Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
BMC Biol ; 19(1): 154, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330275

RESUMEN

BACKGROUND: The skeletal muscle plays a central role in glucose homeostasis through the uptake of glucose from the extracellular medium in response to insulin. A number of factors are known to disrupt the normal response to insulin leading to the emergence of insulin resistance (IR). Advanced age and a high-fat diet are factors that increase the susceptibility to IR, with lipid accumulation in the skeletal muscle being a key driver of this phenomenon. It is debated, however, whether lipid accumulation arises due to dietary lipid overload or from a decline of mitochondrial function. To gain insights into the interplay of diet and age in the flexibility of muscle lipid and glucose handling, we combined lipidomics, proteomics, mitochondrial function analysis and computational modelling to investigate young and aged mice on a low- or high-fat diet (HFD). RESULTS: As expected, aged mice were more susceptible to IR when given a HFD than young mice. The HFD induced intramuscular lipid accumulation specifically in aged mice, including C18:0-containing ceramides and diacylglycerols. This was reflected by the mitochondrial ß-oxidation capacity, which was upregulated by the HFD in young, but not in old mice. Conspicuously, most ß-oxidation proteins were upregulated by the HFD in both groups, but carnitine palmitoyltransferase 1B (CPT1B) declined in aged animals. Computational modelling traced the flux control mostly to CPT1B, suggesting a CPT1B-driven loss of flexibility to the HFD with age. Finally, in old animals, glycolytic protein levels were reduced and less flexible to the diet. CONCLUSION: We conclude that intramuscular lipid accumulation and decreased insulin sensitivity are not due to age-related mitochondrial dysfunction or nutritional overload alone, but rather to their combined effects. Moreover, we identify CPT1B as a potential target to counteract age-dependent intramuscular lipid accumulation and thereby IR.


Asunto(s)
Resistencia a la Insulina , Animales , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Metabolismo de los Lípidos , Lípidos , Ratones , Músculo Esquelético/metabolismo
2.
Blood ; 130(13): 1523-1534, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28827409

RESUMEN

Endogenous DNA damage is causally associated with the functional decline and transformation of stem cells that characterize aging. DNA lesions that have escaped DNA repair can induce replication stress and genomic breaks that induce senescence and apoptosis. It is not clear how stem and proliferating cells cope with accumulating endogenous DNA lesions and how these ultimately affect the physiology of cells and tissues. Here we have addressed these questions by investigating the hematopoietic system of mice deficient for Rev1, a core factor in DNA translesion synthesis (TLS), the postreplicative bypass of damaged nucleotides. Rev1 hematopoietic stem and progenitor cells displayed compromised proliferation, and replication stress that could be rescued with an antioxidant. The additional disruption of Xpc, essential for global-genome nucleotide excision repair (ggNER) of helix-distorting nucleotide lesions, resulted in the perinatal loss of hematopoietic stem cells, progressive loss of bone marrow, and fatal aplastic anemia between 3 and 4 months of age. This was associated with replication stress, genomic breaks, DNA damage signaling, senescence, and apoptosis in bone marrow. Surprisingly, the collapse of the Rev1Xpc bone marrow was associated with progressive mitochondrial dysfunction and consequent exacerbation of oxidative stress. These data reveal that, to protect its genomic and functional integrity, the hematopoietic system critically depends on the combined activities of repair and replication of helix-distorting oxidative nucleotide lesions by ggNER and Rev1-dependent TLS, respectively. The error-prone nature of TLS may provide mechanistic understanding of the accumulation of mutations in the hematopoietic system upon aging.


Asunto(s)
Daño del ADN/genética , Reparación del ADN/genética , Sistema Hematopoyético/fisiología , Estrés Oxidativo , Animales , Apoptosis , Médula Ósea/patología , Proliferación Celular , Senescencia Celular/genética , ADN Polimerasa Dirigida por ADN , Genoma , Células Madre Hematopoyéticas/patología , Ratones , Nucleotidiltransferasas
3.
FASEB J ; 31(7): 3193-3204, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28396344

RESUMEN

The complement system, and specifically C5a, is involved in renal ischemia-reperfusion (IR) injury. The 2 receptors for complement anaphylatoxin C5a (C5aR1 and C5aR2) are expressed on leukocytes as well as on renal epithelium. Extensive evidence shows that C5aR1 inhibition protects kidneys from IR injury; however, the role of C5aR2 in IR injury is less clear as initial studies proposed the hypothesis that C5aR2 functions as a decoy receptor. By Using wild-type, C5aR1-/-, and C5aR2-/- mice in a model of renal IR injury, we found that a deficiency of either of these receptors protected mice from renal IR injury. Surprisingly, C5aR2-/- mice were most protected and had lower creatinine levels and reduced acute tubular necrosis. Next, an in vivo migration study demonstrated that leukocyte chemotaxis was unaffected in C5aR2-/- mice, whereas neutrophil activation was reduced by C5aR2 deficiency. To further investigate the contribution of renal cell-expressed C5aR2 vs leukocyte-expressed C5aR2 to renal IR injury, bone marrow chimeras were created. Our data show that both renal cell-expressed C5aR2 and leukocyte-expressed C5aR2 mediate IR-induced renal dysfunction. These studies reveal the importance of C5aR2 in renal IR injury. They further show that C5aR2 is a functional receptor, rather than a decoy receptor, and may provide a new target for intervention.-Poppelaars, F., van Werkhoven, M. B., Kotimaa, J., Veldhuis, Z. J., Ausema, A., Broeren, S. G. M., Damman, J., Hempel, J. C., Leuvenink, H. G. D., Daha, M. R., van Son, W. J., van Kooten, C., van Os, R. P., Hillebrands, J.-L., Seelen, M. A. Critical role for complement receptor C5aR2 in the pathogenesis of renal ischemia-reperfusion injury.


Asunto(s)
Enfermedades Renales/etiología , Receptor de Anafilatoxina C5a/metabolismo , Daño por Reperfusión/metabolismo , Animales , Movimiento Celular/fisiología , Regulación de la Expresión Génica , Leucocitos/fisiología , Ratones , Ratones Noqueados , Activación Neutrófila , Neutrófilos/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Anafilatoxina C5a/genética
4.
Am J Physiol Lung Cell Mol Physiol ; 308(1): L96-103, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25381025

RESUMEN

Anticholinergics, blocking the muscarinic M3 receptor, are effective bronchodilators for patients with chronic obstructive pulmonary disease. Recent evidence from M(3) receptor-deficient mice (M(3)R(-/-)) indicates that M3 receptors also regulate neutrophilic inflammation in response to cigarette smoke (CS). M(3) receptors are present on almost all cell types, and in this study we investigated the relative contribution of M(3) receptors on structural cells vs. inflammatory cells to CS-induced inflammation using bone marrow chimeric mice. Bone marrow chimeras (C56Bl/6 mice) were generated, and engraftment was confirmed after 10 wk. Thereafter, irradiated and nonirradiated control animals were exposed to CS or fresh air for four consecutive days. CS induced a significant increase in neutrophil numbers in nonirradiated and irradiated control animals (4- to 35-fold). Interestingly, wild-type animals receiving M(3)R(-/-) bone marrow showed a similar increase in neutrophil number (15-fold). In contrast, no increase in the number of neutrophils was observed in M3R(-/-) animals receiving wild-type bone marrow. The increase in keratinocyte-derived chemokine (KC) levels was similar in all smoke-exposed groups (2.5- to 5.0-fold). Microarray analysis revealed that fibrinogen-α and CD177, both involved in neutrophil migration, were downregulated in CS-exposed M(3)R(-/-) animals receiving wild-type bone marrow compared with CS-exposed wild-type animals, which was confirmed by RT-qPCR (1.6-2.5 fold). These findings indicate that the M(3) receptor on structural cells plays a proinflammatory role in CS-induced neutrophilic inflammation, whereas the M(3) receptor on inflammatory cells does not. This effect is probably not mediated via KC release, but may involve altered adhesion and transmigration of neutrophils via fibrinogen-α and CD177.


Asunto(s)
Infiltración Neutrófila , Neutrófilos/metabolismo , Receptor Muscarínico M3/metabolismo , Trastornos Respiratorios/metabolismo , Fumar/efectos adversos , Aloinjertos , Animales , Trasplante de Médula Ósea , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Regulación hacia Abajo/genética , Fibrinógeno/genética , Fibrinógeno/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Noqueados , Neutrófilos/patología , Receptor Muscarínico M3/genética , Trastornos Respiratorios/etiología , Trastornos Respiratorios/genética , Trastornos Respiratorios/patología , Fumar/genética , Fumar/metabolismo , Quimera por Trasplante/genética , Quimera por Trasplante/metabolismo
5.
FEBS J ; 288(7): 2257-2277, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33089625

RESUMEN

Dietary protein restriction has been demonstrated to improve metabolic health under various conditions. However, the relevance of ageing and age-related decline in metabolic flexibility on the effects of dietary protein restriction has not been addressed. Therefore, we investigated the effect of short-term dietary protein restriction on metabolic health in young and aged mice. Young adult (3 months old) and aged (18 months old) C57Bl/6J mice were subjected to a 3-month dietary protein restriction. Outcome parameters included fibroblast growth factor 21 (FGF21) levels, muscle strength, glucose tolerance, energy expenditure (EE) and transcriptomics of brown and white adipose tissue (WAT). Here, we report that a low-protein diet had beneficial effects in aged mice by reducing some aspects of age-related metabolic decline. These effects were characterized by increased plasma levels of FGF21, browning of subcutaneous WAT, increased body temperature and EE, while no changes were observed in glucose homeostasis and insulin sensitivity. Moreover, the low-protein diet used in this study was well-tolerated in aged mice indicated by the absence of adverse effects on body weight, locomotor activity and muscle performance. In conclusion, our study demonstrates that a short-term reduction in dietary protein intake can impact age-related metabolic health alongside increased FGF21 signalling, without negatively affecting muscle function. These findings highlight the potential of protein restriction as a strategy to induce EE and browning of WAT in aged individuals.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Metabolismo Energético/genética , Factores de Crecimiento de Fibroblastos/genética , Factores de Edad , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Restricción Calórica , Proteínas en la Dieta/metabolismo , Humanos , Ratones , Transducción de Señal
6.
Methods Mol Biol ; 430: 31-53, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18370290

RESUMEN

Animal models have added significantly to our understanding of the mechanism(s) of hematopoietic stem and progenitor cell (HSPC) mobilization. Such models suggest that changes in the interaction between the HSPC and the hematopoietic microenvironmental 'niche' (cellular and extracellular components) are critical to the process. The increasing availability of recombinant proteins (growth factors, cytokines, chemokines), antibodies, drugs (agonists and antagonists), and mutant and genetically modified animal models [gene knock-in (KI) and knock-out (KO)] continue to add to the tools available to better understand and manipulate mobilization processes.


Asunto(s)
Células Madre Hematopoyéticas/citología , Animales , Ratones , Ratones Transgénicos
7.
Methods Mol Biol ; 430: 143-57, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18370297

RESUMEN

Various assays exist that measure the function of hematopoietic stemcells (HSCs). In this chapter, in vitro assays are described that measure the frequency of progenitors (colony-forming unit in culture; CFU-C), stem cells (long-term culture-initiating cell; LTC-IC), or both (cobblestone area-forming cell assay; CAFC). These assays measure the potential of a test cell population retrospectively, i.e., at the time its activity is evident when the stem cell itself is often not detectable anymore. Although the in vitro LTC-IC and CAFC assays have been shown to correlate with in vivo activity, in vivo transplantation assays, where it can be shown that cells possess the ability to indefinitely repopulate all blood lineages, are the ultimate proof for HSC activity. Nevertheless, these in vitro assays provide an excellent method to screen for stem cell activity of a putative stem cell population or for screening the effect of a certain treatment on HSCs.


Asunto(s)
Células Madre Hematopoyéticas/citología , Línea Celular , Linaje de la Célula , Humanos , Técnicas In Vitro , Células del Estroma/citología
8.
Aging Cell ; 14(6): 1003-13, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26238505

RESUMEN

Microglia are a proliferative population of resident brain macrophages that under physiological conditions self-renew independent of hematopoiesis. Microglia are innate immune cells actively surveying the brain and are the earliest responders to injury. During aging, microglia elicit an enhanced innate immune response also referred to as 'priming'. To date, it remains unknown whether telomere shortening affects the proliferative capacity and induces priming of microglia. We addressed this issue using early (first-generation G1 mTerc(-/-) )- and late-generation (third-generation G3 and G4 mTerc(-/-) ) telomerase-deficient mice, which carry a homozygous deletion for the telomerase RNA component gene (mTerc). Late-generation mTerc(-/-) microglia show telomere shortening and decreased proliferation efficiency. Under physiological conditions, gene expression and functionality of G3 mTerc(-/-) microglia are comparable with microglia derived from G1 mTerc(-/-) mice despite changes in morphology. However, after intraperitoneal injection of bacterial lipopolysaccharide (LPS), G3 mTerc(-/-) microglia mice show an enhanced pro-inflammatory response. Nevertheless, this enhanced inflammatory response was not accompanied by an increased expression of genes known to be associated with age-associated microglia priming. The increased inflammatory response in microglia correlates closely with increased peripheral inflammation, a loss of blood-brain barrier integrity, and infiltration of immune cells in the brain parenchyma in this mouse model of telomere shortening.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Encéfalo/fisiopatología , Inflamación/inmunología , Microglía/inmunología , Acortamiento del Telómero/genética , Envejecimiento/inmunología , Animales , Encéfalo/citología , Proliferación Celular/genética , Modelos Animales de Enfermedad , Inflamación/patología , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Telomerasa/genética , Telómero/genética
9.
Radiother Oncol ; 108(3): 458-63, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23769181

RESUMEN

INTRODUCTION: During radiotherapy salivary glands of head and neck cancer patients are unavoidably co-irradiated, potentially resulting in life-long impairment. Recently we showed that transplantation of salisphere-derived c-Kit expressing cells can functionally regenerate irradiated salivary glands. This study aims to select a more potent subpopulation of c-Kit(+) cells, co-expressing stem cell markers and to investigate whether long-term tissue homeostasis is restored after stem cell transplantation. METHODS AND RESULTS: Salisphere derived c-Kit(+) cells that co-expressed CD24 and/or CD49f markers, were intra-glandularly injected into 15 Gy irradiated submandibular glands of mice. Particularly, c-Kit(+)/CD24(+)/CD49f(+) cell transplanted mice improved saliva production (54.59 ± 11.1%) versus the irradiated control group (21.5 ± 8.7%). Increase in expression of cells with differentiated duct cell markers like, cytokeratins (CK8, 18, 7 and 14) indicated functional recovery of this compartment. Moreover, ductal stem cell marker expression like c-Kit, CD133, CD24 and CD49f reappeared after transplantation indicating long-term functional maintenance potential of the gland. Furthermore, a normalization of vascularization as indicated by CD31 expression and reduction of fibrosis was observed, indicative of normalization of the microenvironment. CONCLUSIONS: Our results show that stem cell transplantation not only rescues hypo-salivation, but also restores tissue homeostasis of the irradiated gland, necessary for long-term maintenance of adult tissue.


Asunto(s)
Proteínas Proto-Oncogénicas c-kit/fisiología , Traumatismos por Radiación/terapia , Glándulas Salivales/efectos de la radiación , Trasplante de Células Madre , Animales , Antígeno CD24/análisis , Femenino , Neoplasias de Cabeza y Cuello/radioterapia , Homeostasis , Integrina alfa6/análisis , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-kit/análisis , Regeneración , Glándulas Salivales/fisiología
10.
Radiother Oncol ; 107(3): 434-41, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23684587

RESUMEN

BACKGROUND AND PURPOSE: In this study, we investigated whether cancer stem cell marker expressing cells can be identified that predict for the response of esophageal cancer (EC) to CRT. MATERIALS AND METHODS: EC cell-lines OE-33 and OE-21 were used to assess in vitro, stem cell activity, proliferative capacity and radiation response. Xenograft tumors were generated using NOD/SCID mice to assess in vivo proliferative capacity and tumor hypoxia. Archival and fresh EC biopsy tissue was used to confirm our in vitro and in vivo results. RESULTS: We showed that the CD44+/CD24- subpopulation of EC cells exerts a higher proliferation rate and sphere forming potential and is more radioresistant in vitro, when compared to unselected or CD44+/CD24+ cells. Moreover, CD44+/CD24- cells formed xenograft tumors faster and were often located in hypoxic tumor areas. In a study of archival pre-neoadjuvant CRT biopsy material from EC adenocarcinoma patients (N=27), this population could only be identified in 50% (9/18) of reduced-responders to neoadjuvant CRT, but never (0/9) in the complete responders (P=0.009). CONCLUSION: These results warrant further investigation into the possible clinical benefit of CD44+/CD24- as a predictive marker in EC patients for the response to chemoradiation.


Asunto(s)
Adenocarcinoma/terapia , Antígeno CD24/análisis , Quimioradioterapia , Neoplasias Esofágicas/terapia , Receptores de Hialuranos/análisis , Células Madre Neoplásicas/patología , Adenocarcinoma/química , Adenocarcinoma/patología , Animales , Biomarcadores , Línea Celular Tumoral , Proliferación Celular , Neoplasias Esofágicas/química , Neoplasias Esofágicas/patología , Humanos , Ratones , Ratones SCID
11.
Radiother Oncol ; 99(3): 367-72, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21719134

RESUMEN

BACKGROUND: Stem cell therapy could be a potential way for reducing radiation-induced hyposalivation and improving the patient's quality of life. However, the identification and purification of salivary gland stem cells have not been accomplished. This study aims to better characterize the stem/progenitor cell population with regenerative potential residing in the mouse salivary gland. METHODS: Mouse submandibular gland tissue, isolated cells and cultured 3 day old salispheres were tested for their expression of stem cell markers c-Kit, CD133, CD49f, and CD24 using immunohistochemistry for tissue and flow cytometry for cells. Mice were locally irradiated with a single dose of 15 Gy and transplanted with cells expressing defined markers. RESULTS: Cells expressing known stem cell markers are localized in the larger ducts of the mouse salivary gland. Isolated cells and cells from day 3 salispheres also express these markers: c-Kit (0.058% vs. 0.65%), CD133 (6% vs. 5%), CD49f (78% vs. 51%), and CD24 (60% vs. 60%, respectively). Intraglandular transplantation of these cells into irradiated salivary glands of mice resulted in stem cell marker-specific recovery of salivary gland function. CONCLUSIONS: Different stem cell-associated markers are expressed in mouse salivary gland cells, which upon transplantation are able to regenerate the irradiation damaged salivary gland.


Asunto(s)
Glándulas Salivales/citología , Glándulas Salivales/efectos de la radiación , Trasplante de Células Madre/métodos , Células Madre/citología , Células Madre/efectos de la radiación , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Antígeno CD24/metabolismo , Femenino , Citometría de Flujo , Glicoproteínas/metabolismo , Neoplasias de Cabeza y Cuello/radioterapia , Técnicas para Inmunoenzimas , Integrina alfa6/metabolismo , Ratones , Ratones Endogámicos C57BL , Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Regeneración , Células Madre/metabolismo , Xerostomía/etiología , Xerostomía/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA