Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Sci Rep ; 9(1): 3177, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30816220

RESUMEN

Human aldose reductase (hAR, AKR1B1) has been explored as drug target since the 1980s for its implication in diabetic complications. An activated form of hAR was found in cells from diabetic patients, showing a reduced sensitivity to inhibitors in clinical trials, which may prevent its pharmacological use. Here we report the conversion of native hAR to its activated form by X-ray irradiation simulating oxidative stress conditions. Upon irradiation, the enzyme activity increases moderately and the potency of several hAR inhibitors decay before global protein radiation damage appears. The catalytic behavior of activated hAR is also reproduced as the KM increases dramatically while the kcat is not much affected. Consistently, the catalytic tetrad is not showing any modification. The only catalytically-relevant structural difference observed is the conversion of residue Cys298 to serine and alanine. A mechanism involving electron capture is suggested for the hAR activation. We propose that hAR inhibitors should not be designed against the native protein but against the activated form as obtained from X-ray irradiation. Furthermore, since the reactive species produced under irradiation conditions are the same as those produced under oxidative stress, the described irradiation method can be applied to other relevant proteins under oxidative stress environments.


Asunto(s)
Aldehído Reductasa/genética , Inhibidores Enzimáticos/farmacología , Estrés Oxidativo/efectos de la radiación , Alanina/genética , Aldehído Reductasa/antagonistas & inhibidores , Aldehído Reductasa/efectos de la radiación , Catálisis/efectos de los fármacos , Catálisis/efectos de la radiación , Microambiente Celular/efectos de la radiación , Activación Enzimática/efectos de la radiación , Inhibidores Enzimáticos/efectos de la radiación , Humanos , Oxidación-Reducción , Estrés Oxidativo/genética , Serina/genética , Rayos X
2.
Int J Radiat Oncol Biol Phys ; 103(1): 190-194, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30171879

RESUMEN

PURPOSE: Glioblastoma (GBM) remains incurable, despite state-of-the-art treatment involving surgical resection, chemotherapy, and radiation. GBM invariably recurs as a highly invasive and aggressive phenotype, with the majority of recurrences within the radiation therapy treatment field. Although a large body of literature reporting on primary GBM exists, comprehensive studies of how prior irradiation alters recurrent tumor growth are lacking. An animal model that replicates the delayed effects of radiation therapy on the brain microenvironment, and its impact on the development of recurrent GBM, would be a significant advance. METHODS AND MATERIALS: Cohorts of mice received a single fraction of 0, 20, 30, or 40 Gy Gamma Knife irradiation. Naïve, nonirradiated mouse GBM tumor cells were implanted into the ipsilateral hemisphere 6 weeks postirradiation. Tumor growth was measured by magnetic resonance imaging, and animal survival was assessed by monitoring weight loss. Magnetic resonance imaging results were supported by hemotoxylin and eosin histology. RESULTS: Tumorous lesions generated from orthotopic implantation of nonirradiated mouse GBM tumor cells into irradiated mouse brain grew far more aggressively and invasively than implantation of these same cells into nonirradiated brain. Lesions in irradiated brain tissue were significantly larger, more necrotic, and more vascular than those in control animals with increased invasiveness of tumor cells in the periphery, consistent with the histologic features commonly observed in recurrent high-grade tumors in patients. CONCLUSIONS: Irradiation of normal brain primes the targeted cellular microenvironment for aggressive tumor growth when naïve (not previously irradiated) cancer cells are subsequently introduced. The resultant growth pattern is similar to the highly aggressive pattern of tumor regrowth observed clinically after therapeutic radiation therapy. The mouse model offers an avenue for determining the cellular and molecular basis for the aggressiveness of recurrent GBM.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Encéfalo/efectos de la radiación , Microambiente Celular/efectos de la radiación , Glioblastoma/radioterapia , Animales , Encéfalo/patología , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de la radiación , Femenino , Glioblastoma/patología , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica
3.
Stem Cells Dev ; 27(18): 1237-1256, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29698131

RESUMEN

The stem cell compartment of the hematopoietic system constitutes one of the most radiosensitive tissues of the body and leukemias represent one of the most frequent radiogenic cancers with short latency periods. As such, leukemias may pose a particular threat to astronauts during prolonged space missions. Control of hematopoiesis is tightly governed by a specialized bone marrow (BM) microenvironment/niche. As such, any environmental insult that damages cells of this niche would be expected to produce pronounced effects on the types and functionality of hematopoietic/immune cells generated. We recently reported that direct exposure of human hematopoietic stem cells (HSC) to simulated solar energetic particle (SEP) and galactic cosmic ray (GCR) radiation dramatically altered the differentiative potential of these cells, and that simulated GCR exposures can directly induce DNA damage and mutations within human HSC, which led to leukemic transformation when these cells repopulated murine recipients. In this study, we performed the first in-depth examination to define changes that occur in mesenchymal stem cells present in the human BM niche following exposure to accelerated protons and iron ions and assess the impact these changes have upon human hematopoiesis. Our data provide compelling evidence that simulated SEP/GCR exposures can also contribute to defective hematopoiesis/immunity through so-called "biological bystander effects" by damaging the stromal cells that comprise the human marrow microenvironment, thereby altering their ability to support normal hematopoiesis.


Asunto(s)
Células de la Médula Ósea/efectos de la radiación , Radiación Cósmica/efectos adversos , Hematopoyesis/efectos de la radiación , Células Madre Mesenquimatosas/efectos de la radiación , Efecto Espectador , Microambiente Celular/efectos de la radiación , Daño del ADN/efectos de la radiación , Humanos , Hierro/química , Protones/efectos adversos , Energía Solar
4.
Radiat Res ; 189(2): 177-186, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29215326

RESUMEN

Radiation-induced skin fibrosis is a detrimental and chronic disorder that occurs after radiation exposure. The molecular changes underlying the pathogenesis of radiation-induced fibrosis of human skin have not been extensively reported. Technical advances in proteomics have enabled exploration of the biomarkers and molecular pathogenesis of radiation-induced skin fibrosis, with the potential to broaden our understanding of this disease. In this study, we compared protein expression in radiation-induced fibrotic human skin and adjacent normal tissues using iTRAQ-based proteomics technology. We identified 186 preferentially expressed proteins (53 upregulated and 133 downregulated) between radiogenic fibrotic and normal skin tissues. The differentially expressed proteins included keratins (KRT5, KRT6A, KRT16 and KRT17), caspase-14, fatty acid-binding protein 5 (FABP5), SLC2A14 and resistin. Through bioinformatic analysis of the proximal promoters, common motifs and corresponding transcriptional factors were identified that associate with the dysregulated proteins, including PAX5, TBX1, CLOCK and AP2D. In particular, FABP5 (2.15-fold increase in fibrotic skin tissues), a transporter of hydrophobic fatty acids, was investigated in greater detail. Immunohistochemistry confirmed that the protein level of FABP5 was increased in fibrotic human skin tissues, especially in the epidermis. Overexpression of FABP5 resulted in nuclear translocation of SMAD2 and significant activation of the profibrotic TGF-ß signaling pathway in human fibroblast WS1 cells. Moreover, exogenous FABP5 (FABP5-EGFP) could be incorporated by skin cells and intensify TGF-ß signaling, indicating a communication between the microenvironment and skin fibrosis. Taken together, our findings illustrate the molecular changes during radiation-induced human skin fibrosis and the critical role of FABP5 in activating the TGF-ß signaling pathway.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/metabolismo , Traumatismos por Radiación/metabolismo , Piel/patología , Piel/efectos de la radiación , Microambiente Celular/efectos de la radiación , Proteínas de Unión a Ácidos Grasos/genética , Fibrosis , Humanos , Proteómica , Traumatismos por Radiación/patología , Transducción de Señal/efectos de la radiación , Piel/metabolismo , Transcripción Genética/efectos de la radiación , Factor de Crecimiento Transformador beta/metabolismo
5.
Int J Med Sci ; 14(12): 1220-1230, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29104478

RESUMEN

The goal of our research was demonstrated that multiple molecules in microenvironments of the early osteoarthritis (OA) joint tissue may be actively responded to extracorporeal shockwave therapy (ESWT) treatment, which potentially regulated biological function of chondrocytes and synovial cells in early OA knee. We demonstrated that shockwave treatment induced the expression of protein-disulfide isomerase-associated 3 (Pdia-3) which was a significant mediator of the 1α,25-Dihydroxyvitamin D 3 (1α,25(OH)2D3) rapid signaling pathway, using two-dimensional electrophoresis, histological analysis and quantitative polymerase chain reaction (qPCR). We observed that the expression of Pdia-3 at 2 weeks was significantly higher than that of other group at 4, 8, and 12 weeks post-shockwave treatment in early OA rat knee model. The other factors of the rapid membrane signaling pathway, including extracellular signal-regulated protein kinases 1 (ERK1), osteopontin (OPG), alkaline phosphatase (ALP), and matrix metallopeptidase 13 (MMP13) were examined and were found to be significantly increased at 2 weeks post-shockwave treatment by qPCR in early OA of the knee. Our proteomic data revealed significant Pdia-3 expression in microenvironments of OA joint tissue that could be actively responded to ESWT, which may potentially regulate the biological functions of chondrocytes and osteoblasts in the treatment of the early OA of the knee.


Asunto(s)
Tratamiento con Ondas de Choque Extracorpóreas , Osteoartritis de la Rodilla/terapia , Proteína Disulfuro Isomerasas/metabolismo , Transducción de Señal , Vitamina D/análogos & derivados , Animales , Membrana Celular/metabolismo , Membrana Celular/efectos de la radiación , Microambiente Celular/efectos de la radiación , Condrocitos/metabolismo , Condrocitos/efectos de la radiación , Modelos Animales de Enfermedad , Humanos , Articulación de la Rodilla/citología , Articulación de la Rodilla/metabolismo , Articulación de la Rodilla/efectos de la radiación , Masculino , Osteoblastos/metabolismo , Osteoblastos/efectos de la radiación , Proteómica , Ratas , Ratas Sprague-Dawley , Vitamina D/metabolismo
6.
Biomaterials ; 82: 113-23, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26757258

RESUMEN

Cell detachment and migration from the endothelium occurs during vasculogenesis and also in pathological states. Here, we use a novel approach to trigger single cell release from an endothelial monolayer by in-situ opening of adhesive, fibril-like environment using light-responsive ligands and scanning lasers. Cell escapes from the monolayer were observed on the fibril-like adhesive tracks with 3-15 µm width. The frequency of endothelial cell escapes increased monotonically with the fibril width and with the density of the light-activated adhesive ligand. Interestingly, treatment with VEGF induced cohesiveness within the cell layer, preventing cell leaks. When migrating through the tracks, cells presented body lateral reduction and nuclear deformation imposed by the line width and dependent on myosin contractility. Cell migration mode changed from mesenchymal to amoeboid-like when the adhesive tracks narrowed (≤5 µm). Moreover, cell nucleus was shrunk showing packed DNA on lines narrower than the nuclear dimensions in a mechanisms intimately associated with the stress fibers. This platform allows the detailed study of escapes and migratory transitions of cohesive cells, which are relevant processes in development and during diseases such as organ fibrosis and carcinomas.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Microambiente Celular/fisiología , Células Endoteliales/fisiología , Matriz Extracelular/metabolismo , Oligopéptidos/metabolismo , Adhesividad/efectos de la radiación , Adhesión Celular/efectos de la radiación , Movimiento Celular/efectos de la radiación , Células Cultivadas , Microambiente Celular/efectos de la radiación , Células Endoteliales/citología , Células Endoteliales/efectos de la radiación , Endotelio/citología , Endotelio/fisiología , Endotelio/efectos de la radiación , Matriz Extracelular/química , Matriz Extracelular/efectos de la radiación , Humanos , Luz , Oligopéptidos/química , Oligopéptidos/efectos de la radiación
7.
PLoS One ; 10(6): e0128316, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26042591

RESUMEN

The response of the brain to irradiation is complex, involving a multitude of stress inducible pathways that regulate neurotransmission within a dynamic microenvironment. While significant past work has detailed the consequences of CNS radiotherapy following relatively high doses (≥ 45 Gy), few studies have been conducted at much lower doses (≤ 2 Gy), where the response of the CNS (like many other tissues) may differ substantially from that expected from linear extrapolations of high dose data. Low dose exposure could elicit radioadaptive modulation of critical CNS processes such as neurogenesis, that provide cellular input into hippocampal circuits known to impact learning and memory. Here we show that mice deficient for chemokine signaling through genetic disruption of the CCR2 receptor exhibit a neuroprotective phenotype. Compared to wild type (WT) animals, CCR2 deficiency spared reductions in hippocampal neural progenitor cell survival and stabilized neurogenesis following exposure to low dose irradiation. While radiation-induced changes in microglia levels were not found in WT or CCR2 deficient animals, the number of Iba1+ cells did differ between each genotype at the higher dosing paradigms, suggesting that blockade of this signaling axis could moderate the neuroinflammatory response. Interestingly, changes in proinflammatory gene expression were limited in WT animals, while irradiation caused significant elevations in these markers that were attenuated significantly after radioadaptive dosing paradigms in CCR2 deficient mice. These data point to the importance of chemokine signaling under low dose paradigms, findings of potential significance to those exposed to ionizing radiation under a variety of occupational and/or medical scenarios.


Asunto(s)
Microambiente Celular/efectos de la radiación , Hipocampo/citología , Hipocampo/efectos de la radiación , Exposición a la Radiación , Radiación Ionizante , Animales , Biomarcadores/metabolismo , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Corteza Cerebral/metabolismo , Corteza Cerebral/efectos de la radiación , Giro Dentado/citología , Relación Dosis-Respuesta en la Radiación , Regulación de la Expresión Génica/efectos de la radiación , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/efectos de la radiación , Neurogénesis/efectos de la radiación , Receptores CCR2/deficiencia , Receptores CCR2/metabolismo
8.
J Immunol ; 194(7): 3127-35, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25716994

RESUMEN

UV radiation (UVR) induces DNA damage, leading to the accumulation of mutations in epidermal keratinocytes and immunosuppression, which contribute to the development of nonmelanoma skin cancer. We reported previously that the TLR4-MyD88 signaling axis is necessary for UV-induced apoptosis. In the dinitrofluorobenzene contact hypersensitivity model, UV-irradiated MyD88-deficient (MyD88(-/-)) C57BL/6 mice had intact ear swelling, exaggerated inflammation, and higher levels of dinitrofluorobenzene-specific IgG2a compared with wild-type (WT) mice. Even with normal UV-induced, dendritic cell migration, DNA damage in the local lymph nodes was less pronounced in MyD88(-/-) mice compared with WT mice. Cultured, UV-irradiated WT APCs showed cleavage (inactivation) of the DNA damage-recognition molecule PARP, whereas PARP persisted in MyD88(-/-) and TLR4(-/-) APCs. Epidermal DNA from in vivo UV-irradiated MyD88(-/-) mice had an increased resolution rate of cyclobutane pyrimidine dimers. Both in vitro treatment of MyD88(-/-) APCs with and intradermal in vivo injections of PARP inhibitor, PJ-34, caused WT-level cyclobutane pyrimidine dimer repair. Lymphoblasts deficient in DNA repair (derived from a xeroderma pigmentosum group A patient) failed to augment DNA repair after MyD88 knockdown after UVR, in contrast to lymphoblasts from a healthy control. These data suggest that interference with the TLR4/MyD88 pathway may be a useful tool in promoting DNA repair and maintaining immune responses following UVR-induced damage.


Asunto(s)
Reparación del ADN , Terapia de Inmunosupresión , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal/efectos de la radiación , Piel/inmunología , Piel/metabolismo , Receptor Toll-Like 4/metabolismo , Rayos Ultravioleta , Animales , Microambiente Celular/genética , Microambiente Celular/inmunología , Microambiente Celular/efectos de la radiación , Daño del ADN/efectos de la radiación , Femenino , Humanos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/efectos de la radiación , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Piel/efectos de la radiación , Receptor Toll-Like 4/genética
9.
Acta Biomater ; 11: 274-82, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25257315

RESUMEN

Engineered tissue microenvironments impart specialized cues that drive distinct cellular phenotypes and function. Microenvironments with defined properties, such as mechanical properties and fibril alignment, can elicit specific cellular responses that emulate those observed in vivo. Collagen- and glycosaminoglycan (GAG)-based tissue matrices have been popularized due to their biological ubiquity in a broad range of tissues and the ability to tune structure and mechanical properties through a variety of processes. Here, we investigate the combined effects of static magnetic fields, and GAG and cell encapsulation, on the structure (e.g. collagen fibril orientation) and material properties of collagen matrices. We found that magnetic fields align the collagen-GAG matrix, alter equilibrium mechanical properties and provide a method for encapsulating cells within a three-dimensional aligned matrix. Cells are encapsulated prior to polymerization, allowing for controlled cell density and eliminating the need for cell seeding. Increased relative GAG concentrations reduced the ability to magnetically align collagen fibrils, in part through a mechanism involving increased viscosity and polymerization time of the collagen-GAG solution. This work provides a functional design space for the development of pure collagen and hybrid collagen-GAG matrices in the presence of magnetic fields. Additionally, this work shows that magnetic fields are effective for the fabrication of collagen constructs with controlled fibril orientation, and can be coupled with GAG incorporation to modulate mechanical properties and the response of embedded cells.


Asunto(s)
Microambiente Celular/fisiología , Condrocitos/citología , Colágeno/química , Matriz Extracelular/química , Matriz Extracelular/clasificación , Glicosaminoglicanos/química , Ingeniería de Tejidos/métodos , Animales , Materiales Biomiméticos/síntesis química , Bovinos , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Microambiente Celular/efectos de la radiación , Condrocitos/fisiología , Colágeno/efectos de la radiación , Fuerza Compresiva/fisiología , Fuerza Compresiva/efectos de la radiación , Módulo de Elasticidad/fisiología , Módulo de Elasticidad/efectos de la radiación , Campos Magnéticos , Ensayo de Materiales , Mecanotransducción Celular/fisiología , Mecanotransducción Celular/efectos de la radiación , Viscosidad/efectos de la radiación
10.
Mol Cancer Res ; 11(7): 793-807, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23615526

RESUMEN

Evidence suggests that bioactive lipids may regulate pathophysiologic functions such as cancer cell metastasis. Therefore, we determined that the bioactive lipid chemoattractants sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P) strongly enhanced the in vitro motility and adhesion of human rhabdomyosarcoma (RMS) cells. Importantly, this effect was observed at physiologic concentrations for both bioactive lipids, which are present in biologic fluids, and were much stronger than the effects observed in response to known RMS prometastatic factors such as stromal derived factors-1 (SDF-1/CXCL12) or hepatocyte growth factor/scatter factor (HGF/SF). We also present novel evidence that the levels of S1P and C1P were increased in several organs after γ-irradiation or chemotherapy, which indicates an unwanted prometastatic environment related to treatment. Critically, we found that the metastasis of RMS cells in response to S1P can be effectively inhibited in vivo with the S1P-specific binder NOX-S93 that is based on a high-affinity Spiegelmer. These data indicate that bioactive lipids play a vital role in dissemination of RMS and contribute to the unwanted side effects of radio/chemotherapy by creating a prometastatic microenvironment.


Asunto(s)
Antineoplásicos/uso terapéutico , Ceramidas/metabolismo , Lisofosfolípidos/metabolismo , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/radioterapia , Esfingosina/análogos & derivados , Actinas/metabolismo , Animales , Antineoplásicos/farmacología , Aptámeros de Nucleótidos/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Adhesión Celular/efectos de los fármacos , Adhesión Celular/efectos de la radiación , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Microambiente Celular/efectos de los fármacos , Microambiente Celular/efectos de la radiación , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/efectos de la radiación , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/patología , Esfingosina/metabolismo
11.
J Mammary Gland Biol Neoplasia ; 18(1): 3-13, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23325014

RESUMEN

Radiation exposure is a well-documented risk factor for breast cancer in women. Compelling epidemiological evidence in different exposed populations around the world demonstrate that excess breast cancer increases with radiation doses above 10 cGy. Both frequency and type of breast cancer are affected by prior radiation exposure. Many epidemiological studies suggest that radiation risk is inversely related to age at exposure; exposure during puberty poses the greatest risk while exposures past the menopause appear to carry very low risk. These observations are supported by experimental studies in mice and rats, which together provide the basis for the pubertal 'window of susceptibility' hypothesis for carcinogenic exposure. One line of experimental investigation suggests that the pubertal epithelium is more sensitive because DNA damage responses are less efficient, an other suggests that radiation affects stem cells self-renewal. A recent line of investigation suggests that the irradiated microenvironment mediates cancer risk. Studying the biological basis for radiation effects provides potential routes for protection in vulnerable populations, which include survivors of childhood cancers, as well as insights into the biology for certain types of sporadic cancer.


Asunto(s)
Neoplasias de la Mama/etiología , Glándulas Mamarias Humanas/efectos de la radiación , Salud de la Mujer , Factores de Edad , Animales , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/metabolismo , Microambiente Celular/efectos de la radiación , Femenino , Inestabilidad Genómica/efectos de la radiación , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/efectos de la radiación , Glándulas Mamarias Humanas/crecimiento & desarrollo , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Tolerancia a Radiación , Riesgo , Células Madre/efectos de la radiación
12.
Stem Cells ; 31(2): 372-83, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23169593

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs), which continuously maintain all mature blood cells, are regulated within the marrow microenvironment. We previously reported that pharmacologic treatment of naïve mice with prostaglandin E2 (PGE2) expands HSPCs. However, the cellular mechanisms mediating this expansion remain unknown. Here, we demonstrate that PGE2 treatment in naïve mice inhibits apoptosis of HSPCs without changing their proliferation rate. In a murine model of sublethal total body irradiation (TBI), in which HSPCs are rapidly lost, treatment with a long-acting PGE2 analog (dmPGE2) reversed the apoptotic program initiated by TBI. dmPGE2 treatment in vivo decreased the loss of functional HSPCs following radiation injury, as demonstrated both phenotypically and by their increased reconstitution capacity. The antiapoptotic effect of dmPGE2 on HSPCs did not impair their ability to differentiate in vivo, resulting instead in improved hematopoietic recovery after TBI. dmPGE2 also increased microenvironmental cyclooxygenase-2 expression and expanded the α-smooth muscle actin-expressing subset of marrow macrophages, thus enhancing the bone marrow microenvironmental response to TBI. Therefore, in vivo treatment with PGE2 analogs may be particularly beneficial to HSPCs in the setting of injury by targeting them both directly and also through their niche. The current data provide rationale for in vivo manipulation of the HSPC pool as a strategy to improve recovery after myelosuppression.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Dinoprostona/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Protectores contra Radiación/farmacología , Actinas/genética , Actinas/inmunología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Células de la Médula Ósea/patología , Células de la Médula Ósea/efectos de la radiación , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Microambiente Celular/efectos de los fármacos , Microambiente Celular/efectos de la radiación , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/inmunología , Dinoprostona/análogos & derivados , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/efectos de la radiación , Macrófagos/patología , Macrófagos/efectos de la radiación , Masculino , Ratones , Ratones Transgénicos , Traumatismos Experimentales por Radiación/inmunología , Traumatismos Experimentales por Radiación/patología , Irradiación Corporal Total
14.
J Cell Sci ; 125(Pt 9): 2134-40, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22357956

RESUMEN

Cell shape in vitro can be directed by geometrically defined micropatterned adhesion substrates. However conventional methods are limited by the fixed micropattern design, which cannot recapitulate the dynamic changes of the cell microenvironment. Here, we manipulate the shape of living cells in real time by using a tightly focused pulsed laser to introduce additional geometrically defined adhesion sites. The sub-micrometer resolution of the laser patterning allowed us to identify the critical distances between cell adhesion sites required for cell shape extension and contraction. This easy-to-handle method allows the precise control of specific actin-based structures that regulate cell architecture. Actin filament bundles or branched meshworks were induced, displaced or removed in response to specific dynamic modifications of the cell adhesion pattern. Isotropic branched actin meshworks could be forced to assemble new stress fibers locally and polarised in response to specific geometrical cues.


Asunto(s)
Actinas/metabolismo , Forma de la Célula/efectos de la radiación , Epitelio Pigmentado de la Retina/efectos de la radiación , Fibras de Estrés/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular/efectos de la radiación , Polaridad Celular/efectos de la radiación , Microambiente Celular/efectos de la radiación , Vectores Genéticos , Humanos , Rayos Láser , Lentivirus , Microscopía de Fuerza Atómica , Epitelio Pigmentado de la Retina/citología , Transducción Genética
15.
Artículo en Inglés | MEDLINE | ID: mdl-23366161

RESUMEN

Development of novel engineering techniques that can promote new clinical treatments requires implementing multidisciplinary in-vitro and in-vivo approaches. In this study, we have implemented microfluidic devices and in-vivo rat model to study the mechanism of neural stem cell migration and differentiation. These studies can result in the treatment of damages to the neuronal system. In this research, we have shown that by applying appropriate ranges of biochemical and biomechanical factors as well as by exposing the cells to electromagnetic fields, it is possible to improve viability, proliferation, directional migration and differentiation of neural stem cells. The results of this study can be implemented in the design of optimized platforms that can be transplanted into the damaged areas of the neuronal system.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Microambiente Celular/efectos de la radiación , Campos Electromagnéticos , Técnicas Analíticas Microfluídicas/métodos , Células-Madre Neurales/efectos de la radiación , Animales , Fenómenos Biomecánicos , Técnicas de Cultivo de Célula/instrumentación , Procesos de Crecimiento Celular/efectos de la radiación , Modelos Animales de Enfermedad , Colágenos Fibrilares/metabolismo , Técnicas Analíticas Microfluídicas/instrumentación , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Células-Madre Neurales/citología , Ratas
16.
Endocrinology ; 152(12): 4525-36, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22045660

RESUMEN

PTH stimulates bone formation and increases hematopoietic stem cells through mechanisms as yet uncertain. The purpose of this study was to identify mechanisms by which PTH links actions on cells of hematopoietic origin with osteoblast-mediated bone formation. C57B6 mice (10 d) were nonlethally irradiated and then administered PTH for 5-20 d. Irradiation reduced bone marrow cellularity with retention of cells lining trabeculae. PTH anabolic activity was greater in irradiated vs. nonirradiated mice, which could not be accounted for by altered osteoblasts directly or osteoclasts but instead via an altered bone marrow microenvironment. Irradiation increased fibroblast growth factor 2, TGFß, and IL-6 mRNA levels in the bone marrow in vivo. Irradiation decreased B220 cell numbers, whereas the percent of Lin(-)Sca-1(+)c-kit(+) (LSK), CD11b(+), CD68(+), CD41(+), Lin(-)CD29(+)Sca-1(+) cells, and proliferating CD45(-)Nestin(+) cells was increased. Megakaryocyte numbers were reduced with irradiation and located more closely to trabecular surfaces with irradiation and PTH. Bone marrow TGFß was increased in irradiated PTH-treated mice, and inhibition of TGFß blocked the PTH augmentation of bone in irradiated mice. In conclusion, irradiation created a permissive environment for anabolic actions of PTH that was TGFß dependent but osteoclast independent and suggests that a nonosteoclast source of TGFß drives mesenchymal stem cell recruitment to support PTH anabolic actions.


Asunto(s)
Médula Ósea/efectos de la radiación , Microambiente Celular/efectos de la radiación , Hormona Paratiroidea/metabolismo , Animales , Recuento de Células , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Osteoclastos , Hormona Paratiroidea/administración & dosificación , ARN Mensajero/efectos de la radiación , Factor de Crecimiento Transformador beta/fisiología , Irradiación Corporal Total
17.
PLoS One ; 6(10): e26437, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22028878

RESUMEN

To activate clot formation and maintain hemostasis, platelets adhere and spread onto sites of vascular injury. Although this process is well-characterized biochemically, how the physical and spatial cues in the microenvironment affect platelet adhesion and spreading remain unclear. In this study, we applied deep UV photolithography and protein micro/nanostamping to quantitatively investigate and characterize the spatial guidance of platelet spreading at the single cell level and with nanoscale resolution. Platelets adhered to and spread only onto micropatterned collagen or fibrinogen surfaces and followed the microenvironmental geometry with high fidelity and with single micron precision. Using micropatterned lines of different widths, we determined that platelets are able to conform to micropatterned stripes as thin as 0.6 µm and adopt a maximum aspect ratio of 19 on those protein patterns. Interestingly, platelets were also able to span and spread over non-patterned regions of up to 5 µm, a length consistent with that of maximally extended filopodia. This process appears to be mediated by platelet filopodia that are sensitive to spatial cues. Finally, we observed that microenvironmental geometry directly affects platelet biology, such as the spatial organization and distribution of the platelet actin cytoskeleton. Our data demonstrate that platelet spreading is a finely-tuned and spatially-guided process in which spatial cues directly influence the biological aspects of how clot formation is regulated.


Asunto(s)
Plaquetas/citología , Tamaño de la Célula , Microambiente Celular , Adhesividad Plaquetaria , Análisis de la Célula Individual/métodos , Adulto , Plaquetas/metabolismo , Microambiente Celular/efectos de la radiación , Colágeno/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/efectos de la radiación , Fibrinógeno/metabolismo , Humanos , Microtecnología , Nanotecnología , Adhesividad Plaquetaria/efectos de la radiación , Impresión , Seudópodos/metabolismo , Seudópodos/efectos de la radiación , Rayos Ultravioleta
18.
Biointerphases ; 6(4): 143-52, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22239806

RESUMEN

Cellular micropatterning with bio-adhesive and nonadhesive areas has attracted increasing interest for the precise design of cell-to-surface attachment in cell biology studies, tissue engineering, cell-based biosensors, biological assays, and drug development and screening. In this paper we describe a simple and efficient method to create a two-dimensional stable cellular microenvironment, which is based on (1) forming a protein-resistant oligo(ethylene glycol) methyl ether methacrylate polymer layer on the substrates via surface-initiated atom transfer radical polymerization; (2) placing a defined photomask on the substrate and exposing the substrate to ultraviolet light; and (3) immersing the patterned surface in a fibronectin solution to form cell-adhesive protein patterns in a cell-resistant background. The resulting surfaces are tailored into cell-adhesive and cell-resistant regions. Three different types of cells (NIH-3T3, PC12, bone marrow-derived mesenchymal stem cells) are seeded on such patterned surfaces to form cellular patterns. The geometric effects on cell behavior are investigated. The long-term stability is tested by NIH-3T3 fibroblasts and mesenchymal stem cells and excellent retention of cellular patterns is observed. The strategy illustrated here offers an efficient way to create a stable, patterned cellular microenvironment, and could be employed in tissue engineering to study the effect of micropatterns on the proliferation and differentiation of cells, and in particular mesenchymal stem cells.


Asunto(s)
Microambiente Celular , Fibroblastos/citología , Células Madre Mesenquimatosas/citología , Acrilatos/farmacología , Adsorción/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Microambiente Celular/efectos de los fármacos , Microambiente Celular/efectos de la radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibronectinas/farmacología , Fluoresceína-5-Isotiocianato/metabolismo , Fluorescencia , Vidrio/química , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Microscopía de Contraste de Fase , Células 3T3 NIH , Células PC12 , Espectroscopía de Fotoelectrones , Polietilenglicoles/farmacología , Polimerizacion/efectos de los fármacos , Ratas , Propiedades de Superficie/efectos de los fármacos , Factores de Tiempo , Rayos Ultravioleta , Agua
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA