Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(9)2023 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-37175480

RESUMEN

Five million non-melanoma skin cancers occur globally each year, and it is one of the most common malignant cancers. The dysregulation of the endocannabinoid system, particularly cannabinoid receptor 2 (CB2), is implicated in skin cancer development, progression, and metastasis. Comparing wildtype (WT) to systemic CB2 knockout (CB2-/-) mice, we performed a spontaneous cancer study in one-year old mice, and subsequently used the multi-stage chemical carcinogenesis model, wherein cancer is initiated by 7,12-dimethylbenz[a]anthracene (DMBA) and promoted by 12-O-tetradecanoylphorbol-13-acetate (TPA). We found that aging CB2-/- mice have an increased incidence of spontaneous cancerous and precancerous skin lesions compared to their WT counterparts. In the DMBA/TPA model, CB2-/- developed more and larger papillomas, had decreased spontaneous regression of papillomas, and displayed an altered systemic immune profile, including upregulated CD4+ T cells and dendritic cells, compared to WT mice. Immune cell infiltration in the tumor microenvironment was generally low for both genotypes, although a trend of higher myeloid-derived suppressor cells was observed in the CB2-/- mice. CB2 expression in carcinogen-exposed skin was significantly higher compared to naïve skin in WT mice, suggesting a role of CB2 on keratinocytes. Taken together, our data show that endogenous CB2 activation plays an anti-tumorigenic role in non-melanoma skin carcinogenesis, potentially via an immune-mediated response involving the alteration of T cells and myeloid cells coupled with the modulation of keratinocyte activity.


Asunto(s)
Papiloma , Neoplasias Cutáneas , Animales , Ratones , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Carcinogénesis/genética , Carcinogénesis/patología , Carcinógenos/toxicidad , Papiloma/patología , Receptores de Cannabinoides , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Acetato de Tetradecanoilforbol/toxicidad , Microambiente Tumoral
2.
Int J Mol Sci ; 24(4)2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36835468

RESUMEN

The endocannabinoid system, particularly cannabinoid receptor 2 (CB2 in mice and CNR2 in humans), has controversial pathophysiological implications in colon cancer. Here, we investigate the role of CB2 in potentiating the immune response in colon cancer in mice and determine the influence of CNR2 variants in humans. Comparing wild-type (WT) mice to CB2 knockout (CB2-/-) mice, we performed a spontaneous cancer study in aging mice and subsequently used the AOM/DSS model of colitis-associated colorectal cancer and a model for hereditary colon cancer (ApcMin/+). Additionally, we analyzed genomic data in a large human population to determine the relationship between CNR2 variants and colon cancer incidence. Aging CB2-/- mice exhibited a higher incidence of spontaneous precancerous lesions in the colon compared to WT controls. The AOM/DSS-treated CB2-/- and ApcMin/+CB2-/- mice experienced aggravated tumorigenesis and enhanced splenic populations of immunosuppressive myeloid-derived suppressor cells along with abated anti-tumor CD8+ T cells. Importantly, corroborative genomic data reveal a significant association between non-synonymous variants of CNR2 and the incidence of colon cancer in humans. Taken together, the results suggest that endogenous CB2 activation suppresses colon tumorigenesis by shifting the balance towards anti-tumor immune cells in mice and thus portray the prognostic value of CNR2 variants for colon cancer patients.


Asunto(s)
Carcinogénesis , Neoplasias del Colon , Receptor Cannabinoide CB2 , Animales , Humanos , Ratones , Carcinogénesis/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Ratones Noqueados , Receptor Cannabinoide CB2/genética , Receptor Cannabinoide CB2/metabolismo , Pronóstico
3.
Int J Mol Sci ; 23(19)2022 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-36233351

RESUMEN

Erythropoietin (EPO) is a pleiotropic cytokine that classically drives erythropoiesis but can also induce bone loss by decreasing bone formation and increasing resorption. Deletion of the EPO receptor (EPOR) on osteoblasts or B cells partially mitigates the skeletal effects of EPO, thereby implicating a contribution by EPOR on other cell lineages. This study was designed to define the role of monocyte EPOR in EPO-mediated bone loss, by using two mouse lines with conditional deletion of EPOR in the monocytic lineage. Low-dose EPO attenuated the reduction in bone volume (BV/TV) in Cx3cr1Cre EPORf/f female mice (27.05%) compared to controls (39.26%), but the difference was not statistically significant. To validate these findings, we increased the EPO dose in LysMCre model mice, a model more commonly used to target preosteoclasts. There was a significant reduction in both the increase in the proportion of bone marrow preosteoclasts (CD115+) observed following high-dose EPO administration and the resulting bone loss in LysMCre EPORf/f female mice (44.46% reduction in BV/TV) as compared to controls (77.28%), without interference with the erythropoietic activity. Our data suggest that EPOR in the monocytic lineage is at least partially responsible for driving the effect of EPO on bone mass.


Asunto(s)
Eritropoyetina , Receptores de Eritropoyetina , Animales , Eritropoyetina/metabolismo , Eritropoyetina/farmacología , Femenino , Ratones , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Transducción de Señal
4.
Front Pharmacol ; 12: 638128, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025407

RESUMEN

The common use of dental and orthopedic implants calls for special attention to the immune response leading to peri-prosthetic bone loss and implant failure. In addition to the well-established microbial etiology for oral implant failure, wear debris and in particular titanium (Ti) particles (TiP) in the implant vicinity are an important trigger of inflammation and activation of bone resorption around oral and orthopedic implants, presenting an unmet medical need. Here, we employed bacterial-derived lipopolysaccharides (LPS) to model infection and TiP to model aseptic inflammation and osteolysis. We assessed inflammation in vitro by measuring IL1ß, IL6 and TNFα mRNA expression in primary macrophages, osteoclastogenesis in RANKL-induced bone marrow derived pre-osteoclasts and osteolysis in vivo in a mouse calvarial model. We also assessed the trans-epithelial penetrability and safety of the tested compound in rats. Our results show that a lipophilic super-active derivative of vasoactive intestinal peptide (VIP), namely stearyl-norleucine-VIP (SNV) presented superior anti-inflammatory and anti-osteoclastogenic effects compared to VIP in vitro. In the bacterial infection model (LPS), SNV significantly reduced IL1ß expression, while VIP increased IL6 expression. In the aseptic models of osteolysis, SNV showed greater suppression of in vitro osteoclastogenesis than VIP, and significantly inhibited inflammation-induced osteolysis in vivo. We also observed that expression levels of the VIP receptor VPAC-2, but not that of VPAC-1, dramatically decreased during osteoclast differentiation. Importantly, SNV previously shown to have an increased stability compared to VIP, showed here significant trans-epithelial penetration and a clean toxicological profile, presenting a novel drug candidate that could be applied topically to counter both aseptic and infection-related bone destruction.

5.
Int J Mol Sci ; 23(1)2021 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-35008482

RESUMEN

The two erythropoietin (EPO) receptor forms mediate different cellular responses to erythropoietin. While hematopoiesis is mediated via the homodimeric EPO receptor (EPOR), tissue protection is conferred via a heteromer composed of EPOR and CD131. In the skeletal system, EPO stimulates osteoclast precursors and induces bone loss. However, the underlying molecular mechanisms are still elusive. Here, we evaluated the role of the heteromeric complex in bone metabolism in vivo and in vitro by using Cibinetide (CIB), a non-erythropoietic EPO analogue that exclusively binds the heteromeric receptor. CIB is administered either alone or in combination with EPO. One month of CIB treatment significantly increased the cortical (~5.8%) and trabecular (~5.2%) bone mineral density in C57BL/6J WT female mice. Similarly, administration of CIB for five consecutive days to female mice that concurrently received EPO on days one and four, reduced the number of osteoclast progenitors, defined by flow cytometry as Lin-CD11b-Ly6Chi CD115+, by 42.8% compared to treatment with EPO alone. In addition, CIB alone or in combination with EPO inhibited osteoclastogenesis in vitro. Our findings introduce CIB either as a stand-alone treatment, or in combination with EPO, as an appealing candidate for the treatment of the bone loss that accompanies EPO treatment.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Eritropoyetina/metabolismo , Oligopéptidos/farmacología , Osteogénesis/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Hematopoyesis/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo
6.
Front Immunol ; 11: 561294, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193330

RESUMEN

Immunotherapy with anti-CD20-specific antibodies (rituximab), has become the standard of care for B cell lymphoproliferative disorders and many autoimmune diseases. In rheumatological patients the effect of rituximab on bone mass yielded conflicting results, while in lymphoma patients it has not yet been described. Here, we used cross-sectional X-ray imaging (CT/PET-CT) to serially assess bone density in patients with follicular lymphoma receiving rituximab maintenance therapy. Remarkably, this treatment prevented the decline in bone mass observed in the control group of patients who did not receive active maintenance therapy. In accordance with these data, anti-CD20-mediated B cell depletion in normal C57BL/6J female mice led to a significant increase in bone mass, as reflected by a 7.7% increase in bone mineral density (whole femur), and a ~5% increase in cortical as well as trabecular tissue mineral density. Administration of anti-CD20 antibodies resulted in a significant decrease in osteoclastogenic signals, including RANKL, which correlated with a reduction in osteoclastogenic potential of bone marrow cells derived from B-cell-depleted animals. Taken together, our data suggest that in addition to its anti-tumor activity, anti-CD20 treatment has a favorable effect on bone mass. Our murine studies indicate that B cell depletion has a direct effect on bone remodeling.


Asunto(s)
Antígenos CD20/inmunología , Antineoplásicos Inmunológicos/administración & dosificación , Linfocitos B/inmunología , Densidad Ósea/efectos de los fármacos , Resorción Ósea/terapia , Inmunoterapia/métodos , Depleción Linfocítica , Linfoma Folicular/terapia , Rituximab/administración & dosificación , Adulto , Anciano , Animales , Estudios Transversales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Tomografía Computarizada por Tomografía de Emisión de Positrones , Estudios Retrospectivos , Resultado del Tratamiento
7.
Theranostics ; 10(19): 8744-8756, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32754275

RESUMEN

Erythropoietin (EPO) is a key regulator of erythropoiesis. However, EPO receptors (EPO-Rs) are also expressed on non-erythroid cell types, including myeloid and bone cells. Immune cells also participate in bone homeostasis. B cells produce receptor activator of nuclear factor kappa-Β ligand (RANKL) and osteoprotegerin (OPG), two pivotal regulators of bone metabolism. Here we explored the ability of B cells to transdifferentiate into functional osteoclasts and examined the role of EPO in this process in a murine model. Methods: We have combined specifically-designed experimental mouse models and in vitro based osteoclastogenesis assays, as well as PCR analysis of gene expression. Results: (i) EPO treatment in vivo increased RANKL expression in bone marrow (BM) B cells, suggesting a paracrine effect on osteoclastogenesis; (ii) B cell-derived osteoclastogenesis occured in vivo and in vitro, as demonstrated by B cell lineage tracing in murine models; (iii) B-cell-derived osteoclastogenesis in vitro was restricted to Pro-B cells expressing CD115/CSF1-R and is enhanced by EPO; (iv) EPO treatment increased the number of B-cell-derived preosteoclasts (ß3+CD115+), suggesting a physiological rationale for B cell derived osteoclastogenesis; (v) finally, mice with conditional EPO-R knockdown in the B cell lineage (cKD) displayed a higher cortical and trabecular bone mass. Moreover, cKD displayed attenuated EPO-driven trabecular bone loss, an effect that was observed despite the fact that cKD mice attained higher hemoglobin levels following EPO treatment. Conclusions: Our work highlights B cells as an important extra-erythropoietic target of EPO-EPO-R signaling and suggests their involvement in the regulation of bone homeostasis and possibly in EPO-stimulated erythropoietic response. Importantly, we present here for the first time, histological evidence for B cell-derived osteoclastogenesis in vivo.


Asunto(s)
Linfocitos B/citología , Remodelación Ósea/efectos de los fármacos , Eritropoyetina/farmacología , Receptores de Eritropoyetina/genética , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Transdiferenciación Celular/efectos de los fármacos , Femenino , Técnicas de Inactivación de Genes , Ratones , Osteogénesis , Ligando RANK/metabolismo , Receptores de Eritropoyetina/metabolismo
8.
Int J Mol Sci ; 21(11)2020 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-32471308

RESUMEN

Recent studies have demonstrated that erythropoietin (EPO) treatment in mice results in trabecular bone loss. Here, we investigated the dose-response relationship between EPO, hemoglobin (Hgb) and bone loss and examined the reversibility of EPO-induced damage. Increasing doses of EPO over two weeks led to a dose-dependent increase in Hgb in young female mice, accompanied by a disproportionate decrease in trabecular bone mass measured by micro-CT (µCT). Namely, increasing EPO from 24 to 540 IU/week produced a modest 12% rise in Hgb (20.2 ± 1.3 mg/dL vs 22.7 ± 1.3 mg/dL), while trabecular bone volume fraction (BV/TV) in the distal femur decreased dramatically (27 ± 8.5% vs 53 ± 10.2% bone loss). To explore the long-term skeletal effects of EPO, we treated mice for two weeks (540 IU/week) and monitored bone mass changes after treatment cessation. Six weeks post-treatment, there was only a partial recovery of the trabecular microarchitecture in the femur and vertebra. EPO-induced bone loss is therefore dose-dependent and mostly irreversible at doses that offer only a minor advantage in the treatment of anemia. Because patients requiring EPO therapy are often prone to osteoporosis, our data advocate for using the lowest effective EPO dose for the shortest period of time to decrease thromboembolic complications and minimize the adverse skeletal outcome.


Asunto(s)
Resorción Ósea/etiología , Eritropoyetina/efectos adversos , Animales , Resorción Ósea/diagnóstico por imagen , Resorción Ósea/patología , Hueso Esponjoso/diagnóstico por imagen , Hueso Esponjoso/efectos de los fármacos , Células Cultivadas , Eritropoyetina/administración & dosificación , Eritropoyetina/farmacología , Femenino , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Hemoglobinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Columna Vertebral/diagnóstico por imagen , Columna Vertebral/efectos de los fármacos
9.
J Bone Miner Res ; 34(12): 2277-2286, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31398266

RESUMEN

Krox20/EGR2 is a zinc finger transcription factor, implicated in the development of the hindbrain, nerve myelination, and tumor suppression. In skeletal biology, we have demonstrated that Krox20 also regulates adult bone metabolism. We and others have characterized several functions of Krox20 in the osteoclast lineage, namely, preosteoclast proliferation and differentiation, and mature osteoclast apoptosis. We have previously reported that systemically Krox20-haploinsufficient mice have a low bone mass with increased bone resorption. However, new data have now revealed that this phenotype is restricted to females. In addition, we discovered that conditional knockout of Krox20 (cKO) restricted to osteoclast progenitors is sufficient to induce the same female-specific bone loss observed in systemic mutants. To test whether this sexual dimorphism results from an interaction between Krox20 and sex hormones, we examined the sex- and hormone-dependent role of Krox20 deficiency on proliferation and apoptosis in osteoclastic cells. Our results indicate that male and female sex hormones (dihydrotestosterone [DHT] and estradiol [E2], respectively) as well as Krox20 inhibit preosteoclast proliferation and augment osteoclast apoptosis. The observation that Krox20 expression is inhibited by DHT and E2 negates the hypothesis that the effect of sex hormones is mediated by an increase in Krox20 expression. Interestingly, the effect of Krox20 deficiency was observed only with cells derived from female animals, regardless of any sex hormones added in vitro. In addition, we have identified sexual dimorphism in the expression of several Krox20-related genes, including NAB2. This sex-specific epigenetic profile was established at puberty, maintained in the absence of sex hormones, and explains the female-specific skeletal importance of Krox20. The findings described in this study emphasize the medical importance of sex differences, which may be determined at the epigenetic level. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Resorción Ósea/metabolismo , Resorción Ósea/patología , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Caracteres Sexuales , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Proteína 2 de la Respuesta de Crecimiento Precoz/deficiencia , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Haploinsuficiencia/genética , Masculino , Ratones Noqueados , Monocitos/metabolismo , Osteoclastos/efectos de los fármacos , Fenotipo
10.
Front Immunol ; 9: 2963, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619321

RESUMEN

The worldwide number of dental implants and orthopedic prostheses is steadily increasing. Orthopedic implant loosening, in the absence of infection, is mostly attributable to the generation of wear debris. Dental peri-implantitis is characterized by a multifactorial etiology and is the main cause of implant failure. It consists of a peri-implant inflammatory lesion that often results in loss of supporting bone. Disease management includes cleaning the surrounding flora by hand instruments, ultrasonic tips, lasers, or chemical agents. We recently published a paper indicating that US scaling of titanium (Ti) implants releases particles that provoke an inflammatory response and osteolysis. Here we show that a strong inflammatory response occurs; however, very few of the titanium particles are phagocytosed by the macrophages. We then measured a dramatic Ti particle-induced stimulation of IL1ß, IL6, and TNFα secretion by these macrophages using multiplex immunoassay. The particle-induced expression profile, examined by FACS, also indicated an M1 macrophage polarization. To assess how the secreted cytokines contributed to the paracrine exacerbation of the inflammatory response and to osteoclastogenesis, we treated macrophage/preosteoclast cultures with neutralizing antibodies against IL1ß, IL6, or TNFα. We found that anti-TNFα antibodies attenuated the overall expression of both the inflammatory cytokines and osteoclastogenesis. On the other hand, anti-IL1ß antibodies affected osteoclastogenesis but not the paracrine expression of inflammatory cytokines, whereas anti-IL6 antibodies did the opposite. We then tested these neutralizing antibodies in vivo using our mouse calvarial model of Ti particle-induced osteolysis and microCT analysis. Here, all neutralizing antibodies, administered by intraperitoneal injection, completely abrogated the particle-induced osteolysis. This suggests that blockage of paracrine inflammatory stimulation and osteoclastogenesis are similarly effective in preventing bone resorption induced by Ti particles. Blocking both the inflammation and osteoclastogenesis by anti-TNFα antibodies, incorporated locally into a slow-release membrane, also significantly prevented osteolysis. The osteolytic inflammatory response, fueled by ultrasonic scaling of Ti implants, results from an inflammatory positive feedback loop and osteoclastogenic stimulation. Our findings suggest that blocking IL1ß, IL6, and/or TNFα systemically or locally around titanium implants is a promising therapeutic approach for the clinical management of peri-implant bone loss.


Asunto(s)
Anticuerpos Neutralizantes/administración & dosificación , Implantes Dentales/efectos adversos , Macrófagos/inmunología , Osteólisis/inmunología , Periimplantitis/inmunología , Titanio/inmunología , Animales , Células Cultivadas , Citocinas/antagonistas & inhibidores , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Osteogénesis/inmunología , Osteólisis/diagnóstico por imagen , Osteólisis/patología , Osteólisis/prevención & control , Periimplantitis/diagnóstico por imagen , Periimplantitis/patología , Periimplantitis/prevención & control , Cultivo Primario de Células , Cráneo/diagnóstico por imagen , Cráneo/patología , Microtomografía por Rayos X
11.
J Cell Physiol ; 233(1): 371-377, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28266709

RESUMEN

The inbred mouse strain C57BL/6 is commonly used for the generation of transgenic mouse and is a well established strain in bone research. Different vendors supply different substrains of C57BL/6J as wild-type animals when genetic drift did not incur any noticeable phenotype. However, we sporadically observed drastic differences in the bone phenotype of "WT" C57BL/6J mice originating from different labs and speculated that these variations are attributable, at least in part, to the variation between C57BL/6J substrains, which is often overlooked. C57BL/6J-OlaHsd is a commonly used substrain that despite a well defined deletion in the alpha-synuclein (Snca) and multimerin-1 (Mmrn1) genes, was reported to display no obvious phenotype and is used as WT control. Here, we compared the bone phenotype of C57BL/6J-OlaHsd (6J-OLA) to C57BL/6J-RccHsd (6J-RCC) and to the original C57BL/6J (6J-JAX). Using µCT analysis, we found that 6J-OLA mice display a significantly lower trabecular bone mass compared to 6J-RCC and 6J-JAX. PCR analysis revealed that both the Snca and Mmrn1 genes are expressed in bone tissue of 6J-RCC animals but not of 6J-OLA mutants, suggesting either one or both genes play a role in bone metabolism. In vitro analysis demonstrated increase in osteoclasts number and decreased osteoblast mineralization in cells derived from 6J-OLA compared with 6J-RCC. Our data may shed light on unexplained differences in basal bone measurements between different research centers and reiterate the importance of specifying the exact substrain type. In addition, our findings describe the physiological role for Mmrn1 and/or Snca in bone remodeling.


Asunto(s)
Proteínas Sanguíneas/genética , Remodelación Ósea/genética , Moléculas de Adhesión Celular/genética , Mutación , Osteoporosis/genética , alfa-Sinucleína/genética , Animales , Proteínas Sanguíneas/metabolismo , Densidad Ósea , Calcificación Fisiológica , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Fémur/diagnóstico por imagen , Fémur/metabolismo , Fémur/fisiopatología , Predisposición Genética a la Enfermedad , Ratones Endogámicos C57BL , Ratones Mutantes , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Osteoporosis/diagnóstico por imagen , Osteoporosis/metabolismo , Osteoporosis/fisiopatología , Fenotipo , Microtomografía por Rayos X , alfa-Sinucleína/metabolismo
12.
Sci Rep ; 7: 39612, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-28059080

RESUMEN

With millions of new dental and orthopedic implants inserted annually, periprosthetic osteolysis becomes a major concern. In dentistry, peri-implantitis management includes cleaning using ultrasonic scaling. We examined whether ultrasonic scaling releases titanium particles and induces inflammation and osteolysis. Titanium discs with machined, sandblasted/acid-etched and sandblasted surfaces were subjected to ultrasonic scaling and we physically and chemically characterized the released particles. These particles induced a severe inflammatory response in macrophages and stimulated osteoclastogenesis. The number of released particles and their chemical composition and nanotopography had a significant effect on the inflammatory response. Sandblasted surfaces released the highest number of particles with the greatest nanoroughness properties. Particles from sandblasted/acid-etched discs induced a milder inflammatory response than those from sandblasted discs but a stronger inflammatory response than those from machined discs. Titanium particles were then embedded in fibrin membranes placed on mouse calvariae for 5 weeks. Using micro-CT, we observed that particles from sandblasted discs induced more osteolysis than those from sandblasted/acid-etched discs. In summary, ultrasonic scaling of titanium implants releases particles in a surface type-dependent manner and may aggravate peri-implantitis. Future studies should assess whether surface roughening affects the extent of released wear particles and aseptic loosening of orthopedic implants.


Asunto(s)
Implantes Dentales , Raspado Dental/efectos adversos , Osteólisis/etiología , Periimplantitis/inducido químicamente , Titanio/efectos adversos , Animales , Células Cultivadas , Femenino , Ratones Endogámicos C57BL , Osteogénesis
13.
Genome Res ; 26(5): 601-11, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26907635

RESUMEN

During development, enhancers play pivotal roles in regulating gene expression programs; however, their involvement in cancer progression has not been fully characterized. We performed an integrative analysis of DNA methylation, RNA-seq, and small RNA-seq profiles from thousands of patients, including 25 diverse primary malignances and seven body sites of metastatic melanoma. We found that enhancers are consistently the most differentially methylated regions (DMR) as cancer progresses from normal to primary tumors and then to metastases, compared to other genomic features. Remarkably, identification of enhancer DMRs (eDMRs) enabled classification of primary tumors according to physiological organ systems, and in metastasis eDMRs are the most correlated with patient outcome. To further understand the eDMR role in cancer progression, we developed a model to predict genes and microRNAs that are regulated by enhancer and not promotor methylation, which shows high accuracy with chromatin architecture methods and was experimentally validated. Interestingly, among all metastatic melanoma eDMRs, the most correlated with patient survival were eDMRs that "switched" their methylation patterns back and forth between normal, primary, and metastases and target cancer drivers, e.g., KIT We further demonstrated that eDMR target genes were modulated in melanoma by the bone metastasis microenvironment, suggesting that eDMRs respond to microenvironmental cues in metastatic niches. Our findings that aberrant methylation in cancer cells mostly affects enhancers, which contribute to tumor progression and cancer cell plasticity, will facilitate development of epigenetic anticancer approaches.


Asunto(s)
Metilación de ADN , ADN de Neoplasias , Elementos de Facilitación Genéticos , Melanoma , Línea Celular Tumoral , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Femenino , Humanos , Masculino , Melanoma/genética , Melanoma/metabolismo , Melanoma/mortalidad
14.
FASEB J ; 29(5): 1890-900, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25630969

RESUMEN

Erythropoietin (EPO) primarily regulates red blood cell formation, and EPO serum levels are increased on hypoxic stress (e.g., anemia and altitude). In addition to anemia, recent discoveries suggest new therapeutic indications for EPO, unrelated to erythropoiesis. We investigated the skeletal role of EPO using several models of overexpression (Tg6 mice) and EPO administration (intermittent/continuous, high/low doses) in adult C57Bl6 female mice. Using microcomputed tomography, histology, and serum markers, we found that EPO induced a 32%-61% trabecular bone loss caused by increased bone resorption (+60%-88% osteoclast number) and reduced bone formation rate (-19 to -74%; P < 0.05 throughout). EPO targeted the monocytic lineage by increasing the number of bone monocytes/macrophages, preosteoclasts, and mature osteoclasts. In contrast to the attenuated bone formation in vivo, EPO treatment in vitro did not inhibit osteoblast differentiation and activity, suggesting an indirect effect of EPO on osteoblasts. However, EPO had a direct effect on preosteoclasts by stimulating osteoclastogenesis in isolated cultures (+60%) via the Jak2 and PI3K pathways. In summary, our findings demonstrate that EPO negatively regulates bone mass and thus bears significant clinical implications for the potential management of patients with endogenously or therapeutically elevated EPO levels.


Asunto(s)
Resorción Ósea/etiología , Eritropoyetina/fisiología , Osteoclastos/citología , Receptores de Eritropoyetina/metabolismo , Animales , Apoptosis , Western Blotting , Resorción Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoclastos/metabolismo , Osteogénesis/fisiología , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Eritropoyetina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Microtomografía por Rayos X
15.
Am J Sports Med ; 42(12): 2869-76, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25184246

RESUMEN

BACKGROUND: Statins are lipid-lowering drugs with many beneficial pleiotropic effects. Cyclooxygenase (COX2) selective inhibitors that are commonly prescribed in orthopaedic patients may effect healing. Evidence indicates that statins stimulate COX2 activity. HYPOTHESIS: Atorvastatin (ATV) administration will enhance tendon healing by stimulating the acute inflammatory phase via increasing the production of prostaglandin E2 (PGE2). STUDY DESIGN: Controlled laboratory study. METHODS: After experimental rotator cuff (RC) tearing and suturing, 48 Wistar rats were randomly allocated into 4 groups: (1) ATV (20 mg/kg), (2) celecoxib (CEL; COX2 inhibitor) (10 mg/kg), (3) ATV + CEL (20 mg/kg + 10 mg/kg), and (4) saline alone. Animals were sacrificed 3 weeks after RC tears and repair, and tendon integrity was tested biomechanically in tension. To further evaluate the underlying mechanism of action, human and rat primary tenocytes were obtained from the supraspinatus tendon. Cultures were treated with a therapeutic dosage of 5 commonly used statins: CEL, ATV + CEL, PGE2, and a selective antagonist of PGE2 receptor 4 (EP4). Cell proliferation (thymidine incorporation), migration (wound healing assay), and adhesion (iCELLigence) were evaluated. The expression of all PGE2 receptors (EPs) was determined by quantitative reverse transcription polymerase chain reaction. RESULTS: Tension testing of healed tendons demonstrated significantly higher maximal loading and stiffness in the ATV group as compared with the saline (+30% and +20%, respectively; P < .001) and CEL groups (+33% and +50%, respectively; P < .005). Celecoxib alone did not affect tendon healing (P = .88). In line with these in vivo results, tenocytes treated with statins demonstrated significantly higher proliferation rates; CEL abrogated this effect, and PGE2 treatment stimulated tenocyte proliferation even in the presence of CEL. Also, ATV stimulated the migration (wound healing) and adhesion of tenocytes. Among all PGE2 receptors, tenocytes mainly express EP4, and an EP4 selective antagonist blocked the effect of ATV. CONCLUSION: Results indicate that ATV enhances tendon healing by stimulating tenocyte proliferation, migration, and adhesion via increased COX2 activity and autocrine/paracrine PGE2 signaling. Findings also demonstrate that this effect is mediated by EP4 signaling. CLINICAL RELEVANCE: Although chronic inflammation contributes to the development of tendinopathy, study results advocate for a positive role of PGE2 in tendon healing during the acute inflammatory phase that follows tendon surgical repair. It is therefore suggested that ATV should be further investigated as a possible modality to improve tendon healing.


Asunto(s)
Dinoprostona/metabolismo , Ácidos Heptanoicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Pirroles/farmacología , Manguito de los Rotadores/cirugía , Cicatrización de Heridas/efectos de los fármacos , Animales , Atorvastatina , Fenómenos Biomecánicos , Celecoxib , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Modelos Animales , Pirazoles/farmacología , Distribución Aleatoria , Ratas Wistar , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Sulfonamidas/farmacología , Tendones/citología
16.
Biochem J ; 427(2): 305-12, 2010 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-20136632

RESUMEN

EPO (erythropoietin), the major hormone regulating erythropoiesis, functions via activation of its cell-surface receptor (EPO-R) present on erythroid progenitor cells. One of the most striking properties of EPO-R is its low expression on the cell surface, as opposed to its high intracellular levels. The low cell-surface expression of EPO-R may thus limit the efficacy of EPO that is routinely used to treat primary and secondary anaemia. In a recent study [Nahari, Barzilay, Hirschberg and Neumann (2008) Biochem. J. 410, 409-416] we have shown that insertion of an NPVY sequence into the intracellular domain of EPO-R increases its cell-surface expression. In the present study we demonstrate that this NPVY EPO-R insert has a selective effect on EPO-mediated downstream signalling in Ba/F3 cells expressing this receptor (NPVY-EPO-R). This is monitored by increased phosphorylation of the NPVY-EPO-R (on Tyr479), Akt, JAK2 (Janus kinase 2) and ERK1/2 (extracellular-signal-regulated kinase 1/2), but not STAT5 (signal transducer and activator of transcription 5), as compared with cells expressing wild-type EPO-R. This enhanced signalling is reflected in augmented proliferation at low EPO levels (0.05 units/ml) and protection against etoposide-induced apoptosis. Increased cell-surface levels of NPVY-EPO-R are most probably not sufficient to mediate these effects as the A234E-EPO-R mutant that is expressed at high cell-surface levels does not confer an augmented response to EPO. Taken together, we demonstrate that insertion of an NPVY sequence into the cytosolic domain of the EPO-R confers not only improved maturation, but also selectively affects EPO-mediated signalling resulting in an improved responsiveness to EPO reflected in cell proliferation and protection against apoptosis.


Asunto(s)
Eritropoyetina/metabolismo , Receptores de Eritropoyetina/química , Transducción de Señal , Secuencia de Aminoácidos , Animales , Línea Celular , Proliferación Celular , Citosol , Humanos , Ratones , Mutación , Fosforilación , Receptores de Eritropoyetina/análisis , Receptores de Eritropoyetina/genética , Regulación hacia Arriba
17.
J Mol Cell Cardiol ; 42(4): 835-41, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17337000

RESUMEN

We have previously shown that domains involved in binding of protein kinase C (PKC) isozymes to their respective anchoring proteins (RACKs) and short peptides derived from these domains are PKC isozyme-selective antagonists. We also identified PKC isozyme-selective agonists, named psiRACK peptides, derived from a sequence within each PKC with high homology to its respective RACK. We noted that all the psiRACK sequences within each PKC isozyme have at least one non-homologous amino acid difference from their corresponding RACK that constitutes a charge change. Based on this information, we have devised here a new approach to design an isozyme-selective PKC antagonist, derived from the psiRACK sequence. We focused on epsilonPKC psiRACK peptide, where the pseudo-epsilonRACK sequence (psiepsilonRACK; HDAPIGYD; corresponding to epsilonPKC85-92) is different in charge from the homologous RACK-derived sequence (NNVALGYD; corresponding to epsilonRACK285-292) in the second amino acid. Here we show that changing the charge of the psiepsilonRACK peptide through a substitution of only one amino acid (aspartate to asparagine) resulted in a peptide with an opposite activity on the same cell function and a substitution for aspartate with an alanine resulted in an inactive peptide. These data support our hypothesis regarding the mechanism by which pseudo-RACK peptide activates PKC in heart cells and suggest that this approach is applicable to other signaling proteins with inducible protein-protein interactions.


Asunto(s)
Regulación Alostérica , Diseño de Fármacos , Fragmentos de Péptidos/metabolismo , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Células Cultivadas , Drosophila/metabolismo , Masculino , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fragmentos de Péptidos/química , Unión Proteica , Proteína Quinasa C-epsilon/metabolismo , Transporte de Proteínas , Ratas , Transducción de Señal
18.
J Pain ; 6(8): 535-49, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16084468

RESUMEN

UNLABELLED: Ethanol (EtOH) withdrawal increases sensitivity to painful stimuli in adult rats. In this study, withdrawal from a single, acute administration of EtOH dose-dependently produced mechanical allodynia and thermal hyperalgesia in postnatal day 7 (P7) rats. In contrast, P21 rats exhibited earlier and more prolonged mechanical allodynia but not thermal hyperalgesia. For both P7 and P21 rats, blood and spinal cord EtOH levels peaked at 30 minutes after administration, with P7 rats achieving overall higher spinal cord concentrations. Protein kinase C (PKC) has been implicated in mediating pain responses. Inhibitory PKC- and gamma-specific peptides attenuated mechanical allodynia and thermal hyperalgesia in P7 rats, whereas only the PKCgamma inhibitor prevented mechanical allodynia in P21 rats. Immunoreactive PKC in dorsal root ganglion and PKCgamma in lumbar spinal cord increased at 6 hours after EtOH administration in P7 rats. In P21 rats, the density of PKC immunoreactivity remained unchanged, whereas the density of PKCgamma immunoreactivity increased and translocation occurred. These studies demonstrate developmental differences in neonatal nociceptive responses after withdrawal from acute EtOH and implicate a role for specific PKC isozymes in EtOH withdrawal-associated allodynia and hyperalgesia. PERSPECTIVE: This study examines age-specific nociceptive responses after ethanol exposure by using 2 different ages of rats. The results suggest that ethanol age-dependently alters sensitivity to mechanical and thermal stimuli via specific protein kinase C isozymes. These results begin to ascertain the mechanisms that produce abnormal pain after alcohol exposure.


Asunto(s)
Trastornos del Sistema Nervioso Inducidos por Alcohol/enzimología , Etanol/efectos adversos , Hiperalgesia/enzimología , Proteína Quinasa C-epsilon/efectos de los fármacos , Proteína Quinasa C/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/enzimología , Factores de Edad , Envejecimiento/fisiología , Trastornos del Sistema Nervioso Inducidos por Alcohol/fisiopatología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/fisiopatología , Depresores del Sistema Nervioso Central/efectos adversos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Femenino , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Inmunohistoquímica , Isoenzimas/efectos de los fármacos , Isoenzimas/metabolismo , Masculino , Péptidos/farmacología , Proteína Quinasa C/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Síndrome de Abstinencia a Sustancias/fisiopatología
19.
Biochem Biophys Res Commun ; 318(4): 949-54, 2004 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-15147964

RESUMEN

Intracellularly acting peptide modulators of signaling enzymes provide a powerful means to regulate signaling events. Delivery of peptides into cells is facilitated by conjugation to carrier peptides, such as Tat. When peptides are irreversibly conjugated to Tat, Tat-mediated subcellular localization may predominate, resulting in mislocalization of the peptide cargo. We have used intracellularly acting peptides, conjugated to Tat by a disulfide bond, to modulate protein kinase C (PKC) signaling; these PKC-modulating peptides are released from Tat upon intracellular delivery. Previously, the distribution of these peptides within tissue and throughout the body had not been demonstrated. We show here intravascular delivery of a PKC-peptide, reversibly conjugated to Tat, resulted in distribution throughout cardiac tissue. In addition, a single injection resulted in selective modulation of PKC activity in many organs. Therefore, intracellularly acting peptide modulators of signaling enzymes, reversibly conjugated to Tat, have extensive biodistribution and can be used to modulate signaling pathways in vivo.


Asunto(s)
Productos del Gen tat/farmacocinética , Péptidos/farmacocinética , Proteína Quinasa C/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Productos del Gen tat/química , Inyecciones Intraperitoneales , Isoenzimas , Miocardio/metabolismo , Péptidos/química , Péptidos/farmacología , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Ratas , Distribución Tisular , Xantenos/metabolismo
20.
Biochem Cell Biol ; 80(5): 655-65, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12440705

RESUMEN

Adenosine is a physiologically important nucleoside in the cardiovascular system where it can act as a cardioprotectant and modulator of energy usage. Adenosine transporters (ATs) modulate cellular adenosine levels, which, in turn, can affect a number of processes such as receptor activation and glucose uptake, but their role in cardiac physiology is poorly understood. Therefore, we have developed a new cell model by determining various adenosine-related characteristics of HL-1, an immortalized atrial cardiomyocyte murine cell line. Adenosine uptake in HL-1 cells is sodium independent, saturable, and inhibitable by nucleoside transport inhibitors (nitrobenzylthioinosine (NBTI), dipyridamole, dilazep). Reverse transcription--polymerase chain reaction analysis confirmed that HL-1 cells possess mouse equilibrative nucleoside transporters 1 and 2 (mENT1, mENT2) and kinetic analyses indicate moderate-affinity (Km = 51.3 +/- 12.9 microM), NBTI-sensitive adenosine transport. NBTI binds at a high-affinity single site (B(max) = 520 +/- 10 fmol/mg protein, Kd = 0.11 +/- 0.04 nM, 1.6 x 10(5) NBTI-binding sites/cell). HL-1 cells possess adenosine receptor, metabolic enzyme, protein kinase C isoform, and insulin-stimulated glucose transport profiles that match normal mouse heart. Therefore, HL-1 is an excellent model to study ATs within cardiomyocytes and the first model for evaluating in detail the role of the ATs in modulating effects of adenosine.


Asunto(s)
Adenosina/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Transporte de Membrana , Miocitos Cardíacos/metabolismo , Tioinosina/análogos & derivados , Adenosina Desaminasa/metabolismo , Adenosina Quinasa/metabolismo , Marcadores de Afinidad/metabolismo , Marcadores de Afinidad/farmacología , Animales , Transporte Biológico , Northern Blotting , Proteínas Portadoras/genética , Células Cultivadas , Cartilla de ADN/química , Tranportador Equilibrativo 1 de Nucleósido/antagonistas & inhibidores , Tranportador Equilibrativo 1 de Nucleósido/genética , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Transportador Equilibrativo 2 de Nucleósido/antagonistas & inhibidores , Transportador Equilibrativo 2 de Nucleósido/genética , Transportador Equilibrativo 2 de Nucleósido/metabolismo , Glucosa/metabolismo , Insulina/farmacología , Ratones , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Proteínas de Transporte de Nucleósidos , Proteína Quinasa C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Tioinosina/metabolismo , Tioinosina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...