Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 78(23): 7851-7872, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34719737

RESUMEN

Although the development of hematopoietic stem cells (HSC) has been studied in great detail, their heterogeneity and relationships to different cell lineages remain incompletely understood. Moreover, the role of Vascular Adhesion Protein-1 in bone marrow hematopoiesis has remained unknown. Here we show that VAP-1, an adhesin and a primary amine oxidase producing hydrogen peroxide, is expressed on a subset of human HSC and bone marrow vasculature forming a hematogenic niche. Bulk and single-cell RNAseq analyses reveal that VAP-1+ HSC represent a transcriptionally unique small subset of differentiated and proliferating HSC, while VAP-1- HSC are the most primitive HSC. VAP-1 generated hydrogen peroxide acts via the p53 signaling pathway to regulate HSC proliferation. HSC expansion and differentiation into colony-forming units are enhanced by inhibition of VAP-1. Contribution of VAP-1 to HSC proliferation was confirmed with mice deficient of VAP-1, mice expressing mutated VAP-1 and using an enzyme inhibitor. In conclusion, VAP-1 expression allows the characterization and prospective isolation of a new subset of human HSC. Since VAP-1 serves as a check point-like inhibitor in HSC differentiation, the use of VAP-1 inhibitors enables the expansion of HSC.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Sangre Fetal/citología , Hematopoyesis , Células Madre Hematopoyéticas/citología , Molécula 1 de Adhesión Celular Vascular/fisiología , Animales , Trasplante de Médula Ósea , Movimiento Celular , Femenino , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , RNA-Seq , Nicho de Células Madre
2.
J Cell Sci ; 133(10)2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32317394

RESUMEN

Extracellular adenosine mediates diverse anti-inflammatory, angiogenic and vasoactive effects, and has become an important therapeutic target for cancer, which has been translated into clinical trials. This study was designed to comprehensively assess adenosine metabolism in prostate and breast cancer cells. We identified cellular adenosine turnover as a complex cascade, comprising (1) the ectoenzymatic breakdown of ATP via sequential ecto-nucleotide pyrophosphatase/phosphodiesterase-1 (NPP1, officially known as ENPP1), ecto-5'-nucleotidase (CD73, also known as NT5E), and adenosine deaminase reactions, and ATP re-synthesis through a counteracting adenylate kinase and members of the nucleoside diphosphate kinase (NDPK, also known as NME/NM23) family; (2) the uptake of nucleotide-derived adenosine via equilibrative nucleoside transporters; and (3) the intracellular adenosine phosphorylation into ATP by adenosine kinase and other nucleotide kinases. The exposure of cancer cells to 1% O2 for 24 h triggered an ∼2-fold upregulation of CD73, without affecting nucleoside transporters, adenosine kinase activity and cellular ATP content. The ability of adenosine to inhibit the tumor-initiating potential of breast cancer cells via a receptor-independent mechanism was confirmed in vivo using a xenograft mouse model. The existence of redundant pathways controlling extracellular and intracellular adenosine provides a sufficient justification for reexamination of the current concepts of cellular purine homeostasis and signaling in cancer.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Adenosina Trifosfato , Neoplasias , Adenosina , Adenosina Difosfato , Adenilato Quinasa , Animales , Hipoxia , Masculino , Ratones , Neoplasias/genética , Nucleótidos
3.
Int J Cancer ; 139(10): 2270-6, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27459381

RESUMEN

Sunitinib and sorafenib are broad-spectrum tyrosine kinase inhibitors (TKI) targeting, for example, VEGF1-3, PDGFRb, RET, FLT3, CD117 (c-KIT) and CSF-1R cell membrane receptors thus suppressing tumor angiogenesis and cancer cell growth. Recently it has been suggested that the kinases targeted by Sunitinib and/or Sorafenib regulate leukocyte transmigration, which might in part be responsible for the often-observed reduction in tumor-associated myeloid derived suppressor cells and regulatory T cells. The aim of the current study is to determine whether sunitinib or sorafenib inhibit leukocyte extravasation. Sunitinib, sorafenib, or vehicle treated animals did not show any difference in leukocyte trafficking either in peritonitis or in vivo homing experiments, although sunitinib treatment effectively inhibited growth of B16 melanoma tumors in WT, SCID and SCID beige mice. Inhibition of tumor growth was associated with an increased number of infiltrating CD11b+ cells in the tumor, while the numbers of CD8, Gr-1 and F4/80 expressing cells were unchanged. In conclusion, the findings suggest that despite multiple targets with a potential role in leukocyte extravasation, neither sunitinib nor sorafenib effectively inhibits this process in vivo. Thus, the observed specific effect on CD11b cells among tumor infiltrating leukocytes is most likely an indirect effect.


Asunto(s)
Indoles/farmacología , Leucocitos/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Femenino , Leucocitos/enzimología , Leucocitos/inmunología , Leucocitos/patología , Melanoma Experimental/sangre , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Niacinamida/farmacología , Sorafenib , Sunitinib , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Migración Transendotelial y Transepitelial/efectos de los fármacos
4.
Proc Natl Acad Sci U S A ; 113(33): 9298-303, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27474165

RESUMEN

Macrophages are key regulators of fibrosis development and resolution. Elucidating the mechanisms by which they mediate this process is crucial for establishing their therapeutic potential. Here, we use experimental models of liver fibrosis to show that deficiency of the scavenger receptor, stabilin-1, exacerbates fibrosis and delays resolution during the recovery phase. We detected a subset of stabilin-1(+) macrophages that were induced at sites of cellular injury close to the hepatic scar in mouse models of liver fibrosis and in human liver disease. Stabilin-1 deficiency abrogated malondialdehyde-LDL (MDA-LDL) uptake by hepatic macrophages and was associated with excess collagen III deposition. Mechanistically, the lack of stabilin-1 led to elevated intrahepatic levels of the profibrogenic chemokine CCL3 and an increase in GFAP(+) fibrogenic cells. Stabilin-1(-/-) macrophages demonstrated a proinflammatory phenotype during liver injury and the normal induction of Ly6C(lo) monocytes during resolution was absent in stabilin-1 knockouts leading to persistence of fibrosis. Human stabilin-1(+) monocytes efficiently internalized MDA-LDL and this suppressed their ability to secrete CCL3, suggesting that loss of stabilin-1 removes a brake to CCL3 secretion. Experiments with cell-lineage-specific knockouts revealed that stabilin-1 expression in myeloid cells is required for the induction of this subset of macrophages and that increased fibrosis occurs in their absence. This study demonstrates a previously unidentified regulatory pathway in fibrogenesis in which a macrophage scavenger receptor protects against organ fibrosis by removing fibrogenic products of lipid peroxidation. Thus, stabilin-1(+) macrophages shape the tissue microenvironment during liver injury and healing.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Enfermedad Hepática Inducida por Sustancias y Drogas/complicaciones , Homeostasis , Cirrosis Hepática/prevención & control , Macrófagos/fisiología , Animales , Tetracloruro de Carbono , Quimiocina CCL3/fisiología , Deficiencia de Colina/complicaciones , Humanos , Lipoproteínas LDL/metabolismo , Malondialdehído/análogos & derivados , Malondialdehído/metabolismo , Ratones
5.
Duodecim ; 132(20): 1904-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-29190044

RESUMEN

There is usually enough time for identifying the etiology of calculous cholestasis and cholangitis by imaging and laboratory investigations, provided that antimicrobial drug therapy is started immediately after collection of blood cultures and the patient's general condition is good. The situation changes, if the inflammation is caused by Clostridium perfringens, a rare causative agent of severe sepsis and massive intravascular hemolysis. Mortality from the resulting infection and sepsis is as high as over 70%. Quick recognition of the condition, initiation of antimicrobial drug therapy and drainage of a possible focus of infection may save the patient's life.


Asunto(s)
Infecciones por Clostridium/microbiología , Clostridium perfringens/aislamiento & purificación , Absceso Hepático/microbiología , Sepsis/microbiología , Antibacterianos/uso terapéutico , Infecciones por Clostridium/terapia , Terapia Combinada , Drenaje , Hemólisis , Humanos , Absceso Hepático/terapia , Sepsis/terapia
6.
PLoS One ; 10(8): e0134721, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26258883

RESUMEN

The ectoenzyme CD73 catalyzes the hydrolysis of AMP, and is one of the most important producers of extracellular adenosine. On regulatory T cells, CD73 is necessary for immunosuppressive functions, and on Th17 cells CD73-generated adenosine exerts anti-inflammatory effects. However, the expression and function of CD73 in pro-inflammatory M1 and in immunosuppressive M2 macrophages is largely unknown. Here we show that CD73 expression and enzyme activity were induced in in vitro polarized pro-inflammatory human M(LPS+TNF) monocytes/macrophages, while CD73 was absent from immunosuppressive M(IL-4+M-CSF)-polarized macrophages. Inhibition of CD73 activity with the inhibitor AMPCP did not affect the polarization of human monocytes. In mice, CD73 was present on resident peritoneal macrophages. In striking contrast, elicited peritoneal macrophages remained CD73 negative regardless of their polarization towards either a pro-inflammatory M(LPS) or anti-inflammatory M(IL-4c) direction. Finally, the ability of peritoneal macrophages to polarize to pro- and anti-inflammatory cells was perfectly normal in CD73-deficient mice in vivo. These data indicate that, in contrast to other major leukocyte subpopulations, CD73 activity on macrophages does not play a major role in their polarization and that in mice host CD73 on any cell type is not required in vivo for peritoneal macrophage polarization towards either a pro- or an anti-inflammatory direction.


Asunto(s)
5'-Nucleotidasa/metabolismo , Macrófagos Peritoneales/metabolismo , Adenosina/química , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/química , Animales , Citocinas/metabolismo , Citometría de Flujo , Proteínas Ligadas a GPI/metabolismo , Humanos , Inmunosupresores/química , Inflamación , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Lipopolisacáridos/química , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos Peritoneales/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Fenotipo
7.
Antioxid Redox Signal ; 21(10): 1460-74, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24328532

RESUMEN

AIMS: Rat sarcoma virus (RAS)-induced tumorigenesis has been suggested to follow a three-stage model consisting of an initial RAS activation, senescence induction, and evasion of p53-dependent senescence checkpoints. While reactive oxygen species act as second messengers in RAS-induced senescence, they are also involved in oncogenic transformation by inducing proliferation and promoting mutations. In the current work, we investigated the role of extracellular superoxide dismutase (SOD3) in RAS-induced senescence and immortalization in vitro and in vivo. We used a mouse embryonic fibroblast (MEF) primary cell model along with immortalized and transformed human cell lines derived from papillary and anaplastic thyroid cancer. RESULTS: Based on our data, sod3 RNA interference in H-RasV12-transduced cells markedly inhibited cell growth, while sod3 over-expression in MEFs initially caused a proliferative burst followed by the activation of DNA damage checkpoints, induction of p53-p21 signal transduction, and senescence. Subsequently, sod3-transduced MEF cells developed co-operative p21-p16 down-regulation and acquired transformed cell characteristics such as increased telomerase activity, loss of contact inhibition, growth in low-nutrient conditions, and in vivo tumorigenesis. Interestingly, as previously reported with RAS, we showed a dose-dependent response to SOD3 in vitro and in vivo involving transcriptional and non-transcriptional regulatory mechanisms. INNOVATION: SOD3 may mediate H-RasV12-induced initiation of primary cell immortalization. CONCLUSIONS: Our results indicate that SOD3 influences growth signaling in primary and cancer cells downstream of the ras oncogene and could serve as a therapy target at an early tumorigenesis phase.


Asunto(s)
Transformación Celular Neoplásica , Embrión de Mamíferos/metabolismo , Superóxido Dismutasa/metabolismo , Animales , Embrión de Mamíferos/citología , Fibroblastos/citología , Ratones
8.
PLoS One ; 8(12): e84148, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24358335

RESUMEN

The NADPH oxidase 2 (NOX2) complex is a professional producer of reactive oxygen species (ROS) and is mainly expressed in phagocytes. While the activity of the NOX2 complex is essential for immunity against pathogens and protection against autoimmunity, its role in the development of malignant tumors remains unclear. We compared wild type and Ncf1 (m1J) mutated mice, which lack functional NOX2 complex, in four different tumor models. Ncf1 (m1J) mutated mice developed significantly smaller tumors in two melanoma models in which B16 melanoma cells expressing a hematopoietic growth factor FLT3L or luciferase reporter were used. Ncf1 (m1J) mutated mice developed significantly fewer Lewis Lung Carcinoma (LLC) tumors, but the tumors that did develop, grew at a pace that was similar to the wild type mice. In the spontaneously arising prostate carcinoma model (TRAMP), tumor growth was not affected. The lack of ROS-mediated protection against tumor growth was associated with increased production of immunity-associated cytokines. A significant increase in Th2 associated cytokines was observed in the LLC model. Our present data show that ROS regulate rejection of the antigenic B16-luc and LLC tumors, whereas the data do not support a role for ROS in growth of intrinsically generated tumors.


Asunto(s)
Carcinoma/genética , Carcinoma/patología , Melanoma/genética , Melanoma/patología , NADPH Oxidasas/genética , Neoplasias/genética , Neoplasias/patología , Animales , Carcinoma/metabolismo , Carcinoma/mortalidad , Carcinoma Pulmonar de Lewis , Modelos Animales de Enfermedad , Inflamación/genética , Inflamación/metabolismo , Melanoma/metabolismo , Melanoma/mortalidad , Melanoma Experimental , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mutación , NADPH Oxidasas/deficiencia , NADPH Oxidasas/metabolismo , Neoplasias/metabolismo , Neoplasias/mortalidad , Especies Reactivas de Oxígeno/inmunología , Carga Tumoral/genética
9.
Stem Cells ; 31(6): 1218-23, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23404893

RESUMEN

Mesenchymal stromal cells (MSCs) are able to influence the growth abilities of transformed cells. Here, we show that papillary thyroid cancer TPC1 and HEK 293T cells interact physically with human primary bone marrow-derived MSCs followed by evanescence of MSC cytoplasm. Interestingly, transformed cells were able to connect only to apoptotic MSCs that had lost their migration ability, whereas naïve MSCs avoided the direct contact. The interaction stimulated the proliferation of the cocultured transformed cells, activated mitogen and stress signaling, and increased resistance to cytotoxins. Consistent with in vitro data, the MSC interaction stimulated transformed cells had enhanced ability to grow and metastasize in vivo. The parental control cells showed mild tumorigenicity as compared to MSC interaction stimulated cells yielding measurable tumors in 31 days and 7 days, respectively. Our coculture model system describes how adjacent transformed cells absorb stromal cells thereby leading to the stroma-driven evolution of moderately carcinogenic cells to highly aggressive metastatic cells.


Asunto(s)
Células Madre Mesenquimatosas/patología , Animales , Atrofia , Células de la Médula Ósea/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Células HEK293 , Humanos , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal , Células del Estroma/patología , Neoplasias de la Tiroides/patología
10.
J Virol ; 86(24): 13779-84, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23035221

RESUMEN

Canine parvovirus (CPV) infection leads to reorganization of nuclear proteinaceous subcompartments. Our studies showed that virus infection causes a time-dependent increase in the amount of viral nonstructural protein NS1 mRNA. Fluorescence recovery after photobleaching showed that the recovery kinetics of nuclear transcription-associated proteins, TATA binding protein (TBP), transcription factor IIB (TFIIB), and poly(A) binding protein nuclear 1 (PABPN1) were different in infected and noninfected cells, pointing to virus-induced alterations in binding dynamics of these proteins.


Asunto(s)
Infecciones por Parvoviridae/metabolismo , Fracciones Subcelulares/metabolismo , Factores de Transcripción/metabolismo , Animales , Compartimento Celular , Parvovirus Canino/aislamiento & purificación
11.
Mediators Inflamm ; 2012: 730469, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22529530

RESUMEN

Extracellular superoxide dismutase (SOD3), an enzyme mediating dismutation of superoxide into hydrogen peroxide, has been shown to reduce inflammation by inhibiting macrophage migration into injured tissues. In inflamed tissues, superoxide is produced by the phagocytic NOX2 complex, which consists of the catalytic subunit NOX2 and several regulatory subunits (e.g., NCF1). To analyze whether SOD3 can regulate inflammation in the absence of functional NOX2 complex, we injected an adenoviral vector overexpressing SOD3 directly into the arthritic paws of Ncf1(∗/∗) mice with collagen-induced arthritis. SOD3 reduced arthritis severity in both oxidative burst-deficient Ncf1(∗/∗) mice and also in wild-type mice. The NOX2 complex independent anti-inflammatory effect of SOD3 was further characterized in peritonitis, and SOD3 was found to reduce macrophage infiltration independently of NOX2 complex functionality. We conclude that the SOD3-mediated anti-inflammatory effect on arthritis and peritonitis operates independently of NOX2 complex derived oxidative burst.


Asunto(s)
Artritis Experimental/metabolismo , Fagocitos/citología , Superóxido Dismutasa/metabolismo , Adenoviridae/metabolismo , Animales , Células COS , Macrófagos/citología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Peritonitis/metabolismo , Fagocitosis , Ratas , Estallido Respiratorio , Transducción de Señal
12.
PLoS One ; 6(8): e24456, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21909393

RESUMEN

BACKGROUND: Extracellular superoxide dismutase (SOD3), which dismutates superoxide anion to hydrogen peroxide, has been shown to reduce the free radical stress derived apoptosis in tissue injuries. Since both superoxide anion and hydrogen peroxide have a marked impact on signal transduction pathways and could potentially explain a number of apoptosis and survival -related phenomena in different pathological conditions, we clarified the impact of SOD3 on Akt and Erk1/2 cell survival pathways in rat hind limb injury model. METHODOLOGY AND PRINCIPAL FINDINGS: Based on our data, the hind limb ischemic rats treated with virally delivered sod3 have milder injury and less apoptosis than control animals that could be due to parallel activation of pro-proliferative and anti-apoptotic Erk1/2 and Akt pathways. The common downstream factor of both signaling pathways, the apoptosis related forkhead box protein O3a (FoxO3a), was phosphorylated and translocated to the cytoplasm in sod3 treated tissues and cell line. Additionally, we obtained increased mRNA production of elk-1, ets-1, and microRNA 21 (miR-21), whereas synthesis of bim mRNA was decreased in sod3 overexpressing tissues. We further showed that overexpression of sod3 modulated redox related gene expression by downregulating nox2 and inos when compared to injured control animals. CONCLUSIONS AND SIGNIFICANCE: The study shows the complexity of SOD3-derived effects on tissue injury recovery that are not limited to the reduction of superoxide anion caused cellular stress but highlights the impact of SOD3 related signal transduction on tissue functions and suggests an important role for SOD3 in attenuating cell stress effects in different pathological conditions.


Asunto(s)
Apoptosis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factores de Transcripción Forkhead/metabolismo , Isquemia/enzimología , Isquemia/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Superóxido Dismutasa/metabolismo , Animales , Supervivencia Celular , Proteína Forkhead Box O3 , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Biológicos , Células 3T3 NIH , Fosforilación , Ratas , Ratas Endogámicas F344
13.
Eur J Immunol ; 41(5): 1231-41, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21469131

RESUMEN

CD73/ecto-5'-nucleotidase dephosphorylates extracellular AMP into adenosine, and it is a key enzyme in the regulation of adenosinergic signaling. The contribution of host CD73 to tumor growth and anti-tumor immunity has not been studied. Here, we show that under physiological conditions CD73-deficient mice had significantly elevated ATPase and ADPase activities in LN T cells. In a melanoma model, the growth of primary tumors and formation of metastasis were significantly attenuated in mice lacking CD73. Among tumor-infiltrating leukocytes there were fewer Tregs and mannose receptor-positive macrophages, and increased IFN-γ and NOS2 mRNA production in CD73-deficient mice. Treatment of tumor-bearing animals with soluble apyrase, an enzyme hydrolyzing ATP and ADP, significantly inhibited tumor growth and accumulation of intratumoral Tregs and mannose receptor-positive macrophages in the WT C57BL/6 mice but not in the CD73-deficient mice. Pharmacological inhibition of CD73 with α,ß-methylene-adenosine-5'-diphosphate in WT mice retarded tumor progression similarly to the genetic deletion of CD73. Together these data show that increased pericellular ATP degradation in the absence of CD73 activity in the host cells is a novel mechanism controlling anti-tumor immunity and tumor progression, and that the purinergic balance can be manipulated therapeutically to inhibit tumor growth.


Asunto(s)
5'-Nucleotidasa/fisiología , Adenosina Trifosfatasas/metabolismo , Apirasa/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , 5'-Nucleotidasa/antagonistas & inhibidores , 5'-Nucleotidasa/genética , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Animales , Apirasa/farmacología , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Interferón gamma/genética , Lectinas Tipo C/genética , Linfocitos/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia/genética , Óxido Nítrico Sintasa de Tipo II/genética , Purinas/metabolismo , ARN Mensajero/genética , Receptores de Superficie Celular/genética , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
14.
Endocr Relat Cancer ; 17(3): 785-96, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20576801

RESUMEN

Reactive oxygen species, specifically hydrogen peroxide (H(2)O(2)), have a significant role in hormone production in thyroid tissue. Although recent studies have demonstrated that dual oxidases are responsible for the H(2)O(2) synthesis needed in thyroid hormone production, our data suggest a pivotal role for superoxide dismutase 3 (SOD3) as a major H(2)O(2)-producing enzyme. According to our results, Sod3 is highly expressed in normal thyroid, and becomes even more abundant in rat goiter models. We showed TSH-stimulated expression of Sod3 via phospholipase C-Ca(2+) and cAMP-protein kinase A, a pathway that might be disrupted in thyroid cancer. In line with this finding, we demonstrated an oncogene-dependent decrease in Sod3 mRNA expression synthesis in thyroid cancer cell models that corresponded to a similar decrease in clinical patient samples, suggesting that SOD3 could be used as a differentiation marker in thyroid cancer. Finally, the functional analysis in thyroid models indicated a moderate role for SOD3 in regulating normal thyroid cell proliferation being in line with our previous observations.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Diferenciación Celular , Superóxido Dismutasa/metabolismo , Neoplasias de la Tiroides/enzimología , Animales , Western Blotting , Calcio/metabolismo , Carcinoma , Carcinoma Papilar , Proliferación Celular , Regulación hacia Abajo , Humanos , Peróxido de Hidrógeno/metabolismo , Masculino , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/antagonistas & inhibidores , Superóxido Dismutasa/genética , Superóxidos/metabolismo , Cáncer Papilar Tiroideo , Carcinoma Anaplásico de Tiroides , Glándula Tiroides/enzimología , Glándula Tiroides/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Células Tumorales Cultivadas
15.
Cytotherapy ; 11(6): 726-37, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19878059

RESUMEN

BACKGROUND AIMS: Mesenchymal stromal cells (MSC) have been used in a wide variety of pre-clinical experiments and in an increasing number of human clinical trials. Although many of these studies have shown different levels of engraftment, the exact fate of MSC after transplantation and the tissue response to their engraftment have not been investigated in detail. In the present work we studied the distribution of human MSC in a rat hind limb ischemic injury model immediately after transplantation and also analyzed the recipient tissue response to transplanted cells. METHODS: We tracked the in vivo fate of the transplanted MSC utilizing bioluminescence imaging, fluorescence microscopy and gene/protein expression analysis in a rat hind limb ischemia model. We also monitored the viability of transplanted cells by graft versus recipient expression analysis and determined the angiogenic and proliferative effect of transplantation by histologic staining. RESULTS: According to imaging analysis only a small portion of cells persisted for an extended period of time at the site of injury. Interestingly, recipient versus graft expression studies showed increased synthesis of rat-origin angiogenic factors and no human-origin mRNA or protein synthesis in transplanted tissues. More importantly, despite the lack of robust engraftment or growth factor secretion the transplantation procedure exerted a significant pro-angiogenic and pro-proliferative effect, which was mediated by angiogenic and mitogenic signaling pathways. CONCLUSIONS: Our results show an immediate temporal tissue effect in response to MSC transplantation that may represent a novel indirect paracrine mechanism for the beneficial effects of cell transplantation observed in injured tissues.


Asunto(s)
Extremidad Inferior/irrigación sanguínea , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica , Daño por Reperfusión/cirugía , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Expresión Génica/fisiología , Reacción Injerto-Huésped/inmunología , Reacción Huésped-Injerto/inmunología , Humanos , Extremidad Inferior/patología , Extremidad Inferior/cirugía , Masculino , Células Madre Mesenquimatosas/citología , Ratas , Regeneración , Daño por Reperfusión/patología , Transducción de Señal/fisiología , Transducción Genética
16.
PLoS One ; 4(6): e5786, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19495415

RESUMEN

Inflammatory cell migration characteristic of ischemic damages has a dual role providing the tissue with factors needed for tissue injury recovery simultaneously causing deleterious development depending on the quality and the quantity of infiltrated cells. Extracellular superoxide dismutase (SOD3) has been shown to have an anti-inflammatory role in ischemic injuries where it increases the recovery process by activating mitogen signal transduction and increasing cell proliferation. However, SOD3 derived effects on inflammatory cytokine and adhesion molecule expression, which would explain reduced inflammation in vascular lesions, has not been properly characterized. In the present work the effect of SOD3 on the inflammatory cell extravasation was studied in vivo in rat hind limb ischemia and mouse peritonitis models by identifying the migrated cells and analyzing SOD3-derived response on inflammatory cytokine and adhesion molecule expression. SOD3 overexpression significantly reduced TNFalpha, IL1alpha, IL6, MIP2, and MCP-1 cytokine and VCAM, ICAM, P-selectin, and E-selectin adhesion molecule expressions in injured tissues. Consequently the mononuclear cell, especially CD68+ monocyte and CD3+ T cell infiltration were significantly decreased whereas granulocyte migration was less affected. According to our data SOD3 has a selective anti-inflammatory role in ischemic damages preventing the migration of reactive oxygen producing monocyte/macrophages, which in excessive amounts could potentially further intensify the tissue injuries therefore suggesting potential for SOD3 in treatment of inflammatory disorders.


Asunto(s)
Citocinas/biosíntesis , Inflamación , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/fisiología , Animales , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Movimiento Celular , Femenino , Granulocitos/citología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Ratas , Ratas Endogámicas F344 , Especies Reactivas de Oxígeno/metabolismo
17.
Mol Ther ; 17(3): 448-54, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19107121

RESUMEN

Extracellular superoxide dismutase (SOD3) gene therapy has been shown to attenuate tissue damages and to improve the recovery of the tissue injuries, but the cellular events delivering the therapeutic response of the enzyme are not well defined. In the current work, we overexpressed SOD3 in rat hindlimb ischemia model to study the signal transduction and injury healing following the sod3 gene transfer. The data suggest a novel sod3 gene transfer-derived signal transduction cascade through Ras-Mek-Erk mitogenic pathway leading to activation of AP1 and CRE transcription factors, increased vascular endothelial growth factor (VEGF)-A and cyclin D1 expression, increased cell proliferation, and consequently improved metabolic functionality of the injured tissue. Increased cell proliferation could explain the improved metabolic performance and the healing of the tissue damages after the sod3 gene transfer. The present data is a novel description of the molecular mechanism of SOD3-mediated recovery of tissue injury and suggests a new physiological role for SOD3 as a Ras regulatory molecule in signal transduction.


Asunto(s)
Espacio Extracelular/enzimología , Miembro Posterior/enzimología , Miembro Posterior/patología , Isquemia/enzimología , Isquemia/patología , Superóxido Dismutasa/metabolismo , Adenoviridae/genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica , Glucosa/metabolismo , Miembro Posterior/lesiones , Humanos , Isquemia/genética , Sistema de Señalización de MAP Quinasas , Masculino , Conejos , Ratas , Superóxido Dismutasa/genética , Transgenes/genética , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...