical agents or the overexpression of the mutant form of inhibitor κB-α (IκBα) in BM-MSCs markedly reduced the stromal-mediated drug resistance in leukemia cells in vitro and in vivo. In particular, our unique in vivo model of human leukemia BM microenvironment illustrated a direct link between NF-κB activation and stromal-associated chemoprotection. Mechanistic in vitro studies revealed that the interaction between vascular cell adhesion molecule 1 (VCAM-1) and very late antigen-4 (VLA-4) played an integral role in the activation of NF-κB in the stromal and tumor cell compartments. Together, these results suggest that reciprocal NF-κB activation in BM-MSCs and leukemia cells is essential for promoting chemoresistance in the transformed cells, and targeting NF-κB or VLA-4/VCAM-1 signaling could be a clinically relevant mechanism to overcome stroma-mediated chemoresistance in BM-resident leukemia cells.


Asunto(s)
Resistencia a Antineoplásicos , Regulación Leucémica de la Expresión Génica , Integrina alfa4beta1/metabolismo , FN-kappa B/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Huesos/metabolismo , Adhesión Celular , Línea Celular Transformada , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/citología , Perfilación de la Expresión Génica , Humanos , Ratones , ARN Mensajero/metabolismo , Transducción de Señal , Células del Estroma/citología
2.
Cytotherapy ; 15(1): 20-32, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23260083

RESUMEN

BACKGROUND AIMS: Many ovarian cancers originate from ovarian surface epithelium, where they develop from cysts intermixed with stroma. The stromal layer is critical to the progression and survival of the neoplasm and consequently is recruited into the tumor microenvironment. METHODS: Using both syngeneic mouse tumors (ID8-R) and human xenograft (OVCAR3, SKOV3) tumor models, we first confirmed that intraperitoneally injected circulating mesenchymal stem cells (MSCs) could target, preferentially engraft and differentiate into α-smooth muscle actin-positive myofibroblasts, suggesting their role as "reactive stroma" in ovarian carcinoma development and confirming their potential as a targeted delivery vehicle for the intratumoral production of interferon-ß (IFN-ß). Mice with ovarian carcinomas then received weekly intraperitoneal injections of IFN-ß expressing MSCs. RESULTS: Intraperitoneal injections of IFN-ß expressing MSCs resulted in complete eradication of tumors in 70% of treated OVCAR3 mice (P = 0.004) and an increased survival of treated SKOV3 mice compared with controls (P = 0.01). Similar tumor growth control was observed using murine IFN-ß delivered by murine MSCs in ID8-R ovarian carcinoma. As a potential mechanism of tumor killing, MSCs produced IFN-ß-induced caspase-dependent tumor cell apoptosis. CONCLUSIONS: Our results demonstrate that ovarian carcinoma engrafts MSCs to participate in myofibrovascular networks and that IFN-ß produced by MSCs intratumorally modulates tumor kinetics, resulting in prolonged survival.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Neoplasias Ováricas/terapia , Animales , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Femenino , Terapia Genética/métodos , Humanos , Inmunohistoquímica , Interferón beta/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Prev Res (Phila) ; 16(5): 281-291, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36862830

RESUMEN

PREVENTION RELEVANCE: In this prospective population-based cohort study, we show the improved performance of a new risk assessment model compared with a gold-standard model (BCRAT). The classification of at-risk women using this new model highlights the opportunity to improve risk stratification and implement existing clinical risk-reduction interventions.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Estudios de Cohortes , Estudios Prospectivos , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/prevención & control , Medición de Riesgo , Factores de Riesgo
4.
Cytotherapy ; 12(5): 615-25, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20230221

RESUMEN

BACKGROUND AIMS: Because of the inflammatory nature and extensive stromal compartment in pancreatic tumors, we investigated the role of mesenchymal stromal cells (MSC) to engraft selectively in pancreatic carcinomas and serve as anti-tumor drug delivery vehicles to control pancreatic cancer progression. METHODS: Human pancreatic carcinoma cells, PANC-1, expressing renilla luciferase were orthotopically implanted into SCID mice and allowed to develop for 10 days. Firefly luciferase-transduced MSC or MSC expressing interferon (IFN)-beta were then injected intraperitoneally weekly for 3 weeks. Mice were monitored by bioluminescent imaging for expression of renilla (PANC-1) and firefly (MSC) luciferase. RESULTS: MSC selectively homed to sites of primary and metastatic pancreatic tumors and inhibited tumor growth (P=0.032). The production of IFN-beta within the tumor site by MSC-IFN-beta further suppressed tumor growth (P=0.0000083). Prior studies indicated that MSC home to sites of inflammation; therefore, we sought to alter the tumor microenvironment through treatment with a potent anti-inflammatory agent. After treatment, inflammation-associated mediators were effectively down-regulated, including NFkappaB, vascular endothelial growth factor (VEGF) and interleukin (IL)-6 as well as chemokines involved in MSC migration (CCL3 and CCL25). Treatment with the anti-inflammatory agent CDDO-Me before and after MSC-IFN-beta injections resulted in reduction of MSC in the tumors and reversed the positive effect of tumor inhibition by MSC-IFN-beta alone (P=0.041). CONCLUSIONS: These results suggest that MSC exhibit innate anti-tumor effects against PANC-1 cells and can serve as delivery vehicles for IFN-beta for the treatment of pancreatic cancer. However, these beneficial effects may be lost in therapies combining MSC with anti-inflammatory agents.


Asunto(s)
Interferón beta/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Neoplasias Pancreáticas/terapia , Células del Estroma/metabolismo , Animales , Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Procesos de Crecimiento Celular/inmunología , Línea Celular Tumoral , Terapia Genética , Inhibidores de Crecimiento/inmunología , Inhibidores de Crecimiento/uso terapéutico , Humanos , Terapia de Inmunosupresión , Inflamación , Interferón beta/genética , Interferón beta/inmunología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/patología , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Células del Estroma/patología , Células del Estroma/trasplante , Transgenes/genética
5.
Cancer Res ; 78(12): 3233-3242, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29661830

RESUMEN

Mounting clinical and preclinical evidence supports a key role for sustained adrenergic signaling in the tumor microenvironment as a driver of tumor growth and progression. However, the mechanisms by which adrenergic neurotransmitters are delivered to the tumor microenvironment are not well understood. Here we present evidence for a feed-forward loop whereby adrenergic signaling leads to increased tumoral innervation. In response to catecholamines, tumor cells produced brain-derived neurotrophic factor (BDNF) in an ADRB3/cAMP/Epac/JNK-dependent manner. Elevated BDNF levels in the tumor microenvironment increased innervation by signaling through host neurotrophic receptor tyrosine kinase 2 receptors. In patients with cancer, high tumor nerve counts were significantly associated with increased BDNF and norepinephrine levels and decreased overall survival. Collectively, these data describe a novel pathway for tumor innervation, with resultant biological and clinical implications.Significance: Sustained adrenergic signaling promotes tumor growth and metastasis through BDNF-mediated tumoral innervation. Cancer Res; 78(12); 3233-42. ©2018 AACR.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Retroalimentación Fisiológica , Neoplasias/patología , Norepinefrina/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Animales , Línea Celular Tumoral , AMP Cíclico/metabolismo , Femenino , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Neoplasias/mortalidad , Nervios Periféricos/metabolismo , Nervios Periféricos/patología , Receptor trkB/metabolismo , Transducción de Señal , Microambiente Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Exp Med ; 213(9): 1723-40, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27481130

RESUMEN

Primary myelofibrosis (PMF) is a fatal neoplastic disease characterized by clonal myeloproliferation and progressive bone marrow (BM) fibrosis thought to be induced by mesenchymal stromal cells stimulated by overproduced growth factors. However, tissue fibrosis in other diseases is associated with monocyte-derived fibrocytes. Therefore, we sought to determine whether fibrocytes play a role in the induction of BM fibrosis in PMF. In this study, we show that BM from patients with PMF harbors an abundance of clonal, neoplastic collagen- and fibronectin-producing fibrocytes. Immunodeficient mice transplanted with myelofibrosis patients' BM cells developed a lethal myelofibrosis-like phenotype. Treatment of the xenograft mice with the fibrocyte inhibitor serum amyloid P (SAP; pentraxin-2) significantly prolonged survival and slowed the development of BM fibrosis. Collectively, our data suggest that neoplastic fibrocytes contribute to the induction of BM fibrosis in PMF, and inhibiting fibrocyte differentiation with SAP may interfere with this process.


Asunto(s)
Fibroblastos/fisiología , Monocitos/citología , Mielofibrosis Primaria/etiología , Animales , Médula Ósea/patología , Trasplante de Médula Ósea , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibrosis , Proteínas de Homeodominio/farmacología , Humanos , Ratones , Ratones SCID , Nitrilos , Mielofibrosis Primaria/patología , Pirazoles/farmacología , Pirimidinas , Proteínas Recombinantes/farmacología , Componente Amiloide P Sérico/farmacología
7.
J Vis Exp ; (79): e50385, 2013 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-24084599

RESUMEN

With the desire to understand the contributions of multiple cellular elements to the development of a complex tissue; such as the numerous cell types that participate in regenerating tissue, tumor formation, or vasculogenesis, we devised a multi-colored cellular transplant model of tumor development in which cell populations originate from different fluorescently colored reporter gene mice and are transplanted, engrafted or injected in and around a developing tumor. These colored cells are then recruited and incorporated into the tumor stroma. In order to quantitatively assess bone marrow derived tumor stromal cells, we transplanted GFP expressing transgenic whole bone marrow into lethally irradiated RFP expressing mice as approved by IACUC. 0ovarian tumors that were orthotopically injected into the transplanted mice were excised 6-8 weeks post engraftment and analyzed for bone marrow marker of origin (GFP) as well as antibody markers to detect tumor associated stroma using multispectral imaging techniques. We then adapted a methodology we call MIMicc- Multispectral Interrogation of Multiplexed cellular compositions, using multispectral unmixing of fluoroprobes to quantitatively assess which labeled cell came from which starting populations (based on original reporter gene labels), and as our ability to unmix 4, 5, 6 or more spectra per slide increases, we've added additional immunohistochemistry associated with cell lineages or differentiation to increase precision. Utilizing software to detect co-localized multiplexed-fluorescent signals, tumor stromal populations can be traced, enumerated and characterized based on marker staining.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Médula Ósea/métodos , Citometría de Flujo/métodos , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Microscopía Fluorescente/métodos , Neoplasias Ováricas/patología , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Femenino , Técnica del Anticuerpo Fluorescente/métodos , Colorantes Fluorescentes/química , Proteínas Fluorescentes Verdes/análisis , Indoles/química , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Proteína Fluorescente Roja
8.
Cancer Res ; 73(17): 5347-59, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23838935

RESUMEN

Tumor-stroma interactions play a crucial role in cancer progression by eliciting factors that promote proliferative, angiogenic, and invasive supports to the tumor microenvironment. Mesenchymal stromal/stem cells (MSC) contribute to stroma in part as cancer-associated fibroblasts (CAF), but a complete understanding of how MSC contribute to the tumor stroma is lacking. In this study, we show how CAF phenotypes rely upon MSC expression of the multifunctional cell surface glycoprotein CD44, a putative stem cell marker. Through bone marrow transplantation experiments in a transgenic mouse model of cancer, we determined that CD44 deficiency leads to a relative reduction in the contribution of bone marrow-derived cells to tumor stroma. CD44 attenuation in MSC limited their expression of CAF markers induced by tumor conditioning, and these MSC migrated poorly and provided weak angiogenic support compared with wild-type MSC. These defects were linked to deficiencies in the ability of CD44-attenuated MSC to transcriptionally upregulate Twist expression. Together, our results establish that CD44 expression contributes to critical functions in the tumor stroma.


Asunto(s)
Fibroblastos/patología , Receptores de Hialuranos/fisiología , Neoplasias Mamarias Animales/patología , Células Madre Mesenquimatosas/patología , Neoplasias Ováricas/patología , Microambiente Tumoral , Proteína 1 Relacionada con Twist/metabolismo , Animales , Apoptosis , Western Blotting , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Inmunoprecipitación de Cromatina , Medios de Cultivo Condicionados/farmacología , Femenino , Fibroblastos/metabolismo , Humanos , Técnicas para Inmunoenzimas , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Fenotipo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 Relacionada con Twist/genética
9.
Methods Mol Biol ; 904: 173-90, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22890932

RESUMEN

The act of migration is similar for many cell types. The migratory mechanism of mesenchymal stem cells (MSC) is not completely elucidated, yet many of the initial studies have been based on current understanding of the leukocyte migration. A normal function of MSC is the ability of the cell to migrate to and repair wounded tissue. This wound healing property of MSC originates with migration towards inflammatory signals produced by the wounded environment [1]. A tumor and its microenvironment are capable of eliciting a similar inflammatory response from the MSC, thus resulting in migration of the MSC towards the tumor microenvironment. We have shown MSC migration both in vitro and in vivo. In this chapter, we elucidate several in vivo methods to study MSC migration and mobilization to the tumor microenvironment. The first model is an exogenous model of MSC migration that can be performed in both xenograft and syngenic systems with in vitro expanded MSC. The second model utilizes transgenic-fluorescent--colored mice to follow endogenous bone marrow components including MSC. The third technique enables us to analyze data from the transgenic model through multispectral imaging. Furthermore, the migratory phenotype of MSC can be exploited for use in tumor-targeted gene delivery therapy has been efficacious in animal model studies and is an anticipated therapeutic device in clinical trials.


Asunto(s)
Movimiento Celular/fisiología , Rastreo Celular/métodos , Inflamación/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Adipocitos/citología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Línea Celular , Separación Celular/métodos , Condrocitos/citología , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones , Osteocitos/citología , Microambiente Tumoral
10.
J Clin Invest ; 122(6): 2066-78, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22585577

RESUMEN

Cancer stem cells (CSCs) are a small subpopulation of cancer cells that have increased resistance to conventional therapies and are capable of establishing metastasis. However, only a few biomarkers of CSCs have been identified. Here, we report that ganglioside GD2 (a glycosphingolipid) identifies a small fraction of cells in human breast cancer cell lines and patient samples that are capable of forming mammospheres and initiating tumors with as few as 10 GD2+ cells. In addition, the majority of GD2+ cells are also CD44hiCD24lo, the previously established CSC-associated cell surface phenotype. Gene expression analysis revealed that GD3 synthase (GD3S) is highly expressed in GD2+ as well as in CD44hiCD24lo cells and that interference with GD3S expression, either by shRNA or using a pharmacological inhibitor, reduced the CSC population and CSC-associated properties. GD3S knockdown completely abrogated tumor formation in vivo. Also, induction of epithelial-mesenchymal transition (EMT) in transformed human mammary epithelial cells (HMLER cells) dramatically increased GD2 as well as GD3S expression in these cells, suggesting a role of EMT in the origin of GD2+ breast CSCs. In summary, we identified GD2 as a new CSC-specific cell surface marker and GD3S as a potential therapeutic target for CSCs, with the possibility of improving survival and cure rates in patients with breast cancer.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Neoplasias de la Mama/metabolismo , Gangliósidos/biosíntesis , Células Madre Neoplásicas/metabolismo , Animales , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Antígeno CD24/genética , Antígeno CD24/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Gangliósidos/genética , Regulación Enzimológica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/trasplante , Sialiltransferasas/biosíntesis , Sialiltransferasas/genética , Trasplante Heterólogo
11.
Methods Mol Biol ; 750: 241-59, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21618096

RESUMEN

Targeted migration is a necessary attribute for any gene delivery vehicle. Mesenchymal stem cells (MSC) have been used as effective delivery vehicles for treatments against cancer, graft versus host disease, -arthritis, multiple sclerosis, and many other diseases. MSC migrate toward sites of inflammation, however, the true migratory mechanism has yet to be elucidated. There are several receptors and respective chemokines known to be involved in the migration of the MSC. Further insight to MSC migration will be revealed both in vivo and in vitro through the application of migration assays from the most simple, to the more technologically demanding.


Asunto(s)
Adipocitos/citología , Diferenciación Celular/fisiología , Ensayos de Migración Celular , Movimiento Celular , Células Madre Mesenquimatosas/citología , Osteoblastos/citología , Adipocitos/fisiología , Animales , Línea Celular Tumoral , Separación Celular , Técnicas de Transferencia de Gen , Humanos , Inmunohistoquímica , Mediciones Luminiscentes , Proteínas Luminiscentes/análisis , Células Madre Mesenquimatosas/fisiología , Ratones , Neoplasias/patología , Osteoblastos/fisiología
12.
PLoS One ; 4(4): e4992, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19352430

RESUMEN

BACKGROUND: Tumor associated fibroblasts (TAF), are essential for tumor progression providing both a functional and structural supportive environment. TAF, known as activated fibroblasts, have an established biological impact on tumorigenesis as matrix synthesizing or matrix degrading cells, contractile cells, and even blood vessel associated cells. The production of growth factors, cytokines, chemokines, matrix-degrading enzymes, and immunomodulatory mechanisms by these cells augment tumor progression by providing a suitable environment. There are several suggested origins of the TAF including tissue-resident, circulating, and epithelial-to-mesenchymal-transitioned cells. METHODOLOGY/PRINCIPAL FINDINGS: We provide evidence that TAF are derived from mesenchymal stem cells (MSC) that acquire a TAF phenotype following exposure to or systemic recruitment into adenocarcinoma xenograft models including breast, pancreatic, and ovarian. We define the MSC derived TAF in a xenograft ovarian carcinoma model by the immunohistochemical presence of 1) fibroblast specific protein and fibroblast activated protein; 2) markers phenotypically associated with aggressiveness, including tenascin-c, thrombospondin-1, and stromelysin-1; 3) production of pro-tumorigenic growth factors including hepatocyte growth factor, epidermal growth factor, and interleukin-6; and 4) factors indicative of vascularization, including alpha-smooth muscle actin, desmin, and vascular endothelial growth factor. We demonstrate that under long-term tumor conditioning in vitro, MSC express TAF-like proteins. Additionally, human MSC but not murine MSC stimulated tumor growth primarily through the paracrine production of secreted IL6. CONCLUSIONS/SIGNIFICANCE: Our results suggest the dependence of in vitro Skov-3 tumor cell proliferation is due to the presence of tumor-stimulated MSC secreted IL6. The subsequent TAF phenotype arises from the MSC which ultimately promotes tumor growth through the contribution of microvascularization, stromal networks, and the production of tumor-stimulating paracrine factors.


Asunto(s)
Linaje de la Célula , Fibroblastos/citología , Células Madre Mesenquimatosas/citología , Neoplasias Ováricas/patología , Animales , División Celular , Progresión de la Enfermedad , Femenino , Humanos , Trasplante Heterólogo
13.
Scand J Infect Dis ; 39(8): 714-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17654349

RESUMEN

Women visiting Estonian STD clinics were subjected to PCR assay for human papillomavirus (HPV), Chlamydia trachomatis and Ureaplasma urealyticum biovar 2. The overall prevalence of coinfection was 8%. The chlamydial infection was found to be associated with HPV, especially with high-risk HPV (OR=2.5, p<0.005) and most significantly in women over 41 y of age. C. trachomatis infection also occurred more frequently in U. urealyticum-infected than in U. urealyticum-free patients (OR=2.6, p=0.02). U. urealyticum infection did not associate with HPV status. The clinical significance of the association between C. trachomatis and U. urelyticum infection remains to be elucidated.


Asunto(s)
Alphapapillomavirus/genética , Infecciones por Chlamydia/epidemiología , Enfermedades Urogenitales Femeninas/epidemiología , Infecciones por Papillomavirus/epidemiología , Infecciones por Ureaplasma/epidemiología , Adolescente , Adulto , Anciano , Instituciones de Atención Ambulatoria , Infecciones por Chlamydia/complicaciones , Chlamydia trachomatis , Estonia/epidemiología , Femenino , Humanos , Persona de Mediana Edad , Infecciones por Papillomavirus/complicaciones , Prevalencia , Infecciones por Ureaplasma/complicaciones , Ureaplasma urealyticum
14.
Cancer Res ; 67(24): 11687-95, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18089798

RESUMEN

Mesenchymal stem cells (MSC) migrate to and proliferate within sites of inflammation and tumors as part of the tissue remodeling process. Radiation increases the expression of inflammatory mediators that could enhance the recruitment of MSC into the tumor microenvironment. To investigate this, bilateral murine 4T1 breast carcinomas (expressing renilla luciferase) were irradiated unilaterally (1 or 2 Gy). Twenty-four hours later, 2 x 10(5) MSC-expressing firefly luciferase were injected i.v. Mice were then monitored with bioluminescent imaging for expression of both renilla (tumor) and firefly (MSC) luciferase. Forty-eight hours postirradiation, levels of MSC engraftment were 34% higher in tumors receiving 2 Gy (P = 0.004) than in the contralateral unirradiated limb. Immunohistochemical staining of tumor sections from mice treated unilaterally with 2 Gy revealed higher levels of MSC in the parenchyma of radiated tumors, whereas a higher proportion of MSC remained vasculature-associated in unirradiated tumors. To discern the potential mediators involved in MSC attraction, in vitro migration assays showed a 50% to 80% increase in MSC migration towards conditioned media from 1 to 5 Gy-irradiated 4T1 cells compared with unirradiated 4T1 cells. Irradiated 4T1 cells had increased expression of the cytokines, transforming growth factor-beta1, vascular endothelial growth factor, and platelet-derived growth factor-BB, and this up-regulation was confirmed by immunohistochemistry in tumors irradiated in vivo. Interestingly, the chemokine receptor CCR2 was found to be up-regulated in MSC exposed to irradiated tumor cells and inhibition of CCR2 led to a marked decrease of MSC migration in vitro. In conclusion, clinically relevant low doses of irradiation increase the tropism for and engraftment of MSC in the tumor microenvironment.


Asunto(s)
Neoplasias Mamarias Animales/terapia , Mesodermo/fisiología , Neoplasias/radioterapia , Células Madre/fisiología , Adenoviridae/genética , Animales , Línea Celular Tumoral , Ingeniería Genética , Luciferasas/análisis , Luciferasas/genética , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/radioterapia , Mesodermo/efectos de la radiación , Ratones , Ratones Endogámicos BALB C , Neoplasias/patología , Trasplante de Células Madre , Células Madre/patología , Células Madre/efectos de la radiación , Transfección
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda