Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell Biochem ; 477(8): 2059-2071, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35449483

RESUMEN

Non-alcoholic steatohepatitis (NASH) is a rapidly growing liver disease. The chemoattractant chemerin is abundant in hepatocytes, and hepatocyte expressed prochemerin protected from NASH. Prochemerin is inactive and different active isoforms have been described. Here, the effect of hepatocyte expressed muChem-156, a highly active murine chemerin isoform, was studied in the methionine-choline deficient dietary model of NASH. Mice overexpressing muChem-156 had higher hepatic chemerin protein. Serum chemerin levels and the capability of serum to activate the chemerin receptors was unchanged showing that the liver did not release active chemerin. Notably, activation of the chemerin receptors by hepatic vein blood did not increase in parallel to total chemerin protein in patients with liver cirrhosis. In experimental NASH, muChem-156 had no effect on liver lipids. Accordingly, overexpression of active chemerin in hepatocytes or treatment of hepatocytes with recombinant chemerin did not affect cellular triglyceride and cholesterol levels. Importantly, overexpression of muChem-156 in the murine liver did not change the hepatic expression of inflammatory and profibrotic genes. The downstream targets of chemerin such as p38 kinase were neither activated in the liver of muChem-156 producing mice nor in HepG2, Huh7 and Hepa1-6 cells overexpressing this isoform. Recombinant chemerin had no effect on global gene expression of primary human hepatocytes and hepatic stellate cells within 24 h of incubation. Phosphorylation of p38 kinase was, however, increased upon short-time incubation of HepG2 cells with chemerin. These findings show that muChem-156 overexpression in hepatocytes does not protect from liver steatosis and inflammation.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Quimiocinas , Modelos Animales de Enfermedad , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Hígado/metabolismo , Ratones , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Isoformas de Proteínas/metabolismo
2.
Int J Mol Sci ; 21(7)2020 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-32224846

RESUMEN

: The coordinated development and function of bone-forming (osteoblasts) and bone-resorbing (osteoclasts) cells is critical for the maintenance of skeletal integrity and calcium homeostasis. An enhanced adipogenic versus osteogenic potential of bone marrow mesenchymal stem cells (MSCs) has been linked to bone loss associated with diseases such as diabetes mellitus, as well as aging and postmenopause. In addition to an inherent decrease in bone formation due to reduced osteoblast numbers, recent experimental evidence indicates that an increase in bone marrow adipocytes contributes to a disproportionate increase in osteoclast formation. Therefore, a potential strategy for therapeutic intervention in chronic bone loss disorders such as osteoporosis is to interfere with the pro-osteoclastogenic influence of marrow adipocytes. However, application of this approach is limited by the extremely complex regulatory processes in the osteoclastogenic program. For example, key regulators of osteoclastogenesis such as the receptor activator of nuclear factor-kappaB ligand (RANKL) and the soluble decoy receptor osteoprotegerin (OPG) are not only secreted by both osteoblasts and adipocytes, but are also regulated through several cytokines produced by these cell types. In this context, biologically active signaling molecules secreted from bone marrow adipocytes, such as chemerin, adiponectin, leptin, visfatin and resistin, can have a profound influence on the osteoclast differentiation program of hematopoietic stem cells (HSCs), and thus, hold therapeutic potential under disease conditions. In addition to these paracrine signals, adipogenic transcription factors including CCAAT/enhancer binding protein alpha (C/EBPα), C/EBP beta (C/EBPß) and peroxisome proliferator-associated receptor gamma (PPARγ) are also expressed by osteoclastogenic cells. However, in contrast to MSCs, activation of these adipogenic transcription factors in HSCs promotes the differentiation of osteoclast precursors into mature osteoclasts. Herein, we discuss the molecular mechanisms that link adipogenic signaling molecules and transcription factors to the osteoclast differentiation program and highlight therapeutic strategies targeting these mechanisms for promoting bone homeostasis.


Asunto(s)
Adipocitos/citología , Comunicación Celular , Diferenciación Celular , Osteoclastos/citología , Adipocitos/metabolismo , Animales , Humanos , Osteoclastos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Int J Mol Sci ; 21(19)2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-33003572

RESUMEN

Murine chemerin is C-terminally processed to the bioactive isoforms, muChem-156 and muChem-155, among which the longer variant protects from hepatocellular carcinoma (HCC). However, the role of muChem-155 is mostly unknown. Here, we aimed to compare the effects of these isoforms on the proliferation, migration and the secretome of the human hepatocyte cell lines HepG2 and Huh7 and the murine Hepa1-6 cell line. Therefore, huChem-157 and -156 were overexpressed in the human cells, and the respective murine variants, muChem-156 and -155, in the murine hepatocytes. Both chemerin isoforms produced by HepG2 and Hepa1-6 cells activated the chemerin receptors chemokine-like receptor 1 (CMKLR1) and G protein-coupled receptor 1 (GPR1). HuChem-157 was the active isoform in the Huh7 cell culture medium. The potencies of muChem-155 and muChem-156 to activate human GPR1 and mouse CMKLR1 were equivalent. Human CMKLR1 was most responsive to muChem-156. Chemerin variants showed no effect on cell viability and proliferation. Activation of the mitogen-activated protein kinases Erk1/2 and p38, and protein levels of the epithelial-mesenchymal transition marker, E-cadherin, were not regulated by the chemerin variants. Migration was reduced in HepG2 and Hepa1-6 cells by the longer isoform. Protective effects of chemerin in HCC include the modulation of cytokines but huChem-156 and huChem-157 overexpression did not change IL-8, CCL20 or osteopontin in the hepatocytes. The conditioned medium of the transfected hepatocytes failed to alter these soluble factors in the cell culture medium of peripheral blood mononuclear cells (PBMCs). Interestingly, the cell culture medium of Huh7 cells producing the inactive variant huChem-155 reduced CCL2 and IL-8 in PBMCs. To sum up, huChem-157 and muChem-156 inhibited hepatocyte migration and may protect from HCC metastasis. HuChem-155 was the only human isoform exerting anti-inflammatory effects on immune cells.


Asunto(s)
Quimiocinas/genética , Inflamación/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Receptores de Quimiocina/genética , Receptores Acoplados a Proteínas G/genética , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inflamación/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Isoformas de Proteínas/genética
4.
Int J Mol Sci ; 21(20)2020 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-33066326

RESUMEN

The chemokine chemerin exists as C-terminally processed isoforms whose biological functions are mostly unknown. A highly active human chemerin variant (huChem-157) was protective in experimental hepatocellular carcinoma (HCC) models. Hepatic stellate cells (HSCs) are central mediators of hepatic fibrogenesis and carcinogenesis and express the chemerin receptors chemokine-like receptor 1 (CMKLR1) and G protein-coupled receptor 1 (GPR1). Here we aimed to analyse the effect of chemerin isoforms on the viability, proliferation and secretome of the human HSC cell line LX-2. Therefore, huChem-157, 156 and 155 were over-expressed in LX-2 cells, which have low endogenous chemerin levels. HuChem-157 produced in LX-2 cells activated CMKLR1 and GPR1, and huChem-156 modestly induced GPR1 signaling. HuChem-155 is an inactive chemerin variant. Chemerin isoforms had no effect on cell viability and proliferation. Cellular expression of the fibrotic proteins galectin-3 and alpha-smooth muscle actin was not regulated by any chemerin isoform. HuChem-156 increased IL-6, IL-8 and galectin-3 in cell media. HuChem-157 was ineffective, and accordingly, did not enhance levels of these proteins in media of primary human hepatic stellate cells when added exogenously. These analyses provide evidence that huChem-156 is the biologic active chemerin variant in hepatic stellate cells and acts as a pro-inflammatory factor.


Asunto(s)
Quimiocinas/metabolismo , Células Estrelladas Hepáticas/metabolismo , Actinas/metabolismo , Línea Celular , Proliferación Celular , Células Cultivadas , Quimiocinas/genética , Galectina 3/metabolismo , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Quimiocina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
5.
FASEB J ; : fj201800479, 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29906243

RESUMEN

Measures of the adipokine chemerin are elevated in multiple cardiovascular diseases, including hypertension, but little mechanistic work has been done to implicate chemerin as being causative in such diseases. The chemerin knockout (KO) rat was created to test the hypothesis that removal of chemerin would reduce pressure in the normal and hypertensive state. Western analyses confirmed loss of chemerin in the plasma and tissues of the KO vs. wild-type (WT) rats. Chemerin concentration in plasma and tissues was lower in WT females than in WT males, as determined by Western analysis. Conscious male and female KO rats had modest differences in baseline measures vs. the WT that included systolic, diastolic, mean arterial and pulse pressures, and heart rate, all measured telemetrically. The mineralocorticoid deoxycorticosterone acetate (DOCA) and salt water, combined with uninephrectomy as a hypertensive stimulus, elevated mean and systolic blood pressures of the male KO higher than the male WT. By contrast, all pressures in the female KO were lower than their WT throughout DOCA-salt treatment. These results revealed an unexpected sex difference in chemerin expression and the ability of chemerin to modify blood pressure in response to a hypertensive challenge.-Watts, S. W., Darios, E. S., Mullick, A. E., Garver, H., Saunders, T. L., Hughes, E. D., Filipiak, W. E., Zeidler, M. G., McMullen, N., Sinal, C. J., Kumar, R. K., Ferland, D. J., Fink, G. D. The chemerin knockout rat reveals chemerin dependence in female, but not male, experimental hypertension.

6.
Int J Mol Sci ; 20(19)2019 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-31561459

RESUMEN

Chemerin is widely recognized as an adipokine, with diverse biological roles in cellular differentiation and metabolism, as well as a leukocyte chemoattractant. Research investigating the role of chemerin in the obesity-cancer relationship has provided evidence both for pro- and anti-cancer effects. The tumor-promoting effects of chemerin primarily involve direct effects on migration, invasion, and metastasis as well as growth and proliferation of cancer cells. Chemerin can also promote tumor growth via the recruitment of tumor-supporting mesenchymal stromal cells and stimulation of angiogenesis pathways in endothelial cells. In contrast, the majority of evidence supports that the tumor-suppressing effects of chemerin are immune-mediated and result in a shift from immunosuppressive to immunogenic cell populations within the tumor microenvironment. Systemic chemerin and chemerin produced within the tumor microenvironment may contribute to these effects via signaling through CMKLR1 (chemerin1), GPR1 (chemerin2), and CCLR2 on target cells. As such, inhibition or activation of chemerin signaling could be beneficial as a therapeutic approach depending on the type of cancer. Additional studies are required to determine if obesity influences cancer initiation or progression through increased adipose tissue production of chemerin and/or altered chemerin processing that leads to changes in chemerin signaling in the tumor microenvironment.


Asunto(s)
Adipoquinas/metabolismo , Quimiocinas/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Transducción de Señal , Adipoquinas/genética , Animales , Biomarcadores , Susceptibilidad a Enfermedades , Humanos , Inmunomodulación , Neoplasias/patología , Obesidad/complicaciones , Obesidad/metabolismo , Especificidad de Órganos , Unión Proteica
7.
Int J Mol Sci ; 21(1)2019 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-31905933

RESUMEN

The tumor inhibitory potential of the highly active chemerin-156 isoform was described in orthotopic models of hepatocellular carcinoma (HCC). The majority of HCC arises in the fibrotic liver, which was not reproduced in these studies. Here, a potential therapeutic activity of chemerin-156 was evaluated in diethylnitrosamine (DEN)-induced liver cancer, which mimics fibrosis-associated HCC. Mice were infected with adeno-associated virus (AAV) six months after DEN injection to overexpress chemerin-156 in the liver, and animals injected with non-recombinant-AAV served as controls. Three months later, the animals were killed. Both groups were comparable with regard to liver steatosis and fibrosis. Of note, the number of very small tumors was reduced by chemerin-156. Anyhow, the expression of inflammatory and profibrotic genes was similar in larger tumors of control and chemerin-156-AAV-infected animals. Although genes with a role in lipid metabolism, like 3-hydroxy-3-methylglutaryl-coenzym-A--reductase, were overexpressed in tumors of animals with high chemerin-156, total hepatic cholesterol, diacylglycerol and triglyceride levels, and distribution of individual lipid species were normal. Chemerin-156-AAV-infected mice had elevated hepatic and systemic chemerin. Ex vivo activation of the chemerin receptor chemokine-like receptor 1 increased in parallel with serum chemerin, illustrating the biological activity of the recombinant protein. In the tumors, chemerin-155 was the most abundant variant. Chemerin-156 was not detected in tumors of the controls and was hardly found in chemerin-156-AAV infected animals. In conclusion, the present study showed that chemerin-156 overexpression caused a decline in the number of small lesions but did not prevent the growth of pre-existing neoplasms.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Quimiocinas/metabolismo , Dietilnitrosamina/efectos adversos , Hepatocitos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Carga Tumoral/fisiología , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Quimiocinas/sangre , Quimiocinas/genética , Colesterol/metabolismo , Diglicéridos/metabolismo , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/genética , Metabolismo de los Lípidos , Hígado/lesiones , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Endogámicos C3H , Isoformas de Proteínas , Receptores de Quimiocina , Triglicéridos/metabolismo
8.
Cytokine ; 104: 42-45, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29414326

RESUMEN

OBJECTIVES: Chemerin is an adipokine with established roles in inflammation, adipogenesis and the regulation of glucose and lipid homeostasis. Extracellular proteolytic processing of chemerin generates a spectrum of isoforms that differ significantly with respect to the ability to activate the cognate receptors chemokine-like receptor 1 (CMKLR1) and G-protein-coupled receptor 1 (GPR1). Increased total serum chemerin has been widely reported in obese humans as well as in preclinical rodent models of adiposity. However, very little information is available regarding the correspondence, if any, of changes in total serum chemerin protein with chemerin bioactivity. METHODS: Total serum chemerin and ex vivo CMKLR1 and GPR1 activation was compared using two widely used murine obesity models: high fat diet feeding (HFD) and leptin deficiency (ob/ob). RESULTS: Total serum chemerin levels and ex vivo CMKLR1 and GPR1 activation were significantly induced in HFD. The bioactivity ratio (bioactive chemerin/total chemerin) was also increased when measured with CMKLR1, but not GPR1. In contrast, while ob/ob mice exhibited increased total serum chemerin protein, ex vivo receptor activation was observed with GPR1, but not CMKLR1. There was no change in bioactivity ratio for either receptor. Of note, GPR1 but not CMKLR1 bioactivity positively correlated with adipose tissue inflammation. CONCLUSIONS: While increased total serum chemerin is a consistent finding among rodent obesity models, this may not accurately reflect changes in chemerin bioactivity which is the major determinant of biological effects.


Asunto(s)
Quimiocinas/sangre , Péptidos y Proteínas de Señalización Intercelular/sangre , Obesidad/sangre , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Leptina/deficiencia , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Receptores de Quimiocina , Receptores Acoplados a Proteínas G/metabolismo
9.
Stem Cells ; 35(3): 711-724, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27733019

RESUMEN

Bone remodeling is a dynamic process requiring the coordinated action of formative (osteoblast) and resorptive (osteoclast) cell populations. An imbalance of the development and function of these cell types underlies several chronic bone loss disorders such as osteoporosis. Increased bone marrow adipocyte numbers commonly occur with bone loss disorders and numerous studies have documented an inverse relationship between bone marrow fat and bone formation. Osteoblasts and adipocytes derive in a competitive fashion from a common mesenchymal stem cell (MSC) precursor. Generally, factors that promote MSC adipogenesis inhibit osteoblastogenesis and thereby, reduce bone formation. Previously we established that the secreted protein chemerin regulates adipogenic and osteoblastogenic differentiation of MSCs by signaling through chemokine-like receptor 1 (CMKLR1). However, the fundamental mechanisms by which chemerin/CMKLR1 influences lineage determination remain largely uncharacterized. Herein, we provide experimental evidence that chemerin/CMKLR1 regulates canonical Wnt signaling in MSCs by influencing the expression, subcellular location, and transcriptional activity of the central Wnt transducer, ß-catenin. Moreover, we provide evidence that CMKLR1 is a novel Wnt responsive gene that functions in a negative feedback loop to limit osteoblastogenic Wnt signaling. Mechanistically, this entails Notch-dependent changes in the expression and function of key adipogenic and osteoblastogenic transcription factors, cell cycle proteins and chromatin remodeling enzymes. Consistent with this, MSCs from CMKLR1 knockout (-/-) mice exhibited similar dependency on Notch signaling to maintain osteoblastogenic differentiation. Taken together, our findings support a fundamental biological function for chemerin/CMKLR1 to balance osteoblastogenic and adipogenic signaling and thereby contribute to the maintenance of pluripotency in MSCs. Stem Cells 2017;35:711-724.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Receptores Acoplados a Proteínas G/genética , Vía de Señalización Wnt/genética , Adipogénesis/genética , Animales , Diferenciación Celular/genética , Quimiocinas/metabolismo , Ciclina D1/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Osteogénesis/genética , PPAR gamma/metabolismo , Receptores de Quimiocina , Receptores Notch/metabolismo , Proteínas Represoras/metabolismo , Ubiquitinación , beta Catenina/metabolismo
10.
Curr Osteoporos Rep ; 16(4): 434-442, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29855795

RESUMEN

PURPOSE OF REVIEW: To summarize and discuss recent progress and novel signaling mechanisms relevant to bone marrow adipocyte formation and its physiological/pathophysiological implications for bone remodeling. RECENT FINDINGS: Skeletal remodeling is a coordinated process entailing removal of old bone and formation of new bone. Several bone loss disorders such as osteoporosis are commonly associated with increased bone marrow adipose tissue. Experimental and clinical evidence supports that a reduction in osteoblastogenesis from mesenchymal stem cells at the expense of adipogenesis, as well as the deleterious effects of adipocyte-derived signaling, contributes to the etiology of osteoporosis as well as bone loss associated with aging, diabetes mellitus, post-menopause, and chronic drug therapy. However, this view is challenged by findings indicating that, in some contexts, bone marrow adipose tissue may have a beneficial impact on skeletal health. Further research is needed to better define the role of marrow adipocytes in bone physiology/pathophysiology and to determine the therapeutic potential of manipulating mesenchymal stem cell differentiation.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Médula Ósea/metabolismo , Remodelación Ósea , Osteoblastos/metabolismo , Osteogénesis , Osteoporosis/metabolismo , Adipocitos/citología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/patología , Médula Ósea/patología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Humanos , Células Madre Mesenquimatosas/citología , Osteoblastos/citología , Osteoporosis/patología , Transducción de Señal
11.
Biochim Biophys Acta ; 1861(3): 260-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26721419

RESUMEN

Chemerin is a novel adipokine associated with obesity and insulin resistance. α-Lipoic acid (α-LA) has shown beneficial properties on diabetes and obesity. The aim of this study was to examine the effects of α-LA on chemerin production in adipocytes in absence or presence of TNF-α, insulin and AICAR. The potential signaling pathways involved in α-LA effects on chemerin were also analyzed. α-LA actions on chemerin were tested in differentiated 3T3-L1 adipocytes and in some cases in human subcutaneous and omental adipocytes. Chemerin mRNA levels were measured by RT-PCR and the amount of chemerin secreted to culture media was determined by ELISA. α-LA induced a concentration-dependent inhibition on both chemerin secretion and mRNA levels in 3T3-L1 adipocytes. The AMPK activator AICAR and the PI3K inhibitor LY294002 dramatically abrogated both chemerin secretion and gene expression, and further potentiated the inhibitory effect of α-LA on chemerin secretion. Insulin was able to partially reverse the inhibitory action of α-LA on chemerin secretion. α-LA also reduced basal chemerin secretion in both subcutaneous and omental adipocytes from overweight/obese subjects. Moreover, α-LA was able to abolish the stimulatory effects of the pro-inflammatory cytokine TNF-α on chemerin secretion. Our data demonstrated the ability of α-LA to inhibit chemerin production, an adipokine associated to obesity and metabolic syndrome, suggesting that the reduction of chemerin could contribute to the antiobesity/antidiabetic properties described for α-LA.


Asunto(s)
Adipocitos/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Quimiocinas/metabolismo , Hipoglucemiantes/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ácido Tióctico/farmacología , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Quimiocinas/sangre , Quimiocinas/genética , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Humanos , Insulina/farmacología , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , PPAR gamma/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Ratas Wistar , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos , Transfección , Factor de Necrosis Tumoral alfa/farmacología
12.
IUBMB Life ; 66(3): 147-155, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24638917

RESUMEN

Throughout life, bone is constantly remodeled through the complementary processes of bone resorption and bone formation. Highly coordinated regulation of these activities is essential for maintaining consistent bone quality and quantity. Normally, the development and function of bone-forming (osteoblast) and bone-resorbing (osteoclast) cells are tightly regulated by signaling molecules secreted by these two cell types. Within the bone marrow microenvironment, osteoblasts arise from mesenchymal stem cells (MSCs), which are in close contact with the hematopoietic stem cell (HSC) precursors that differentiate into mature osteoclasts. Signaling molecules secreted by osteoblasts (e.g., receptor activator of nuclear factor kappa B ligand and osteoprotegerin) and osteoclasts (e.g., bone morphogenetic protein 6, wingless-type MMTV integration site family member 10B, sphingosine-1-phosphate, and ephrin-B2) play a key role in bone remodeling by guiding the differentiation, localization, and function of bone cells. In addition to osteoblasts, bone marrow MSCs can also differentiate into adipocytes that affect bone remodeling by competitively suppressing intracellular osteogenic signals, including runt-related transcription factor 2, osterix, and beta-catenin, while simultaneously promoting the secretion of adipogenic signaling molecules such as leptin, adiponectin, chemerin, omentin-1, resistin, and visfatin. Secreted adipogenic factors have also been shown to affect the osteoclastogenic differentiation of HSCs. Herein, we discuss the impact of bone marrow adipocytes on the coupling of osteoblast and osteoclast differentiation, and the relevance to bone-loss disorders such as osteoporosis. © 2014 IUBMB Life, 66(3):147-155, 2014.

13.
Stem Cells ; 31(10): 2172-82, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23766088

RESUMEN

Bone is a dynamic tissue that is continuously remodeled through the action of formative osteoblasts and resorptive osteoclasts. Chemerin is a secreted protein that activates chemokine-like receptor 1 (CMKLR1), a G protein-coupled receptor expressed by various cell types including adipocytes, osteoblasts, mesenchymal stem cells (MSCs), and macrophages. Previously, we identified chemerin as a regulator of adipocyte and osteoblast differentiation of MSCs. Herein we examined the role of chemerin in Lin(-) Sca1(+) c-kit(+) CD34(+) hematopoietic stem cell (HSC) osteoclastogenesis. We found that HSCs expressed both chemerin and CMKLR1 mRNA and secreted chemerin protein into the extracellular media. Neutralization of chemerin with a blocking antibody beginning prior to inducing osteoclast differentiation resulted in a near complete loss of osteoclastogenesis as evidenced by reduced marker gene expression and matrix resorption. This effect was conserved in an independent model of RAW264.7 cell osteoclastogenesis. Reintroduction of chemerin by reversal of neutralization rescued osteoclast differentiation indicating that chemerin signaling is essential to permit HSC differentiation into osteoclasts but following blockade the cells maintained the potential to differentiate into osteoclasts. Mechanistically, neutralization of chemerin blunted the early receptor activator of nuclear factor-kappa B ligand induction of nuclear factor of activated T-cells 2 (NFAT2), Fos, Itgb3, and Src associated with preosteoclast formation. Consistent with a central role for NFAT2, induction or activation of NFAT2 by forced expression or stimulation of intracellular calcium release rescued the impairment of HSC osteoclastogenesis caused by chemerin neutralization. Taken together, these data support a novel autocrine/paracrine role for chemerin in regulating osteoclast differentiation of HSCs through modulating intracellular calcium and NFAT2 expression/activation.


Asunto(s)
Diferenciación Celular , Factores Quimiotácticos/fisiología , Células Madre Hematopoyéticas/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Animales , Anticuerpos/farmacología , Línea Celular , Quimiocinas , Factores Quimiotácticos/antagonistas & inhibidores , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Osteoclastos/metabolismo , Ligando RANK/fisiología , Receptores de Quimiocina , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
14.
Arterioscler Thromb Vasc Biol ; 33(6): 1320-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23559624

RESUMEN

OBJECTIVE: Obesity and hypertension are comorbid in epidemic proportion, yet their biological connection is largely a mystery. The peptide chemerin is a candidate for connecting fat deposits around the blood vessel (perivascular adipose tissue) to arterial contraction. We presently tested the hypothesis that chemerin is expressed in perivascular adipose tissue and is vasoactive, supporting the existence of a chemerin axis in the vasculature. APPROACH AND RESULTS: Real-time polymerase chain reaction, immunohistochemistry, and Western analyses supported the synthesis and expression of chemerin in perivascular adipose tissue, whereas the primary chemerin receptor ChemR23 was expressed both in the tunica media and endothelial layer. The ChemR23 agonist chemerin-9 caused receptor, concentration-dependent contraction in the isolated rat thoracic aorta, superior mesenteric artery, and mesenteric resistance artery, and contraction was significantly amplified (more than 100%) when nitric oxide synthase was inhibited and the endothelial cell mechanically removed or tone was placed on the arteries. The novel ChemR23 antagonist CCX832 inhibited phenylephrine-induced and prostaglandin F2α-induced contraction (+perivascular adipose tissue), suggesting that endogenous chemerin contributes to contraction. Arteries from animals with dysfunctional endothelium (obese or hypertensive) demonstrated a pronounced contraction to chemerin-9. Finally, mesenteric arteries from obese humans demonstrate amplified contraction to chemerin-9. CONCLUSIONS: These data support a new role for chemerin as an endogenous vasoconstrictor that operates through a receptor typically attributed to function only in immune cells.


Asunto(s)
Adipoquinas/metabolismo , Tejido Adiposo/metabolismo , Aorta Torácica/metabolismo , Quimiocinas/metabolismo , Arterias Mesentéricas/metabolismo , Músculo Liso Vascular/fisiología , Vasoconstricción/fisiología , Tejido Adiposo/efectos de los fármacos , Angiotensina II/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Western Blotting , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Hipertensión/metabolismo , Inmunoquímica , Péptidos y Proteínas de Señalización Intercelular , Arterias Mesentéricas/efectos de los fármacos , Obesidad/metabolismo , Fenilefrina/farmacología , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Sensibilidad y Especificidad
15.
Am J Physiol Cell Physiol ; 302(11): C1621-31, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22460713

RESUMEN

The chemokine-like receptor-1 (CMKLR1) is a G protein-coupled receptor that is activated by chemerin, a secreted plasma leukocyte attractant and adipokine. Previous studies identified that CMKLR1 is expressed in skeletal muscle in a stage-specific fashion during embryogenesis and in adult mice; however, its function in skeletal muscle remains unclear. Based on the established function of CMKLR1 in cell migration and differentiation, we investigated the hypothesis that CMKLR1 regulates the differentiation of myoblasts into myotubes. In C(2)C(12) mouse myoblasts, CMKLR1 expression increased threefold with differentiation into multinucleated myotubes. Decreasing CMKLR1 expression by adenoviral-delivered small-hairpin RNA (shRNA) impaired the differentiation of C(2)C(12) myoblasts into mature myotubes and reduced the mRNA expression of myogenic regulatory factors myogenin and MyoD while increasing Myf5 and Mrf4. At embryonic day 12.5 (E12.5), CMKLR1 knockout (CMKLR1(-/-)) mice appeared developmentally delayed and displayed significantly lower wet weights and a considerably diminished myotomal component of somites as revealed by immunolocalization of myosin heavy chain protein compared with wild-type (CMKLR1(+/+)) mouse embryos. These changes were associated with increased Myf5 and decreased MyoD protein expression in the somites of E12.5 CMKLR1(-/-) mouse embryos. Adult male CMKLR1(-/-) mice had significantly reduced bone-free lean mass and weighed less than the CMKLR1(+/+) mice. We conclude that CMKLR1 is essential for myogenic differentiation of C(2)C(12) cells in vitro, and the CMKLR1 null mice have a subtle skeletal muscle deficit beginning from embryonic life that persists during postnatal life.


Asunto(s)
Células Musculares/metabolismo , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Absorciometría de Fotón , Animales , Diferenciación Celular , Células Cultivadas , Masculino , Ratones , Ratones Noqueados , Células Musculares/fisiología , Fibras Musculares Esqueléticas/fisiología , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Músculo Esquelético/embriología , Músculo Esquelético/fisiología , Proteína MioD/biosíntesis , Proteína MioD/genética , Factor 5 Regulador Miogénico/biosíntesis , Factor 5 Regulador Miogénico/genética , Factor 5 Regulador Miogénico/metabolismo , Factores Reguladores Miogénicos/biosíntesis , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Receptores de Quimiocina
16.
J Biol Chem ; 286(27): 23982-95, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21572083

RESUMEN

Chemerin is an adipocyte-secreted protein that regulates adipogenesis and the metabolic function of mature adipocytes via activation of chemokine-like receptor 1 (CMKLR1). Herein we report the interaction of peroxisome proliferator-activated receptor γ (PPARγ) and chemerin in the context of adipogenesis. Knockdown of chemerin or CMKLR1 expression or antibody neutralization of secreted chemerin protein arrested adipogenic clonal expansion of bone marrow mesenchymal stem cells (BMSCs) by inducing a loss of G(2)/M cyclins (cyclin A2/B2) but not the G(1)/S cyclin D2. Forced expression of PPARγ in BMSCs did not completely rescue this loss of clonal expansion and adipogenesis following chemerin or CMKLR1 knockdown. However, forced expression and/or activation of PPARγ in BMSCs as well as non-adipogenic cell types such as NIH-3T3 embryonic fibroblasts and MCA38 colon carcinoma cells significantly induced chemerin expression and secretion. Sequence analysis revealed a putative PPARγ response element (PPRE) sequence within the chemerin promoter. This PPRE was able to confer PPARγ responsiveness on a heterologous promoter, and mutation of this sequence abolished activation of the chemerin promoter by PPARγ. Chromatin immunoprecipitation confirmed the direct association of PPARγ with this PPRE. Treatment of mice with rosiglitazone elevated chemerin mRNA levels in adipose tissue and bone marrow coincident with an increase in circulating chemerin levels. Together, these findings support a fundamental role for chemerin/CMKLR1 signaling in clonal expansion during adipocyte differentiation as well as a role for PPARγ in regulating chemerin expression.


Asunto(s)
Adipogénesis/fisiología , Factores Quimiotácticos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , PPAR gamma/metabolismo , Animales , Ciclo Celular , Quimiocinas , Factores Quimiotácticos/genética , Técnicas de Silenciamiento del Gen , Péptidos y Proteínas de Señalización Intercelular/genética , Células Madre Mesenquimatosas/citología , Ratones , Células 3T3 NIH , PPAR gamma/genética , Receptores de Quimiocina , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Elementos de Respuesta/fisiología
17.
Front Endocrinol (Lausanne) ; 13: 903925, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35903271

RESUMEN

When compared to adipocytes in other anatomical sites, the interaction of bone marrow resident adipocytes with the other cells in their microenvironment is less well understood. Bone marrow adipocytes originate from a resident, self-renewing population of multipotent bone marrow stromal cells which can also give rise to other lineages such as osteoblasts. The differentiation fate of these mesenchymal progenitors can be influenced to favour adipogenesis by several factors, including the administration of thiazolidinediones and increased age. Experimental data suggests that increases in bone marrow adipose tissue volume may make bone both more attractive to metastasis and conducive to cancer cell growth. Bone marrow adipocytes are known to secrete a variety of lipids, cytokines and bioactive signaling molecules known as adipokines, which have been implicated as mediators of the interaction between adipocytes and cancer cells. Recent studies have provided new insight into the impact of bone marrow adipose tissue volume expansion in regard to supporting and exacerbating the effects of bone metastasis from solid tumors, focusing on prostate, breast and lung cancer and blood cancers, focusing on multiple myeloma. In this mini-review, recent research developments pertaining to the role of factors which increase bone marrow adipose tissue volume, as well as the role of adipocyte secreted factors, in the progression of bone metastatic prostate and breast cancer are assessed. In particular, recent findings regarding the complex cross-talk between adipocytes and metastatic cells of both lung and prostate cancer are highlighted.


Asunto(s)
Células de la Médula Ósea , Neoplasias , Adipocitos/patología , Adipogénesis , Comunicación Celular , Humanos , Masculino , Neoplasias/patología , Microambiente Tumoral
18.
Biomedicines ; 10(1)2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-35052810

RESUMEN

Non-alcoholic steatohepatitis (NASH) is marked by macrophage infiltration and inflammation. Chemerin is a chemoattractant protein and is abundant in hepatocytes. The aim of this study was to gain insight into the role of hepatocyte-produced prochemerin in NASH. Therefore, mice were infected with adeno-associated virus 8 to direct hepatic overexpression of prochemerin in a methionine-choline deficient dietary model of NASH. At the end of the study, hepatic and serum chemerin were higher in the chemerin-expressing mice. These animals had less hepatic oxidative stress, F4/80 and CC-chemokine ligand 2 (CCL2) protein, and mRNA levels of inflammatory genes than the respective control animals. In order to identify the underlying mechanisms, prochemerin was expressed in hepatocytes and the hepatic stellate cells, LX-2. Here, chemerin had no effect on cell viability, production of inflammatory, or pro-fibrotic factors. Notably, cultivation of human peripheral blood mononuclear cells (PBMCs) in the supernatant of Huh7 cells overexpressing chemerin reduced CCL2, interleukin-6, and osteopontin levels in cell media. CCL2 was also low in RAW264.7 cells exposed to Hepa1-6 cell produced chemerin. In summary, the current study showed that prochemerin overexpression had little effect on hepatocytes and hepatic stellate cells. Of note, hepatocyte-produced chemerin deactivated PBMCs and protected against inflammation in experimental NASH.

19.
Nat Commun ; 13(1): 1559, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35322809

RESUMEN

CHKB encodes one of two mammalian choline kinase enzymes that catalyze the first step in the synthesis of the membrane phospholipid phosphatidylcholine. In humans and mice, inactivation of the CHKB gene (Chkb in mice) causes a recessive rostral-to-caudal muscular dystrophy. Using Chkb knockout mice, we reveal that at no stage of the disease is phosphatidylcholine level significantly altered. We observe that in affected muscle a temporal change in lipid metabolism occurs with an initial inability to utilize fatty acids for energy via mitochondrial ß-oxidation resulting in shunting of fatty acids into triacyglycerol as the disease progresses. There is a decrease in peroxisome proliferator-activated receptors and target gene expression specific to Chkb-/- affected muscle. Treatment of Chkb-/- myocytes with peroxisome proliferator-activated receptor agonists enables fatty acids to be used for ß-oxidation and prevents triacyglyerol accumulation, while simultaneously increasing expression of the compensatory choline kinase alpha (Chka) isoform, preventing muscle cell injury.


Asunto(s)
Enfermedades Musculares , Distrofias Musculares , Animales , Colina Quinasa/genética , Colina Quinasa/metabolismo , Ácidos Grasos , Metabolismo de los Lípidos/genética , Mamíferos/metabolismo , Ratones , Distrofias Musculares/genética , Distrofias Musculares/terapia , Fosfatidilcolinas/metabolismo
20.
Pharmacol Res ; 62(5): 408-15, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20654716

RESUMEN

Although amiodarone is the most effective antiarrhythmic agent currently available, concerns regarding adverse effects, including liver, lung and thyroid toxicity, often limit its use. Previously, we reported that amiodarone-induced hepatic steatosis in mice was associated with an upregulation of target genes modulated by peroxisome proliferator-activated receptor-alpha (PPARα). Because amiodarone does not directly stimulate PPARα, target gene induction may reflect a compensatory reaction countering some adverse effects of amiodarone. To test this, we examined co-treatment with the PPARα agonist, fenofibrate, and amiodarone in both PPARα(+/+) and PPARα(-/-) mice. Amiodarone treated PPARα(-/-) mice exhibited significantly greater weight loss and higher serum aspartate aminotransferase (AST) compared to PPARα(+/+) mice. Fenofibrate co-treatment reduced weight loss in amiodarone treated PPARα(-/-) mice, but not PPARα(+/+) mice. Fenofibrate stimulation of PPARα reduced serum amiodarone concentrations in normal mice. Serum amiodarone concentrations were higher in mice without PPARα expression given at 40-80 mg/kg amiodarone doses. These results are consistent with a protective influence of PPARα in reducing amiodarone-induced hepatic toxicity. In addition to PPARα-dependent effects, fenofibrate also demonstrated PPARα-independent actions that suggest a complex interaction modulating both hepatic lipid metabolism and amiodarone disposition. Further studies of the beneficial effect of fenofibrate and the interplay between lipid metabolism and amiodarone pharmacokinetics are required.


Asunto(s)
Amiodarona/toxicidad , Antiarrítmicos/toxicidad , Hígado/efectos de los fármacos , PPAR alfa/metabolismo , Amiodarona/sangre , Animales , Antiarrítmicos/sangre , Aspartato Aminotransferasas/sangre , Aspartato Aminotransferasas/metabolismo , Relación Dosis-Respuesta a Droga , Fenofibrato/farmacología , Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos , Lípidos/sangre , Hígado/enzimología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR alfa/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pérdida de Peso/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA