Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Nephrol ; 52(7): 588-601, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34515038

RESUMEN

INTRODUCTION: The nonsteroidal mineralocorticoid receptor (MR) antagonist finerenone and sodium-glucose cotransporter-2 (SGLT2) inhibitors have demonstrated clinical benefits in chronic kidney disease patients with type 2 diabetes. Precise molecular mechanisms responsible for these benefits are incompletely understood. Here, we investigated potential direct anti-fibrotic effects and mechanisms of nonsteroidal MR antagonism by finerenone or SGLT2 inhibition by empagliflozin in 2 relevant mouse kidney fibrosis models: unilateral ureter obstruction and sub-chronic ischemia reperfusion injury. METHODS: Kidney fibrosis was induced in mice via unilateral ureteral obstruction or ischemia. In a series of experiments, mice were treated orally with the MR antagonist finerenone (3 or 10 mg/kg), the SGLT2 inhibitor empagliflozin (10 or 30 mg/kg), or in a direct comparison of both drugs. Interstitial myofibroblast accumulation was quantified via alpha-smooth muscle actin and interstitial collagen deposition via Sirius Red/Fast Green staining in both models. Secondary analyses included the assessment of inflammatory cells, kidney mRNA expression of fibrotic markers as well as functional parameters (serum creatinine and albuminuria) in the ischemic model. Blood pressure was measured via telemetry in healthy conscious compound-treated animals. RESULTS: Finerenone dose-dependently decreased pathological myofibroblast accumulation and collagen deposition with no effects on systemic blood pressure and inflammatory markers in the tested dose range. Reduced kidney fibrosis was paralleled by reduced kidney plasminogen activator inhibitor-1 (PAI-1) and naked cuticle 2 (NKD2) expression in finerenone-treated mice. In contrast, treatment with empagliflozin strongly increased urinary glucose excretion in both models and reduced ischemia-induced albuminuria but had no effects on kidney myofibroblasts or collagen deposition. DISCUSSION/CONCLUSION: Finerenone has direct anti-fibrotic properties resulting in reduced myofibroblast and collagen deposition accompanied by a reduction in renal PAI-1 and NKD2 expression in mouse models of progressive kidney fibrosis at blood pressure-independent dosages.


Asunto(s)
Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología , Riñón/patología , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Naftiridinas/uso terapéutico , Actinas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Albuminuria/tratamiento farmacológico , Animales , Compuestos de Bencidrilo/uso terapéutico , Presión Sanguínea/efectos de los fármacos , Proteínas de Unión al Calcio/genética , Colágeno/genética , Colágeno/metabolismo , Creatinina/sangre , Modelos Animales de Enfermedad , Fibrosis , Expresión Génica/efectos de los fármacos , Glucósidos/uso terapéutico , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Linfocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Antagonistas de Receptores de Mineralocorticoides/farmacología , Monocitos/patología , Miofibroblastos/patología , Naftiridinas/farmacología , ARN Mensajero/metabolismo , Daño por Reperfusión/complicaciones , Serpina E2/genética , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Obstrucción Ureteral/complicaciones
2.
Int J Mol Sci ; 22(15)2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34360780

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe and progressive muscle wasting disorder, affecting one in 3500 to 5000 boys worldwide. The NO-sGC-cGMP pathway plays an important role in skeletal muscle function, primarily by improving blood flow and oxygen supply to the muscles during exercise. In fact, PDE5 inhibitors have previously been investigated as a potential therapy for DMD, however, a large-scale Phase III clinical trial did not meet its primary endpoint. Since the efficacy of PDE5i is dependent on sufficient endogenous NO production, which might be impaired in DMD, we investigated if NO-independent sGC stimulators, could have therapeutic benefits in a mouse model of DMD. Male mdx/mTRG2 mice aged six weeks were given food supplemented with the sGC stimulator, BAY-747 (150 mg/kg of food) or food alone (untreated) ad libitum for 16 weeks. Untreated C57BL6/J mice were used as wild type (WT) controls. Assessments of the four-limb hang, grip strength, running wheel and serum creatine kinase (CK) levels showed that mdx/mTRG2 mice had significantly reduced skeletal muscle function and severe muscle damage compared to WT mice. Treatment with BAY-747 improved grip strength and running speed, and these mice also had reduced CK levels compared to untreated mdx/mTRG2 mice. We also observed increased inflammation and fibrosis in the skeletal muscle of mdx/mTRG2 mice compared to WT. While gene expression of pro-inflammatory cytokines and some pro-fibrotic markers in the skeletal muscle was reduced following BAY-747 treatment, there was no reduction in infiltration of myeloid immune cells nor collagen deposition. In conclusion, treatment with BAY-747 significantly improves several functional and pathological parameters of the skeletal muscle in mdx/mTRG2 mice. However, the effect size was moderate and therefore, more studies are needed to fully understand the potential treatment benefit of sGC stimulators in DMD.


Asunto(s)
Activadores de Enzimas/farmacología , Músculo Esquelético/enzimología , Distrofia Muscular de Duchenne/tratamiento farmacológico , Guanilil Ciclasa Soluble/metabolismo , Animales , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/enzimología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología
3.
Sci Rep ; 11(1): 13251, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34168267

RESUMEN

Mononuclear phagocytes (MNPs) participate in inflammation and repair after kidney injury, reflecting their complex nature. Dissection into refined functional subunits has been challenging and would benefit understanding of renal pathologies. Flow cytometric approaches are limited to classifications of either different MNP subsets or functional state. We sought to combine these two dimensions in one protocol that considers functional heterogeneity in each MNP subset. We identified five distinct renal MNP subsets based on a previously described strategy. In vitro polarization of bone marrow-derived macrophages (BMDM) into M1- and M2-like cells suggested functional distinction of CD86 + MHCII + CD206- and CD206 + cells. Combination of both distinction methods identified CD86 + MHCII + CD206- and CD206 + cells in all five MNP subsets, revealing their heterologous nature. Our approach revealed that MNP composition and their functional segmentation varied between different mouse models of kidney injury and, moreover, was dynamically regulated in a time-dependent manner. CD206 + cells from three analyzed MNP subsets had a higher ex vivo phagocytic capacity than CD86 + MHCII + CD206- counterparts, indicating functional uniqueness of each subset. In conclusion, our novel flow cytometric approach refines insights into renal MNP heterogeneity and therefore could benefit mechanistic understanding of renal pathology.


Asunto(s)
Citometría de Flujo/métodos , Fagocitos/metabolismo , Animales , Antígenos de Superficie , Antígeno B7-2/inmunología , Genes MHC Clase II/inmunología , Riñón/lesiones , Riñón/patología , Lectinas Tipo C/inmunología , Macrófagos/clasificación , Macrófagos/metabolismo , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagocitos/clasificación , Receptores de Superficie Celular/inmunología
4.
Cardiovasc Res ; 117(6): 1510-1522, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-32717023

RESUMEN

AIMS: Monocytes are central for atherosclerotic vascular inflammation. The human non-classical, patrolling subtype, which expresses high levels of CD16 and fractalkine receptor CX3CR1, strongly associates with cardiovascular events. This is most marked in renal failure, a condition with excess atherosclerosis morbidity. The underlying mechanism is not understood. This study investigated how human CD16+ monocytes modulate endothelial cell function. METHODS AND RESULTS: In patients with kidney failure, CD16+ monocyte counts were elevated and dynamically decreased within a year after transplantation, chiefly due to a drop in CD14+CD16+ cells. The CX3CR1 ligand CX3CL1 was similarly elevated in the circulation of humans and mice with renal impairment. CX3CL1 up-regulation was also observed close to macrophage rich human coronary artery plaques. To investigate a mechanistic basis of this association, CD16+CX3CR1HIGH monocytes were co-incubated with primary human endothelium in vitro. Compared to classical CD14+ monocytes or transwell cocultures, CD16+ monocytes enhanced endothelial STAT1 and NF-κB p65 phosphorylation, up-regulated expression of CX3CL1 and interleukin-1ß, numerous CCL and CXCL chemokines and molecules promoting leucocyte patrolling and adhesion such as ICAM1 and VCAM1. Genes required for vasodilatation including endothelial nitric oxide synthase decreased while endothelial collagen production increased. Uraemic patients' monocytes enhanced endothelial CX3CL1 even more markedly. Their receptor CX3CR1 was required for enhanced aortic endothelial stiffness in murine atherosclerosis with renal impairment. CX3CR1 dose-dependently modulated monocyte-contact-dependent gene expression in human endothelium. CONCLUSION: By demonstrating endothelial proatherosclerotic gene regulation in direct contact with CD16+ monocytes, in part via cellular CX3CR1-CX3CL1 interaction, our data delineate a mechanism how this celltype can increase cardiovascular risk.


Asunto(s)
Aterosclerosis/metabolismo , Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CX3CL1/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Monocitos/metabolismo , Placa Aterosclerótica , Receptores de IgG/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Receptor 1 de Quimiocinas CX3C/genética , Comunicación Celular , Células Cultivadas , Quimiocina CX3CL1/genética , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Proteínas Ligadas a GPI/metabolismo , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Enfermedades Renales/inmunología , Enfermedades Renales/metabolismo , Enfermedades Renales/terapia , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Fenotipo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transducción de Señal , Uremia/inmunología , Uremia/metabolismo
5.
J Leukoc Biol ; 109(4): 741-751, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32803826

RESUMEN

The G protein-coupled free fatty acid receptor 2 (FFA2R) is highly expressed on neutrophils and was previously described to regulate neutrophil activation. Allosteric targeting of G protein-coupled receptors (GPCRs) is increasingly explored to create distinct pharmacology compared to endogenous, orthosteric ligands. The consequence of allosteric versus orthosteric FFA2R activation for neutrophil response, however, is currently largely elusive. Here, different FFA2R desensitization profiles in human neutrophils following allosteric or orthosteric activation are reported. Using a set of neutrophil functional assays to measure calcium flux, pERK1/2, chemotaxis, cellular degranulation, and oxidative burst together with holistic and pathway-unbiased whole cell sensing based on dynamic mass redistribution, it is found that the synthetic positive allosteric modulator agonist 4-CMTB potently activates neutrophils and simultaneously alters FFA2R responsiveness toward the endogenous, orthosteric agonist propionic acid (C3) after homologous and heterologous receptor desensitization. Stimulation with C3 or the hierarchically superior chemokine receptor activator IL-8 led to strong FFA2R desensitization and rendered neutrophils unresponsive toward repeated stimulation with C3. In contrast, stimulation with allosteric 4-CMTB engaged a distinct composition of signaling pathways as compared to orthosteric receptor activation and was able to activate neutrophils that underwent homologous and heterologous desensitization with C3 and IL-8, respectively. Moreover, allosteric FFA2R activation could re-sensitize FFA2 toward the endogenous agonist C3 after homologous and heterologous desensitization. Given the fact that receptor desensitization is critical in neutrophils to sense and adapt to their current environment, these findings are expected to be useful for the discovery of novel pharmacological mechanisms to modulate neutrophil responsiveness therapeutically.


Asunto(s)
Neutrófilos/metabolismo , Receptores de Superficie Celular/metabolismo , Regulación Alostérica/efectos de los fármacos , Calcio/metabolismo , Quimiotaxis/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Activación Neutrófila/efectos de los fármacos , Neutrófilos/química , Neutrófilos/efectos de los fármacos , Propionatos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Biomolecules ; 10(11)2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33228017

RESUMEN

Peritoneal dialysis (PD) is limited by glucose-mediated peritoneal membrane (PM) fibrosis, angiogenesis, and ultrafiltration failure. Influencing PM integrity by pharmacologically targeting sodium-dependent glucose transporter (SGLT)-mediated glucose uptake has not been studied. In this study, wildtype C57Bl/6N mice were treated with high-glucose dialysate via an intraperitoneal catheter, with or without addition of selective SGLT2 inhibitor dapagliflozin. PM structural changes, ultrafiltration capacity, and peritoneal equilibration testing (PET) status for glucose, urea, and creatinine were analyzed. Expression of SGLT and facilitative glucose transporters (GLUT) was analyzed by real-time PCR, immunofluorescence, and immunohistochemistry. Peritoneal effluents were analyzed for cellular and cytokine composition. We found that peritoneal SGLT2 was expressed in mesothelial cells and in skeletal muscle. Dapagliflozin significantly reduced effluent transforming growth factor (TGF-ß) concentrations, peritoneal thickening, and fibrosis, as well as microvessel density, resulting in improved ultrafiltration, despite the fact that it did not affect development of high-glucose transporter status. In vitro, dapagliflozin reduced monocyte chemoattractant protein-1 release under high-glucose conditions in human and murine peritoneal mesothelial cells. Proinflammatory cytokine release in macrophages was reduced only when cultured in high-glucose conditions with an additional inflammatory stimulus. In summary, dapagliflozin improved structural and functional peritoneal health in the context of high-glucose PD.


Asunto(s)
Compuestos de Bencidrilo/uso terapéutico , Soluciones para Diálisis/toxicidad , Glucosa/toxicidad , Glucósidos/uso terapéutico , Fibrosis Peritoneal/tratamiento farmacológico , Peritoneo/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Transportador 2 de Sodio-Glucosa/biosíntesis , Adolescente , Animales , Compuestos de Bencidrilo/farmacología , Línea Celular Transformada , Células Cultivadas , Femenino , Glucósidos/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Peritoneo/metabolismo , Peritoneo/patología , Células RAW 264.7 , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Ultrafiltración/métodos
7.
Kidney Int ; 95(6): 1405-1417, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30948201

RESUMEN

Peritoneal dialysis (PD) is limited by chronic fibrotic remodeling of the peritoneal wall, a transforming growth factor-ß (TGF-ß)-mediated process. The fractalkine (CX3CL1) receptor CX3CR1 is expressed on macrophages and monocytes, where it is a marker of TGFß expression. Detection of its ligand CX3CL1 on the peritoneal mesothelium led us to hypothesize a pathophysiologic role of CX3CL1-CX3CR1 interaction in peritoneal fibrosis. We found that CX3CL1 was expressed on peritoneal mesothelial cells from PD patients and in a murine PD model. CX3CR1, mostly expressed on macrophages in the peritoneal wall, promoted fibrosis induced by chronic dialysate exposure in the mouse model. Our data suggest a positive feedback loop whereby direct interaction with CX3CR1-expressing macrophages promotes mesothelial expression of CX3CL1 and TGFß expression. In turn, TGFß upregulates CX3CR1 in murine and human monocytic cells. Upstream, macrophage cytokines including interleukin-1ß (IL-1ß) promote mesothelial CX3CR1 and TGFß expression, providing a starting point for CX3CL1-CX3CR1 interaction. IL-1ß expression was enhanced by exposure to dialysate both in vitro and in the mouse models. Our data suggest that macrophage-mesothelial cell crosstalk through CX3CR1-CX3CL1 interaction enhances mesothelial TGFß production, promoting peritoneal fibrosis in response to dialysate exposure. This interaction could be a novel therapeutic target in PD-associated chronic peritoneal fibrosis.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CX3CL1/metabolismo , Fibrosis Peritoneal/patología , Anciano , Animales , Comunicación Celular , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Soluciones para Diálisis/toxicidad , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Femenino , Humanos , Interleucina-1beta/metabolismo , Leucocitos Mononucleares , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/etiología , Peritoneo/citología , Peritoneo/patología , Cultivo Primario de Células , Insuficiencia Renal Crónica/terapia , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba
8.
Cytokine ; 122: 154089, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-28663097

RESUMEN

This review summarizes the current data on the interleukin (IL)-17A pathway in experimental atherosclerosis and clinical data. IL-17A is a prominent cytokine for early T cell response produced by both innate and adaptive leukocytes. In atherosclerosis, increased total IL-17A levels and expression in CD4+ T helper and γδ T cells have been demonstrated. Cytokines including IL-6 and TGFß that increase IL-17A expression are elevated. Many other factors such as lipids, glucose and sodium chloride concentrations as well as vitamins and arylhydrocarbon receptor agonists that promote IL-17A expression are closely associated with cardiovascular risk in the human population. In acute inflammation models, IL-17A mediates innate leukocyte recruitment of both neutrophils and monocytes. In atherosclerosis, IL-17A increased aortic macrophage and T cell infiltration in most models. Secondary recruitment effects via the endothelium and according to recent data also pericytes have been demonstrated. IL-17 receptor A is highly expressed on monocytes and direct effects have been reported as well. Beyond leukocyte accumulation, IL-17A may affect other factors of plaque formation such as endothelial function, and according to some reports, fibrous cap formation and vascular relaxation with an increase in blood pressure. Anti-IL-17A agents are now available for clinical use. Cardiovascular side effect profiles are benign at this point. IL-17A appears to be a differential regulator of atherosclerosis and its effects in mouse models suggest that its modulation may have contradictory effects on plaque size and possibly stability in different patient populations.


Asunto(s)
Aterosclerosis/metabolismo , Interleucina-17/antagonistas & inhibidores , Interleucina-17/metabolismo , Placa Aterosclerótica/metabolismo , Receptores de Interleucina-17/metabolismo , Animales , Aorta/metabolismo , Aterosclerosis/inmunología , Aterosclerosis/fisiopatología , Citocinas/metabolismo , Glucosa/metabolismo , Humanos , Inflamación/metabolismo , Metabolismo de los Lípidos , Macrófagos/metabolismo , Monocitos/metabolismo , Insuficiencia Renal Crónica/metabolismo , Factores de Riesgo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
9.
FASEB J ; 33(2): 2278-2289, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30260706

RESUMEN

Endothelial cells can acquire a mesenchymal phenotype upon irritation [endothelial-to-mesenchymal transition (EndMT)]. Macrophages accumulate in the atherosclerotic plaque. This study addressed whether macrophages modulate EndMT and delineated a reciprocal effect of EndMT on macrophage functions in atherosclerosis. In atherosclerotic murine and human aortas, endothelial cells with mesenchymal markers were elevated by confocal microscopy and flow cytometric analysis. Increased EndMT master transcription factor Snai1 expression and extracellular matrix are consistent with enhanced EndMT in this condition. Hypoxia was detected in individual aortic EndMT cells in vivo and rapidly induced a similar EndMT phenotype in vitro. As a novel inducer of EndMT, macrophages, which are abundant in the atherosclerotic lesions, enhance mesothelial marker expression during coculture in vitro. In the reverse relationship, EndMT altered endothelial colony-stimulating factor expression. Functionally, EndMT cell-conditioned media attenuated macrophage proliferation, antigen-presenting cell marker expression, and TNF-α production in response to oxidized LDL but increased oxidized LDL uptake and scavenger receptor expression. These experiments demonstrate that macrophages promote partial EndMT. In turn, EndMT cells modulate macrophage phenotype and lipid uptake. Our data suggest that EndMT shapes macrophage and endothelial cell phenotypes, thus affecting internal atherosclerotic plaque in addition to surface structure.-Helmke, A., Casper, J., Nordlohne, J., David, S., Haller, H., Zeisberg, E. M., von Vietinghoff, S. Endothelial-to-mesenchymal transition shapes the atherosclerotic plaque and modulates macrophage function.


Asunto(s)
Endotelio/patología , Macrófagos/citología , Mesodermo/patología , Placa Aterosclerótica/patología , Animales , Antígenos/inmunología , Aorta/metabolismo , Biomarcadores/metabolismo , Antígenos CD36/metabolismo , Hipoxia de la Célula , Proliferación Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Endotelio/metabolismo , Matriz Extracelular/patología , Femenino , Humanos , Lipoproteínas LDL/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/inmunología , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Factores de Transcripción de la Familia Snail/metabolismo
10.
JACC Basic Transl Sci ; 3(1): 54-66, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30062194

RESUMEN

Effective therapy of atherosclerotic complications in patients with chronic kidney disease (CKD) is an unmet clinical need. Cardiovascular events are the most common cause of death. At a glomerular filtration rate ≤60 ml/min, these events are increased also after correction for common risk factors. Previous studies have reported enhanced vascular inflammation in mice and recently also in humans. Our current data show, in a mouse model of atherosclerosis in moderate renal impairment, that interleukin-17 receptor A is instrumental in this condition, and blockade of this pathway can normalize arterial inflammation even in advanced atherosclerosis.

11.
Clin Case Rep ; 4(8): 840-1, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27525098

RESUMEN

Multicystic back masses can be of infectious, metastatic, or local pre- or malignant origin. We present a case of a rapidly evolving mass in a hemodialysis patient with severe "chronic kidney disease-associated mineral bone disease" (CKD-MBD), that also highlights limitations of chest x-ray for diagnosis of bone disease.

12.
J Am Soc Nephrol ; 27(6): 1753-64, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26449606

RESUMEN

Reduced kidney function increases the risk for atherosclerosis and cardiovascular death. Leukocytes in the arterial wall contribute to atherosclerotic plaque formation. We investigated the role of fractalkine receptor CX3CR1 in atherosclerotic inflammation in renal impairment. Apoe(-/-) (apolipoprotein E) CX3CR1(-/-) mice with renal impairment were protected from increased aortic atherosclerotic lesion size and macrophage accumulation. Deficiency of CX3CR1 in bone marrow, only, attenuated atherosclerosis in renal impairment in an independent atherosclerosis model of LDL receptor-deficient (LDLr(-/-)) mice as well. Analysis of inflammatory leukocytes in atherosclerotic mixed bone-marrow chimeric mice (50% wild-type/50% CX3CR1(-/-) bone marrow into LDLr(-/-) mice) showed that CX3CR1 cell intrinsically promoted aortic T cell accumulation much more than CD11b(+)CD11c(+) myeloid cell accumulation and increased IL-17-producing T cell counts. In vitro, fewer TH17 cells were obtained from CX3CR1(-/-) splenocytes than from wild-type splenocytes after polarization with IL-6, IL-23, and TGFß Polarization of TH17 or TREG cells, or stimulation of splenocytes with TGFß alone, increased T cell CX3CR1 reporter gene expression. Furthermore, TGFß induced CX3CR1 mRNA expression in wild-type cells in a dose- and time-dependent manner. In atherosclerotic LDLr(-/-) mice, CX3CR1(+/-) T cells upregulated CX3CR1 and IL-17A production in renal impairment, whereas CX3CR1(-/-) T cells did not. Transfer of CX3CR1(+/-) but not Il17a(-/-) T cells into LDLr(-/-)CX3CR1(-/-) mice increased aortic lesion size and aortic CD11b(+)CD11c(+) myeloid cell accumulation in renal impairment. In summary, T cell CX3CR1 expression can be induced by TGFß and is instrumental in enhanced atherosclerosis in renal impairment.


Asunto(s)
Aterosclerosis/etiología , Inflamación/etiología , Receptores de Quimiocina/fisiología , Insuficiencia Renal/complicaciones , Animales , Receptor 1 de Quimiocinas CX3C , Células Cultivadas , Femenino , Masculino , Ratones , Receptores de Quimiocina/genética
13.
Br J Haematol ; 164(2): 251-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24164493

RESUMEN

Hodgkin lymphoma (HL) and systemic anaplastic large cell lymphoma (sALCL) are rare lymphoproliferative cancer types. Although most HL patients can be cured by chemo- and radio-therapy, 4-50% of patients relapse and have a poor prognosis. The need for improved therapeutic options for patients with relapsed or refractory disease has been addressed by CD30-specific antibody-based immunotherapeutics. However, available CD30-specific monoclonal antibodies (mAbs), antibody drug conjugates (ADCs) or chimeric immunotoxins suffer from the requirement of a functional host immunity, undesirable immune reactions or heterogeneity and instability, respectively. Here, we present a new fusion protein comprised of the CD30-specific antibody single-chain fragment Ki4(scFv) and the human pro-apoptotic effector protein, microtubule-associated protein tau (MAPT). Ki4(scFv)-MAP selectively induced apoptosis in rapidly proliferating L540cy, L428, and Karpas 299 cells in a dose-dependent manner. Tubulin polymerization assays confirmed that Ki4(scFv)-MAP stabilizes microtubules, suggesting a mechanism for its pro-apoptotic action. Dose-finding experiments proved that Ki4(scFv)-MAP is well tolerated in mice compared to the previously reported Ki4(scFv)-ETA'. Ki4(scFv)-MAP significantly inhibited growth of subcutaneous L540cy xenograft tumours in mice. Our data present a novel approach for the treatment of CD30(+) lymphomas, combining the binding specificity of a target-specific antibody fragment with the selective cytotoxicity of MAPT towards proliferating lymphoma cells.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Enfermedad de Hodgkin/metabolismo , Enfermedad de Hodgkin/patología , Antígeno Ki-1/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos de Cadena Única/farmacología , Proteínas tau/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Antígeno Ki-1/inmunología , Ratones , Microtúbulos/metabolismo , Unión Proteica , Estabilidad Proteica , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas tau/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...