Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Res Ther ; 13(1): 37, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-35093170

RESUMEN

BACKGROUND: The bone marrow niche supports hematopoietic cell development through intimate contact with multipotent stromal mesenchymal stem cells; however, the intracellular signaling, function, and regulation of such supportive niche cells are still being defined. Our study was designed to understand how G protein receptor kinase 3 (GRK3) affects bone marrow mesenchymal stem cell function by examining primary cells from GRK3-deficient mice, which we have previously published to have a hypercellular bone marrow and leukocytosis through negative regulation of CXCL12/CXCR4 signaling. METHODS: Murine GRK3-deficient bone marrow mesenchymal stromal cells were harvested and cultured to differentiate into three lineages (adipocyte, chondrocyte, and osteoblast) to confirm multipotency and compared to wild type cells. Immunoblotting, modified-TANGO experiments, and flow cytometry were used to further examine the effects of GRK3 deficiency on bone marrow mesenchymal stromal cell receptor signaling. Microcomputed tomography was used to determine trabecular and cortical bone composition of GRK3-deficient mice and standard ELISA to quantitate CXCL12 production from cellular cultures. RESULTS: GRK3-deficient, bone marrow-derived mesenchymal stem cells exhibit enhanced and earlier osteogenic differentiation in vitro. The addition of a sphingosine kinase inhibitor abrogated the osteogenic proliferation and differentiation, suggesting that sphingosine-1-phosphate receptor signaling was a putative G protein-coupled receptor regulated by GRK3. Immunoblotting showed prolonged ERK1/2 signaling after stimulation with sphingosine-1-phosphate in GRK3-deficient cells, and modified-TANGO assays suggested the involvement of ß-arrestin-2 in sphingosine-1-phosphate receptor internalization. CONCLUSIONS: Our work suggests that GRK3 regulates sphingosine-1-phosphate receptor signaling on bone marrow mesenchymal stem cells by recruiting ß-arrestin to the occupied GPCR to promote internalization, and lack of such regulation affects mesenchymal stem cell functionality.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , Animales , Diferenciación Celular , Proliferación Celular , Células Madre Mesenquimatosas/metabolismo , Ratones , Receptores de Esfingosina-1-Fosfato , Microtomografía por Rayos X
2.
Mol Immunol ; 106: 12-21, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30576947

RESUMEN

Chemerin receptor (CMKLR1) is a G protein-coupled receptor (GPCR) implicated in macrophage-mediated inflammation and in several forms of human arthritis. Analogous to other GPCR, CMKLR1 is likely regulated by G protein-coupled receptor kinase (GRK) phosphorylation of intracellular domains in an activation-dependent manner, which leads to recruitment and termination of intracellular signaling via desensitization and internalization of the receptor. The ubiquitously expressed GRK family members include GRK2, GRK3, GRK5, and GRK6, but it is unknown which GRK regulates CMKLR1 cellular and signaling functions. Our data show that activation of CMKLR1 by chemerin in primary macrophages leads to signaling and functional outcomes that are regulated by GRK6 and ß-arrestin 2. We show that arrestin recruitment to CMKLR1 following chemerin stimulation is enhanced with co-expression of GRK6. Further, internalization of endogenous CMKLR1, following the addition of chemerin, is decreased in inflammatory macrophages from GRK6- and ß-arrestin 2-deficient mice. These GRK6- and ß-arrestin 2-deficient macrophages display increased migration toward chemerin and altered AKT and Extracellular-signal Related Kinase (ERK) signaling. Our findings show that chemerin-activated CMKLR1 regulation in inflammatory macrophages is largely GRK6 and ß-arrestin mediated, which may impact innate immunity and have therapeutic implications in rheumatic disease.


Asunto(s)
Quimiocinas/inmunología , Quinasas de Receptores Acoplados a Proteína-G/inmunología , Inmunidad Innata , Péptidos y Proteínas de Señalización Intercelular/inmunología , Macrófagos/inmunología , Receptores Acoplados a Proteínas G/inmunología , Arrestina beta 2/inmunología , Animales , Línea Celular , Quimiocinas/genética , Quinasas de Receptores Acoplados a Proteína-G/genética , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Macrófagos/patología , Ratones , Ratones Noqueados , Receptores de Quimiocina , Receptores Acoplados a Proteínas G/genética , Enfermedades Reumáticas/genética , Enfermedades Reumáticas/inmunología , Enfermedades Reumáticas/patología , Arrestina beta 2/genética
3.
PLoS One ; 11(4): e0152856, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27049755

RESUMEN

Triple negative breast cancer (TNBC) is a heterogeneous disease that has a poor prognosis and limited treatment options. Chemokine receptor interactions are important modulators of breast cancer metastasis; however, it is now recognized that quantitative surface expression of one important chemokine receptor, CXCR4, may not directly correlate with metastasis and that its functional activity in breast cancer may better inform tumor pathogenicity. G protein coupled receptor kinase 3 (GRK3) is a negative regulator of CXCR4 activity, and we show that GRK expression correlates with tumorigenicity, molecular subtype, and metastatic potential in human tumor microarray analysis. Using established human breast cancer cell lines and an immunocompetent in vivo mouse model, we further demonstrate that alterations in GRK3 expression levels in tumor cells directly affect migration and invasion in vitro and the establishment of distant metastasis in vivo. The effects of GRK3 modulation appear to be specific to chemokine-mediated migration behaviors without influencing tumor cell proliferation or survival. These data demonstrate that GRK3 dysregulation may play an important part in TNBC metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Quinasa 3 del Receptor Acoplado a Proteína-G/fisiología , Animales , Femenino , Quinasa 3 del Receptor Acoplado a Proteína-G/genética , Silenciador del Gen , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia
4.
Nanomedicine ; 11(7): 1797-807, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26093057

RESUMEN

Nanoparticles (NPs) are cleared by monocytes and macrophages. Chemokines CCL2 and CCL5 are key mediators for recruitment of these immune cells into tumors and tissues. The purpose of this study was to investigate effects of CCL2 and CCL5 on the pharmacokinetics (PKs) of NPs. Mice deficient in CCL2 or CCL5 demonstrated altered clearance and tissue distribution of polyethylene glycol tagged liposomal doxorubicin (PLD) compared to control mice. The PK studies using mice bearing SKOV3 ovarian cancer xenografts revealed that the presence of tumor cells and higher expression of chemokines were significantly associated with greater clearance of PLD compared to non-tumor bearing mice. Plasma exposure of encapsulated liposomal doxorubicin positively correlated with the total exposure of plasma CCL2 and CCL5 in patients with recurrent epithelial ovarian cancer treated with PLD. These data emphasize that the interplay between PLD and chemokines may have an important role in optimizing PLD therapy. FROM THE CLINICAL EDITOR: The use of nanoparticles as drug delivery carriers is gaining widespread acceptance in the clinical setting. However, the underlying pharmacokinetics of these novel drugs has not really been elucidated. In this interesting article, the authors carried out experiments using mice deficient in CCL2 or CCL5 to study the clearance of liposomal system. They showed the important role the immune system played and would enable better designs of future drug delivery systems.


Asunto(s)
Quimiocina CCL2/sangre , Quimiocina CCL5/sangre , Doxorrubicina/análogos & derivados , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Animales , Carcinoma Epitelial de Ovario , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Ratones , Nanopartículas/administración & dosificación , Nanopartículas/química , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Neoplasias Glandulares y Epiteliales/sangre , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Distribución Tisular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Leukoc Biol ; 94(6): 1243-51, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23935208

RESUMEN

Chemokine receptor interactions coordinate leukocyte migration in inflammation. Chemokine receptors are GPCRs that when activated, are phosphorylated by GRKs to turn off G protein-mediated signaling yet recruit additional signaling machinery. Recently, GRK3 was identified as a negative regulator of CXCL12/CXCR4 signaling that is defective in human WHIM syndrome. Here, we report that GRK3-/- mice exhibit numerous features of human WHIM, such as impaired CXCL12-mediated desensitization, enhanced CXCR4 signaling to ERK activation, altered granulocyte migration, and a mild myelokathexis. Moreover, GRK3-/- protects mice from two acute models of inflammatory arthritis (K/BxN serum transfer and CAIA). In these granulocyte-dependent disease models, protection of GRK3-/- mice is mediated by retention of cells in the marrow, fewer circulating granulocytes in the peripheral blood, and reduced granulocytes in the joints during active inflammation. In contrast to WHIM, GRK3-/- mice have minimal hypogammaglobulinemia and a peripheral leukocytosis with increased lymphocytes and absent neutropenia. Thus, we conclude that the loss of GRK3-mediated regulation of CXCL12/CXCR4 signaling contributes to some, but not all, of the complete WHIM phenotype and that GRK3 inhibition may be beneficial in the treatment of inflammatory arthritis.


Asunto(s)
Quinasa 3 del Receptor Acoplado a Proteína-G/inmunología , Síndromes de Inmunodeficiencia/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Verrugas/inmunología , Animales , Línea Celular Transformada , Quimiocina CXCL12/genética , Quimiocina CXCL12/inmunología , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Quinasa 3 del Receptor Acoplado a Proteína-G/genética , Quinasa 3 del Receptor Acoplado a Proteína-G/metabolismo , Granulocitos/enzimología , Granulocitos/inmunología , Granulocitos/patología , Humanos , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Inflamación/enzimología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Verrugas/enzimología , Verrugas/genética , Verrugas/patología
6.
Mol Immunol ; 54(2): 193-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23280397

RESUMEN

Polymorphism at the GPSM3 gene locus is inversely associated with four systemic autoimmune diseases, including rheumatoid arthritis and ankylosing spondylitis. G-protein signaling modulator-3 (GPSM3) expression is most pronounced in myeloid cells, in which it targets heterotrimeric G-protein Gαi subunits of chemokine receptors, critical to immune function. To begin to explore the regulatory role of GPSM3 in monocytes, human THP-1 and primary mouse myeloid cells were cultured under stimulus conditions; GPSM3 was found by immunoblotting to be expressed at highest levels in the mature monocyte. To evaluate the effects of GPSM3 deficiency on a myeloid-dependent autoimmune disease, collagen antibody-induced arthritis (CAIA) was induced in Gpsm3-/- and control mice, which were then analyzed for clinical score, paw swelling, intra-articular proinflammatory markers, and histopathology. Mice lacking GPSM3 were protected from CAIA, and expression of monocyte-representative pro-inflammatory chemokine receptors and cytokines in paws of Gpsm3-/- mice were decreased. Flow cytometry, apoptosis, and transwell chemotaxis experiments were conducted to further characterize the effect of GPSM3 deficiency on survival and chemokine responsiveness of monocytes. GPSM3-deficient myeloid cells had reduced migration ex vivo to CCL2, CX3CL1, and chemerin and enhanced apoptosis in vitro. Our results suggest that GPSM3 is an important regulator of monocyte function involving mechanisms of differentiation, survival, and chemotaxis, and deficiency in GPSM3 expression is protective in acute inflammatory arthritis.


Asunto(s)
Artritis Experimental/genética , Artritis Experimental/inmunología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Inhibidores de Disociación de Guanina Nucleótido/genética , Monocitos/inmunología , Animales , Supervivencia Celular/genética , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Inhibidores de Disociación de Guanina Nucleótido/inmunología , Mediadores de Inflamación/inmunología , Ratones , Ratones Noqueados , Monocitos/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología
7.
Curr Allergy Asthma Rep ; 12(6): 495-510, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23054625

RESUMEN

Many biologic agents that were first approved for the treatment of malignancies are now being actively investigated and used in a variety of autoimmune diseases such as rheumatoid arthritis (RA), antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis, systemic lupus erythematosus (SLE), and Sjogren's syndrome. The relatively recent advance of selective immune targeting has significantly changed the management of autoimmune disorders and in part can be attributed to the progress made in understanding effector cell function and their signaling pathways. In this review, we will discuss the recent FDA-approved biologic therapies that directly target immune cells as well as the most promising investigational drugs affecting immune cell function and signaling for the treatment of autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Autoinmunidad/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Animales , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Antineoplásicos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Autoinmunidad/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Terapia Biológica , Humanos , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Proteínas Tirosina Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas/inmunología , Rituximab , Síndrome de Sjögren/tratamiento farmacológico , Síndrome de Sjögren/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
8.
Arthritis Rheum ; 64(5): 1379-87, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22144035

RESUMEN

OBJECTIVE: CX(3) CR1 is a chemokine receptor that uniquely binds to its ligand fractalkine (CX(3) CL1) and has been shown to be important in inflammatory arthritis responses, largely due to its effects on cellular migration. This study was undertaken to test the hypothesis that genetic deficiency of CX(3) CR1 is protective in the chronic inflammatory arthritis model collagen-induced arthritis (CIA). Because CX(3) CR1 is expressed on T cells and antigen-presenting cells, we also examined adaptive immune functions in this model. METHODS: Autoantibody formation, clinical, histologic, T cell proliferative, and cytokine responses were evaluated in wild-type and CX(3) CR1-deficient DBA/1J mice after immunization with heterologous type II collagen (CII). RESULTS: CX(3) CR1(-/-) mice had an ∼30% reduction in arthritis severity compared to wild-type mice, as determined by 2 independent measures, paw swelling (P < 0.01) and clinical disease score (P < 0.0001). Additionally, compared to wild-type mice, CX(3) CR1(-/-) mice had an ∼50% decrease in anti-CII autoantibody formation (P < 0.05), decreased Th17 intraarticular cytokine expression (P < 0.01 for interleukin-17 [IL-17] and P < 0.001 for IL-23), and decreased total numbers of Th17 cells in inflamed joints (P < 0.05). CONCLUSION: Our findings indicate that CX(3) CR1 deficiency is protective in inflammatory arthritis and may have effects that extend beyond migration that involve adaptive immune responses in autoimmune disease.


Asunto(s)
Artritis Experimental/inmunología , Inmunidad Humoral/inmunología , Receptores de Quimiocina/inmunología , Células Th17/inmunología , Inmunidad Adaptativa , Animales , Artritis Experimental/genética , Artritis Experimental/patología , Receptor 1 de Quimiocinas CX3C , Movimiento Celular , Progresión de la Enfermedad , Miembro Posterior , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Receptores de Quimiocina/deficiencia , Rodilla de Cuadrúpedos/patología , Células Th17/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...