Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Microsc ; 286(2): 120-125, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34664277

RESUMEN

Carbonated wollastonite clinker (CS) may be suitable as supplementary cementitious material (SCM) for mortar and concrete. The microstructure of unground CS clinker, carbonated CS slurry and a mortar blended with carbonated CS are investigated by scanning electron microscopy. Additionally, a reference mortar with pure Portland cement and one with a cement replacement level of 30 mass-% by carbonated CS are produced to assess its contribution to compressive strength development. The calcium silicates are decalcified during carbonation resulting in CaCO3 and amorphous SiO2 . The latter reacts when used as SCM in mortar influencing the Ca/Si ratio of calcium-silicate-hydrate and contributing to compressive strength development.


Asunto(s)
Calcio , Dióxido de Silicio , Compuestos de Calcio/química , Silicatos/química
2.
Cells ; 10(11)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34831306

RESUMEN

In diabetic patients, medial vascular calcification is common and associated with increased cardiovascular mortality. Excessive glucose concentrations can activate the nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-kB) and trigger pro-calcific effects in vascular smooth muscle cells (VSMCs), which may actively augment vascular calcification. Zinc is able to mitigate phosphate-induced VSMC calcification. Reduced serum zinc levels have been reported in diabetes mellitus. Therefore, in this study the effects of zinc supplementation were investigated in primary human aortic VSMCs exposed to excessive glucose concentrations. Zinc treatment was found to abrogate the stimulating effects of high glucose on VSMC calcification. Furthermore, zinc was found to blunt the increased expression of osteogenic and chondrogenic markers in high glucose-treated VSMCs. High glucose exposure was shown to activate NF-kB in VSMCs, an effect that was blunted by additional zinc treatment. Zinc was further found to increase the expression of TNFα-induced protein 3 (TNFAIP3) in high glucose-treated VSMCs. The silencing of TNFAIP3 was shown to abolish the protective effects of zinc on high glucose-induced NF-kB-dependent transcriptional activation, osteogenic marker expression, and the calcification of VSMCs. Silencing of the zinc-sensing receptor G protein-coupled receptor 39 (GPR39) was shown to abolish zinc-induced TNFAIP3 expression and the effects of zinc on high glucose-induced osteogenic marker expression. These observations indicate that zinc may be a protective factor during vascular calcification in hyperglycemic conditions.


Asunto(s)
Glucosa/toxicidad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Osteogénesis/efectos de los fármacos , Zinc/farmacología , Aorta/patología , Biomarcadores/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Clin Sci (Lond) ; 135(3): 515-534, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33479769

RESUMEN

In chronic kidney disease (CKD), hyperphosphatemia is a key factor promoting medial vascular calcification, a common complication associated with cardiovascular events and high mortality. Vascular calcification involves osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs), but the complex signaling events inducing pro-calcific pathways are incompletely understood. The present study investigated the role of acid sphingomyelinase (ASM)/ceramide as regulator of VSMC calcification. In vitro, both, bacterial sphingomyelinase and phosphate increased ceramide levels in VSMCs. Bacterial sphingomyelinase as well as ceramide supplementation stimulated osteo-/chondrogenic transdifferentiation during control and high phosphate conditions and augmented phosphate-induced calcification of VSMCs. Silencing of serum- and glucocorticoid-inducible kinase 1 (SGK1) blunted the pro-calcific effects of bacterial sphingomyelinase or ceramide. Asm deficiency blunted vascular calcification in a cholecalciferol-overload mouse model and ex vivo isolated-perfused arteries. In addition, Asm deficiency suppressed phosphate-induced osteo-/chondrogenic signaling and calcification of cultured VSMCs. Treatment with the functional ASM inhibitors amitriptyline or fendiline strongly blunted pro-calcific signaling pathways in vitro and in vivo. In conclusion, ASM/ceramide is a critical upstream regulator of vascular calcification, at least partly, through SGK1-dependent signaling. Thus, ASM inhibition by repurposing functional ASM inhibitors to reduce the progression of vascular calcification during CKD warrants further study.


Asunto(s)
Transdiferenciación Celular , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Esfingomielina Fosfodiesterasa/farmacología , Calcificación Vascular/patología , Amitriptilina/farmacología , Animales , Células Cultivadas , Ceramidas/metabolismo , Condrogénesis/efectos de los fármacos , Fendilina/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Fosfatos/farmacología
4.
J Mol Med (Berl) ; 99(3): 373-382, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33409552

RESUMEN

Vascular calcification may result from stimulation of osteogenic signalling with upregulation of the transcription factors CBFA1, MSX2 and SOX9, as well as alkaline phosphatase (ALPL), which degrades and thus inactivates the calcification inhibitor pyrophosphate. Osteogenic signalling further involves upregulation of the Ca2+-channel ORAI1. The channel is activated by STIM1 and then accomplishes store-operated Ca2+ entry. ORAI1 and STIM1 are upregulated by the serum & glucocorticoid inducible kinase 1 (SGK1) which is critically important for osteogenic signalling. Stimulators of vascular calcification include vasopressin. The present study explored whether exposure of human aortic smooth muscle cells (HAoSMCs) to vasopressin upregulates ORAI1 and/or STIM1 expression, store-operated Ca2+ entry and osteogenic signalling. To this end, HAoSMCs were exposed to vasopressin (100 nM, 24 h) without or with additional exposure to ORAI1 blocker MRS1845 (10 µM) or SGK1 inhibitor GSK-650394 (1 µM). Transcript levels were measured using q-RT-PCR, cytosolic Ca2+-concentration ([Ca2+]i) by Fura-2-fluorescence, and store-operated Ca2+ entry from increase of [Ca2+]i following re-addition of extracellular Ca2+ after store depletion with thapsigargin (1 µM). As a result, vasopressin enhanced the transcript levels of ORAI1 and STIM1, store-operated Ca2+ entry, as well as the transcript levels of CBFA1, MSX2, SOX9 and ALPL. The effect of vasopressin on store-operated Ca2+ entry as well as on transcript levels of CBFA1, MSX2, SOX9 and ALPL was virtually abrogated by MRS1845 and GSK-650394. In conclusion, vasopressin stimulates expression of ORAI1/STIM1, thus augmenting store-operated Ca2+ entry and osteogenic signalling. In HAoSMCs, vasopressin (VP) upregulates Ca2+ channel ORAI1 and its activator STIM1. VP upregulates store-operated Ca2+ entry (SOCE) and osteogenic signalling (OS). VP-induced SOCE, OS and Ca2+-deposition are disrupted by ORAI1 inhibitor MRS1845. VP-induced SOCE, OS and Ca2+-deposition are disrupted by SGK1 blocker GSK-650394. KEY MESSAGES: • In HAoSMCs, vasopressin (VP) upregulates Ca2+ channel ORAI1 and its activator STIM1. • VP upregulates store-operated Ca2+ entry (SOCE) and osteogenic signalling (OS). • VP-induced SOCE, OS and Ca2+-deposition are disrupted by ORAI1 inhibitor MRS1845. • VP-induced SOCE, OS and Ca2+-deposition are disrupted by SGK1 blocker GSK-650394.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Proteína ORAI1/biosíntesis , Calcificación Vascular/metabolismo , Vasopresinas/farmacología , Aorta/citología , Benzoatos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/fisiología , Células Cultivadas , Evaluación Preclínica de Medicamentos , Humanos , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/fisiología , Miocitos del Músculo Liso/metabolismo , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Nitrendipino/análogos & derivados , Nitrendipino/farmacología , Proteína ORAI1/antagonistas & inhibidores , Proteína ORAI1/genética , Osteogénesis/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/fisiología , Molécula de Interacción Estromal 1/biosíntesis , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/fisiología , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Calcificación Vascular/prevención & control
5.
Artículo en Inglés | MEDLINE | ID: mdl-29780355

RESUMEN

Medial vascular calcification, a major pathophysiological process associated with cardiovascular disease and mortality, involves osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs). In chronic kidney disease (CKD), osteo-/chondrogenic transdifferentiation of VSMCs and, thus, vascular calcification is mainly driven by hyperphosphatemia, resulting from impaired elimination of phosphate by the diseased kidneys. Hyperphosphatemia with subsequent vascular calcification is a hallmark of klotho-hypomorphic mice, which are characterized by rapid development of multiple age-related disorders and early death. In those animals, hyperphosphatemia results from unrestrained formation of 1,25(OH)2D3 with subsequent retention of calcium and phosphate. Analysis of klotho-hypomorphic mice and mice with vitamin D3 overload uncovered several pathophysiological mechanisms participating in the orchestration of vascular calcification and several therapeutic opportunities to delay or even halt vascular calcification. The present brief review addresses the beneficial effects of bicarbonate, carbonic anhydrase inhibition, magnesium supplementation, mineralocorticoid receptor (MR) blockage, and ammonium salts. The case is made that bicarbonate is mainly effective by decreasing intestinal phosphate absorption, and that carbonic anhydrase inhibition leads to metabolic acidosis, which counteracts calcium-phosphate precipitation and VSMC transdifferentiation. Magnesium supplementation, MR blockage and ammonium salts are mainly effective by interference with osteo-/chondrogenic signaling in VSMCs. It should be pointed out that the, by far, most efficient substances are ammonium salts, which may virtually prevent vascular calcification. Future research will probably uncover further therapeutic options and, most importantly, reveal whether these observations in mice can be translated into treatment of patients suffering from vascular calcification, such as patients with CKD.

6.
Cell Physiol Biochem ; 46(4): 1305-1316, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29689558

RESUMEN

BACKGROUND/AIMS: Fibulin-3, an extracellular matrix glycoprotein, inhibits vascular oxidative stress and remodeling in hypertension. Oxidative stress is prevalent in chronic kidney disease (CKD) patients and is an important mediator of osteo-/chondrogenic transdifferentiation and calcification of vascular smooth muscle cells (VSMCs) during hyperphosphatemia. Therefore, the present study explored the effects of Fibulin-3 on phosphate-induced vascular calcification. METHODS: Experiments were performed in primary human aortic smooth muscle cells (HAoSMCs) treated with control or with phosphate without or with additional treatment with recombinant human Fibulin-3 protein or with hydrogen peroxide as an exogenous source of oxidative stress. RESULTS: Treatment with calcification medium significantly increased calcium deposition in HAoSMCs, an effect significantly blunted by additional treatment with Fibulin-3. Moreover, phosphate-induced alkaline phosphatase activity and mRNA expression of osteogenic and chondrogenic markers MSX2, CBFA1, SOX9 and ALPL were all significantly reduced by addition of Fibulin-3. These effects were paralleled by similar regulation of oxidative stress in HAoSMCs. Phosphate treatment significantly up-regulated mRNA expression of the oxidative stress markers NOX4 and CYBA, down-regulated total antioxidant capacity and increased the expression of downstream effectors of oxidative stress PAI-1, MMP2 and MMP9 as well as BAX/BLC2 ratio in HAoSMCs, all effects blocked by additional treatment with Fibulin-3. Furthermore, the protective effects of Fibulin-3 on phosphate-induced osteogenic and chondrogenic markers expression in HAoSMCs were reversed by additional treatment with hydrogen peroxide. CONCLUSIONS: Fibulin-3 attenuates phosphate-induced osteo-/ chondrogenic transdifferentiation and calcification of VSMCs, effects involving inhibition of oxidative stress. Up-regulation or supplementation of Fibulin-3 may be beneficial in reducing the progression of vascular calcification during hyperphosphatemic conditions such as CKD.


Asunto(s)
Calcificación Fisiológica/efectos de los fármacos , Proteínas de la Matriz Extracelular/farmacología , Glicerofosfatos/farmacología , Estrés Oxidativo/efectos de los fármacos , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Línea Celular , Transdiferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , Osteogénesis/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo
7.
J Am Soc Nephrol ; 29(6): 1636-1648, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29654213

RESUMEN

Background The high cardiovascular morbidity and mortality of patients with CKD may result in large part from medial vascular calcification, a process promoted by hyperphosphatemia and involving osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs). Reduced serum zinc levels have frequently been observed in patients with CKD, but the functional relevance of this remains unclear.Methods We performed experiments in primary human aortic VSMCs; klotho-hypomorphic (kl/kl), subtotal nephrectomy, and cholecalciferol-overload mouse calcification models; and serum samples from patients with CKD.Results In cultured VSMCs, treatment with zinc sulfate (ZnSO4) blunted phosphate-induced calcification, osteo-/chondrogenic signaling, and NF-κB activation. ZnSO4 increased the abundance of zinc-finger protein TNF-α-induced protein 3 (TNFAIP3, also known as A20), a suppressor of the NF-κB pathway, by zinc-sensing receptor ZnR/GPR39-dependent upregulation of TNFAIP3 gene expression. Silencing of TNFAIP3 in VSMCs blunted the anticalcific effects of ZnSO4 under high phosphate conditions. kl/kl mice showed reduced plasma zinc levels, and ZnSO4 supplementation strongly blunted vascular calcification and aortic osteoinduction and upregulated aortic Tnfaip3 expression. ZnSO4 ameliorated vascular calcification in mice with chronic renal failure and mice with cholecalciferol overload. In patients with CKD, serum zinc concentrations inversely correlated with serum calcification propensity. Finally, ZnSO4 ameliorated the osteoinductive effects of uremic serum in VSMCs.Conclusions Zinc supplementation ameliorates phosphate-induced osteo-/chondrogenic transdifferentiation of VSMCs and vascular calcification through an active cellular mechanism resulting from GPR39-dependent induction of TNFAIP3 and subsequent suppression of the NF-κB pathway. Zinc supplementation may be a simple treatment to reduce the burden of vascular calcification in CKD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fallo Renal Crónico/sangre , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Calcificación Vascular/prevención & control , Sulfato de Zinc/farmacología , Animales , Aorta , Transdiferenciación Celular , Células Cultivadas , Suplementos Dietéticos , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Silenciador del Gen , Glucuronidasa/genética , Humanos , Hidroxietilrutósido , Hiperfosfatemia/sangre , Hiperfosfatemia/complicaciones , Proteínas Klotho , Ratones , FN-kappa B/antagonistas & inhibidores , Nefrectomía , Nefrocalcinosis/prevención & control , Fosfatos , Transducción de Señal , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Calcificación Vascular/sangre , Calcificación Vascular/etiología , Zinc/sangre
8.
Biochim Biophys Acta Biomembr ; 1860(5): 1114-1124, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29317198

RESUMEN

Particularly in Asia medicinal plants with antimicrobial activity are used for therapeutic purpose. One such plant-derived antibiotic is rhodomyrtone (Rom) isolated from Rhodomyrtus tomentosa leaves. Rom shows high antibacterial activity against a wide range of Gram-positive bacteria, however, its mode of action is still unclear. Reporter gene assays and proteomic profiling experiments in Bacillus subtilis indicate that Rom does not address classical antibiotic targets like translation, transcription or DNA replication, but acts at the cytoplasmic membrane. In Staphylococcus aureus, Rom decreases the membrane potential within seconds and at low doses, causes release of ATP and even the excretion of cytoplasmic proteins (ECP), but does not induce pore-formation as for example nisin. Lipid staining revealed that Rom induces local membrane damage. Rom's antimicrobial activity can be antagonized in the presence of a very narrow spectrum of saturated fatty acids (C15:0, C16:0, or C18:0) that most likely contribute to counteract the membrane damage. Gram-negative bacteria are resistant to Rom, presumably due to reduced penetration through the outer membrane and its neutralization by LPS. Rom is cytotoxic for many eukaryotic cells and studies with human erythrocytes showed that Rom induces eryptosis accompanied by erythrocyte shrinkage, cell membrane blebbing, and membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. Rom's distinctive interaction with the cytoplasmic membrane reminds on the amphipathic, alpha-helical peptides, the phenol-soluble modulins (PSMs), and renders Rom an important tool for the investigation of membrane physiology.


Asunto(s)
Antiinfecciosos/farmacología , Membranas/efectos de los fármacos , Xantonas/farmacología , Animales , Células 3T3 BALB , Bacillus subtilis , Células Cultivadas , Células HeLa , Hemólisis/efectos de los fármacos , Humanos , Potenciales de la Membrana/efectos de los fármacos , Membranas/fisiología , Ratones , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus
9.
Cell Physiol Biochem ; 38(1): 359-67, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26824455

RESUMEN

BACKGROUND: The serum & glucocorticoid inducible kinase isoform SGK3 is a powerful regulator of several transporters, ion channels and the Na+/K+ ATPase. Targets of SGK3 include the ubiquitin ligase Nedd4-2, which is in turn a known regulator of the voltage gated K+ channel Kv1.5 (KCNA5). The present study thus explored whether SGK3 modifies the activity of the voltage gated K+ channel KCNA5, which participates in the regulation of diverse functions including atrial cardiac action potential, activity of vascular smooth muscle cells, insulin release and tumour cell proliferation. METHODS: cRNA encoding KCNA5 was injected into Xenopus oocytes with and without additional injection of cRNA encoding wild-type SGK3, constitutively active S419DSGK3, inactive K191NSGK3 and/or wild type Nedd4-2. Voltage gated K+ channel activity was quantified utilizing dual electrode voltage clamp. RESULTS: Voltage gated current in KCNA5 expressing Xenopus oocytes was significantly enhanced by wild-type SGK3 and S419DSGK3, but not by K191NSGK3. SGK3 was effective in the presence of ouabain (1 mM) and thus did not require Na+/K+ ATPase activity. Coexpression of Nedd4-2 decreased the voltage gated current in KCNA5 expressing Xenopus oocytes, an effect largely reversed by additional coexpression of SGK3. CONCLUSION: SGK3 is a positive regulator of KCNA5, which is at least partially effective by abrogating the effect of Nedd4-2.


Asunto(s)
Canal de Potasio Kv1.5/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Ubiquitina-Proteína Ligasas Nedd4 , Oocitos/metabolismo , Ouabaína/farmacología , Técnicas de Placa-Clamp , Proteínas Serina-Treonina Quinasas/genética , ARN Complementario/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Xenopus/crecimiento & desarrollo , Xenopus/metabolismo , Proteínas de Xenopus
10.
Kidney Blood Press Res ; 40(4): 403-12, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26227001

RESUMEN

BACKGROUND/AIMS: Epidemiological evidence suggests that vitamin D deficiency is associated with anemia. The potent metabolite 1,25(OH)2 vitamin D3 [1,25(OH)2D3] activates various signaling cascades regulating a myriad of cellular functions including suicidal cell death or apoptosis. Suicidal death of erythrocytes or eryptosis is characterized by cell shrinkage and cell membrane scrambling leading to phosphatidylserine (PS) externalization. Stimulation of eryptosis may limit lifespan of circulating erythrocytes and thus cause anemia. In the present study, we explored the effect of a high vitamin D diet (10,000 I.U. vitamin D for 14 days) in mice on eryptosis. METHODS: Plasma concentrations of erythropoietin were estimated using an immunoassay kit, blood count using an electronic hematology particle counter, relative reticulocyte numbers using Retic-COUNT® reagent, PS exposure at the cell surface from annexin V binding, cell volume from forward scatter, and cytosolic Ca(2+) ([Ca(2+)]i) from Fluo3-fluorescence in FACS analysis. RESULTS: Vitamin D treatment decreased mean corpuscular volume, reticulocyte count, and plasma erythropoietin levels. Vitamin D treatment slightly but significantly decreased forward scatter but did not significantly modify spontaneous PS exposure and [Ca(2+)]i of freshly drawn erythrocytes. Vitamin D treatment augmented the stimulation of PS exposure and cell shrinkage following exposure to hyperosmotic shock (addition of 550 mM sucrose) or energy depletion (glucose removal) without significantly modifying [Ca(2+)]i. CONCLUSIONS: The present observations point to a subtle effect of exogenous vitamin D supplementation on erythrocyte survival.


Asunto(s)
Envejecimiento Eritrocítico/efectos de los fármacos , Vitamina D/uso terapéutico , Vitaminas/uso terapéutico , Animales , Recuento de Células Sanguíneas , Calcio/metabolismo , Tamaño de la Célula/efectos de los fármacos , Dieta , Membrana Eritrocítica/efectos de los fármacos , Eritropoyetina/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Presión Osmótica/efectos de los fármacos , Fosfatidilserinas/sangre
11.
Nephron ; 130(3): 221-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26112741

RESUMEN

BACKGROUND/AIMS: Kinases involved in the regulation of epithelial transport include SPAK (SPS1-related proline/alanine-rich kinase) and OSR1 (oxidative stress-responsive kinase 1). SPAK and OSR1 are both regulated by WNK (with-no-K(Lys)) kinases. The present study explored whether SPAK and/or OSR1 influence the excitatory amino acid transporter EAAT3, which accomplishes glutamate and aspartate transport in kidney, intestine and brain. METHODS: cRNA encoding EAAT3 was injected into Xenopus laevis oocytes with or without additional injection of cRNA encoding wild-type SPAK, constitutively active (T233E)SPAK, WNK insensitive (T233A)SPAK, catalytically inactive (D212A)SPAK, wild-type OSR1, constitutively active (T185E)OSR1, WNK insensitive (T185A)OSR1 and catalytically inactive (D164A)OSR1. Glutamate-induced current was taken as measure of electrogenic glutamate transport and was quantified utilizing dual electrode voltage clamp. Furthermore, Ussing chamber was employed to determine glutamate transport in the intestine from gene-targeted mice carrying WNK insensitive SPAK (spak(tg/tg)) and from corresponding wild-type mice (spak(+/+)). RESULTS: EAAT3 activity was significantly decreased by wild-type SPAK and (T233E)SPAK, but not by (T233A)SPAK and (D212A)SPAK. SPAK decreased maximal transport rate without affecting significantly affinity of the carrier. Similarly, EAAT3 activity was significantly downregulated by wild-type OSR1 and (T185E)OSR1, but not by (T185A)OSR1 and (D164A)OSR1. Again OSR1 decreased maximal transport rate without affecting significantly affinity of the carrier. Intestinal electrogenic glutamate transport was significantly lower in spak(+/+) than in spak(tg/tg) mice. CONCLUSION: Both, SPAK and OSR1 are negative regulators of EAAT3 activity.


Asunto(s)
Transportador 3 de Aminoácidos Excitadores/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Ácido Aspártico/metabolismo , Transportador 3 de Aminoácidos Excitadores/genética , Ácido Glutámico/metabolismo , Humanos , Ratones , Oocitos/metabolismo , Técnicas de Placa-Clamp , Proteínas Serina-Treonina Quinasas/genética , ARN Complementario/biosíntesis , ARN Complementario/genética , Agua/metabolismo , Xenopus laevis
12.
PLoS One ; 10(6): e0129365, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26046984

RESUMEN

USP18 (Ubiquitin-like specific protease 18) is an enzyme cleaving ubiquitin from target proteins. USP18 plays a pivotal role in antiviral and antibacterial immune responses. On the other hand, ubiquitination participates in the regulation of several ion channels and transporters. USP18 sensitivity of transporters has, however, never been reported. The present study thus explored, whether USP18 modifies the activity of the peptide transporters PEPT1 and PEPT2, and whether the peptide transporters are sensitive to the ubiquitin ligase Nedd4-2. To this end, cRNA encoding PEPT1 or PEPT2 was injected into Xenopus laevis oocytes without or with additional injection of cRNA encoding USP18. Electrogenic peptide (glycine-glycine) transport was determined by dual electrode voltage clamp. As a result, in Xenopus laevis oocytes injected with cRNA encoding PEPT1 or PEPT2, but not in oocytes injected with water or with USP18 alone, application of the dipeptide gly-gly (2 mM) was followed by the appearance of an inward current (Igly-gly). Coexpression of USP18 significantly increased Igly-gly in both PEPT1 and PEPT2 expressing oocytes. Kinetic analysis revealed that coexpression of USP18 increased maximal Igly-gly. Conversely, overexpression of the ubiquitin ligase Nedd4-2 decreased Igly-gly. Coexpression of USP30 similarly increased Igly-gly in PEPT1 expressing oocytes. In conclusion, USP18 sensitive cellular functions include activity of the peptide transporters PEPT1 and PEPT2.


Asunto(s)
Dipéptidos/metabolismo , Endopeptidasas/metabolismo , Simportadores/metabolismo , Animales , Transporte Biológico , Dipéptidos/farmacología , Endopeptidasas/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Femenino , Humanos , Inyecciones , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Mediciones Luminiscentes/métodos , Potenciales de la Membrana/efectos de los fármacos , Ubiquitina-Proteína Ligasas Nedd4 , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Oocitos/fisiología , Técnicas de Placa-Clamp , Transportador de Péptidos 1 , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , ARN Complementario/administración & dosificación , ARN Complementario/genética , Conejos , Simportadores/genética , Ubiquitina Tiolesterasa , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Xenopus , Xenopus laevis
13.
Amino Acids ; 47(9): 1703-13, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25929587

RESUMEN

Homoarginine (hArg) is an endogenous, nonproteinogenic amino acid which differs from arginine by an additional methylene (CH2) group in the backbone. In this brief narrative review, we summarize the current literature on hArg in the renal and cardiovascular systems. Epidemiological studies have identified low hArg levels as an independent risk marker for cardiovascular, cerebrovascular, and renal diseases as well as for mortality. The relatively low correlation of hArg with established cardiovascular risk factors underlines its great potential as an emerging biomarker to improve risk prediction because plasma hArg concentrations might reflect previously unrecognized pathophysiological processes. hArg may be involved in the pathogenesis of various diseases due to its effects on nitric oxide (NO) and energy metabolism. In view of its structural similarities with arginine, it has been proposed that hArg impacts on arginine metabolism and subsequently also on NO synthesis. The key enzyme for hArg synthesis, arginine:glycine amidinotransferase (AGAT), is involved in the synthesis of energy metabolites including guanidinoacetate, the precursor of creatine. Therefore, the involvement of hArg in energy metabolism could partially explain the close association between hArg and cardiovascular diseases such as heart failure. Whether hArg supplementation or modification of key enzymes of hArg metabolism such as AGAT activity is effective for the treatment of chronic diseases remains to be elucidated.


Asunto(s)
Trastornos Cerebrovasculares/sangre , Metabolismo Energético , Insuficiencia Cardíaca/sangre , Homoarginina/sangre , Enfermedades Renales/sangre , Amidinotransferasas/metabolismo , Animales , Biomarcadores/sangre , Trastornos Cerebrovasculares/mortalidad , Glicina/análogos & derivados , Glicina/sangre , Insuficiencia Cardíaca/mortalidad , Humanos , Enfermedades Renales/mortalidad , Óxido Nítrico/sangre
14.
Kidney Blood Press Res ; 40(2): 153-65, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25871296

RESUMEN

BACKGROUND/AIMS: Since the discovery of FGF23, secondary hyperparathyroidism (SHPT) in renal disease has been considered to result primarily from phosphorus retention rather than vitamin D deficiency. However, the impact of phosphorus restriction and vitamin D supplementation on SHPT is still ill defined. METHODS: We investigated the development of SHPT in a doxorubicin-induced proteinuric mouse model and tested different treatment strategies including a low phosphorus diet and substitution with native or active vitamin D in 129 S1/SvImJ wild-type mice. RESULTS: Development of SHPT at day 30 was strongly related to the magnitude of induced proteinuria. In mice with a proteinuria <100 mg/mg creatinine, SHPT was mild (PTH increase 2.4-fold), and serum levels of FGF23, phosphate and urea remained almost stable, whereas mice with heavy proteinuria (>100 mg/mg creatinine) developed marked SHPT (PTH increase 10.1-fold) accompanied by massive increase in FGF23 (27.0-fold increase), hyperphosphatemia (1.8-fold increase), renal failure (7.3-fold urea increase) and depletion of both 25-OH vitamin D and 1,25-OH vitamin D. Substitution with native or active vitamin D was unable to suppress SHPT, whereas a low-phosphorus diet (Pi content 0.013%) completely suppressed SHPT in mice with both mild and heavy proteinuria. CONCLUSIONS: The development of SHPT resulted from phosphate retention in this proteinuric model and could completely be suppressed with a low-phosphorus diet.


Asunto(s)
Hiperparatiroidismo Secundario/dietoterapia , Hiperparatiroidismo Secundario/tratamiento farmacológico , Fósforo , Proteinuria/dietoterapia , Vitamina D/uso terapéutico , Vitaminas/uso terapéutico , Animales , Doxorrubicina , Factor-23 de Crecimiento de Fibroblastos , Glucuronidasa/biosíntesis , Glucuronidasa/genética , Hiperparatiroidismo Secundario/inducido químicamente , Riñón/patología , Proteínas Klotho , Ratones , Proteinuria/inducido químicamente , Proteinuria/etiología , Insuficiencia Renal/prevención & control , Vitamina D3 24-Hidroxilasa/biosíntesis , Vitamina D3 24-Hidroxilasa/genética
15.
Endocrinology ; 155(10): 3899-908, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25057793

RESUMEN

Glucocorticoids (GCs) constitute a highly pleiotropic class of drugs predominantly employed in the treatment of inflammatory diseases. In our search for new mechanisms of action, we identified a hitherto unknown effect of GCs in the gastrointestinal tract. We found that oral administration of dexamethasone (Dex) to mice caused an enlargement of the stomach due to the induction of gastroparesis and that this effect was abolished in GR(dim) mice carrying the A458T mutation in the GC receptor (GR). Gastroparesis was unrelated to the enhanced gastric acid secretion observed after Dex treatment, although both effects were mediated by the same molecular mechanism of the GR. Using conditional GR-knockout mice, we could further rule out that GC effects on enterocytes or myeloid cells were involved in the induction of gastroparesis. In contrast, we found that Dex upregulated arginase 2 (Arg2) in the stomach both at the mRNA and protein level. This suggests that GC treatment leads to a depletion of l-arginine thereby impeding the production of nitric oxide (NO), which is required for gastric motility. We tested this hypothesis by supplementing the drinking water of the mice with exogenous l-arginine to compensate for the presumed shortage of this major substrate of NO synthases. Importantly, this measure completely prevented both the enlargement of the stomach and the induction of gastroparesis after Dex treatment. Our findings raise considerations of combining orally applied GCs with l-arginine to improve tolerability of GC treatment and provide a possible explanation for the antiemetic effects of GCs widely exploited in chemotherapy.


Asunto(s)
Arginina/deficiencia , Dexametasona/efectos adversos , Gastroparesia/inducido químicamente , Glucocorticoides/efectos adversos , Animales , Arginasa/genética , Arginasa/metabolismo , Dexametasona/administración & dosificación , Femenino , Gastroparesia/genética , Gastroparesia/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucocorticoides/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Regulación hacia Arriba/efectos de los fármacos
16.
Toxins (Basel) ; 6(1): 54-65, 2013 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-24368324

RESUMEN

The polyphenolic 1,2,3,4,6-penta-O-galloyl-beta-D-glucose from several medicinal herbs triggers apoptosis and has, thus, been proposed for treatment of malignancy. The substance is at least partially effective through caspase activation. In analogy to apoptosis of nucleated cells, erythrocytes may enter suicidal death or eryptosis, which is characterized by cell shrinkage and by phosphatidylserine translocation to the erythrocyte surface. Eryptosis is triggered by increase of cytosolic Ca2+-activity ([Ca2+]i). The sensitivity to [Ca2+]i is enhanced by ceramide. The present study explored whether penta-O-galloyl-ß-D-glucose stimulates eryptosis. Cell volume was estimated from forward scatter, phosphatidylserine exposure from annexin V binding, hemolysis from hemoglobin-release, [Ca2+]i from Fluo3-fluorescence and ceramide abundance from fluorescent antibodies. A 48-h exposure of human erythrocytes to penta-O-galloyl-ß-D-glucose significantly decreased forward scatter (50 µM) and significantly increased annexin V binding (10 µM). Up to 50 µM penta-O-galloyl-ß-D-glucose did not significantly modify [Ca2+]i. However, the effect of penta-O-galloyl-ß-D-glucose (25 µM) induced annexin V binding was slightly, but significantly, blunted by removal of extracellular Ca2+, pointing to sensitization of erythrocytes to the scrambling effect of Ca2+. Penta-O-galloyl-ß-D-glucose (25 µM) further increased ceramide formation. In conclusion, penta-O-galloyl-ß-D-glucose stimulates suicidal erythrocyte death or eryptosis, an effect partially due to stimulation of ceramide formation with subsequent sensitization of erythrocytes to Ca2+.


Asunto(s)
Apoptosis/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Hemólisis , Taninos Hidrolizables/farmacología , Anexina A5/metabolismo , Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Ceramidas/metabolismo , Eritrocitos/metabolismo , Humanos , Fosfatidilserinas/metabolismo , Polifenoles/farmacología
17.
Phytomedicine ; 20(12): 1119-23, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23827666

RESUMEN

Trans-cinnamaldehyde, a component of leaves from Cinnamomum osmophloeum kaneh, has been shown to counteract tumor growth. The substance exerts its effect at least in part by triggering apoptosis. The propapoptotic signaling involves altered gene expression and mitochondrial depolarization. In analogy to apoptosis of nucleated cells, erythrocytes may enter eryptosis, the suicidal death characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine-exposure at the cell surface. Triggers of eryptosis include increase of cytosolic Ca(2+)-activity ([Ca(2+)]i). The present study explored, whether trans-cinnamaldehyde triggers eryptosis. Cell volume has been estimated from forward scatter, phosphatidylserine-exposure from annexin-V-binding, hemolysis from hemoglobin release, and [Ca(2+)]i from Fluo3-fluorescence. A 48 h exposure to trans-cinnamaldehyde (30 µM) significantly decreased forward scatter and increased annexin-V-binding, effects paralleled by increase of [Ca(2+)]i. Trans-cinnamaldehyde exposure was followed by a slight but significant increase of hemolysis. Removal of extracellular Ca(2+) virtually abolished the effect of trans-cinnamaldehyde (30 µM) on annexin-V-binding. The present observations show that trans-cinnamaldehyde triggers suicidal death of erythrocytes, i.e. cells devoid of mitochondria and gene expression.


Asunto(s)
Acroleína/análogos & derivados , Anexina A5/metabolismo , Calcio/metabolismo , Eritrocitos/efectos de los fármacos , Acroleína/farmacología , Apoptosis/efectos de los fármacos , Calcio/análisis , Tamaño de la Célula/efectos de los fármacos , Eritrocitos/fisiología , Humanos , Fosfatidilserinas
18.
FEBS Lett ; 587(11): 1663-8, 2013 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23603386

RESUMEN

Klotho, a hormone and enzyme, is a powerful regulator of ageing and life span. Klotho deficiency leads to cardiac arrythmia and sudden cardiac death. We thus explored whether klotho modifies cardiac K(+)-channel hERG. Current was determined utilizing dual electrode voltage clamp and hERG protein abundance utilizing immunohistochemistry and chemiluminescence in Xenopus oocytes expressing hERG with or without klotho. Coexpression of klotho increased cell membrane hERG-protein abundance and hERG current at any given voltage without significantly modifying the voltage required to activate the channel. The effect of klotho coexpression was mimicked by recombinant klotho protein and reversed by ß-glucuronidase-inhibitor D-saccharic acid-1,4-lactone.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/fisiología , Glucuronidasa/metabolismo , Animales , Células Cultivadas , Canal de Potasio ERG1 , Ácido Glucárico/farmacología , Glucuronidasa/antagonistas & inhibidores , Glucuronidasa/genética , Activación del Canal Iónico , Proteínas Klotho , Lactonas/farmacología , Potenciales de la Membrana , Oocitos/fisiología , Técnicas de Placa-Clamp , ARN Complementario/genética , Xenopus
19.
Cell Physiol Biochem ; 30(4): 876-88, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22907570

RESUMEN

BACKGROUND: Sorafenib (Nexavar(®)), a polytyrosine kinase inhibitor, stimulates apoptosis and is thus widely used for chemotherapy in hepatocellular carcinoma (HCC). Hematological side effects of Nexavar(®) chemotherapy include anemia. Erythrocytes may undergo apoptosis-like suicidal death or eryptosis, which is characterized by cell shrinkage and phosphatidylserine-exposure at the cell surface. Signaling leading to eryptosis include increase in cytosolic Ca(2+)activity ([Ca(2+)](i)), formation of ceramide, ATP-depletion and oxidative stress. The present study explored, whether sorafenib triggers eryptosis in vitro and in vivo. METHODS: [Ca(2+)](i )was estimated from Fluo3-fluorescence, cell volume from forward scatter, phosphatidylserine-exposure from annexin-V-binding, hemolysis from hemoglobin release, ceramide with antibody binding-dependent fluorescence, cytosolic ATP with a luciferin-luciferase-based assay, and oxidative stress from 2',7' dichlorodihydrofluorescein diacetate (DCFDA) fluorescence. RESULTS: A 48 h exposure of erythrocytes to sorafenib (≥0.5 µM) significantly increased Fluo 3 fluorescence, decreased forward scatter, increased annexin-V-binding and triggered slight hemolysis (≥5 µM), but did not significantly modify ceramide abundance and cytosolic ATP. Sorafenib treatment significantly enhanced DCFDA-fluorescence and the reducing agents N-acetyl-L-cysteine and tiron significantly blunted sorafenib-induced phosphatidylserine exposure. Nexavar(®) chemotherapy in HCC patients significantly enhanced the number of phosphatidylserine-exposing erythrocytes. CONCLUSIONS: The present observations disclose novel effects of sorafenib, i.e. stimulation of suicidal erythrocyte death or eryptosis, which may contribute to the pathogenesis of anemia in Nexavar(®)-based chemotherapy.


Asunto(s)
Antineoplásicos/efectos adversos , Eritrocitos/efectos de los fármacos , Eritrocitos/patología , Niacinamida/análogos & derivados , Compuestos de Fenilurea/efectos adversos , Fosfatidilserinas/metabolismo , Inhibidores de Proteínas Quinasas/efectos adversos , Adenosina Trifosfato/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Ceramidas/metabolismo , Eritrocitos/citología , Eritrocitos/metabolismo , Hemólisis/efectos de los fármacos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Niacinamida/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Sorafenib
20.
J Nucl Med ; 53(5): 823-30, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22492733

RESUMEN

UNLABELLED: Noninvasive in vivo imaging of biologic processes using PET is an important tool in preclinical studies. We observed significant differences in 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) uptake in arthritic ankles and carcinomas between dynamic and static PET measurements when mice breathed oxygen. Thus, we suspected that air or oxygen breathing and the anesthesia protocol might influence (18)F-FLT tracer uptake. METHODS: We injected arthritic, healthy, and CT26 colon carcinoma-bearing mice with (18)F-FLT before static or dynamic small-animal PET measurements. The spontaneously oxygen- or air-breathing mice were kept conscious or anesthetized with ketamine and xylazine during (18)F-FLT uptake before the 10-min static PET measurements. For dynamic PET scans, mice were anesthetized during the entire measurement. (18)F-FLT uptake was reported in percentage injected dose per cubed centimeter by drawing regions of interest around ankles, carcinomas, and muscle tissue. Additionally, venous blood samples were collected before (18)F-FLT injection and after PET measurement to analyze pH, carbon dioxide partial pressure (pCO(2)), and lactate values. RESULTS: A significantly reduced (18)F-FLT uptake was measured in arthritic ankles and in CT26 colon carcinomas when the mice breathed oxygen and were conscious during tracer uptake, compared with mice that were anesthetized during (18)F-FLT uptake. Breathing air completely abolished this phenomenon. Analysis of blood samples that were obtained from the mice before (18)F-FLT injection and after the PET scan implicated respiratory acidosis that was induced by oxygen breathing and consciousness during tracer uptake. Acidosis was found to be the primary factor responsible for the reduced (18)F-FLT uptake, as reflected by increased pCO(2) and reduced pH and lactate values. CONCLUSION: Oxygen-breathing conscious mice sustained respiratory acidosis and, consequently, reduced cell proliferation and (18)F-FLT uptake in arthritic ankles and CT26 colon carcinomas. Thus, we suggest the use of air instead of oxygen breathing for (18)F-FLT PET measurements.


Asunto(s)
Artritis/metabolismo , Artefactos , Neoplasias del Colon/metabolismo , Didesoxinucleósidos/metabolismo , Oxígeno/farmacología , Respiración , Anestesia , Animales , Tobillo/diagnóstico por imagen , Artritis/inducido químicamente , Artritis/diagnóstico por imagen , Transporte Biológico/efectos de los fármacos , Dióxido de Carbono/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/diagnóstico por imagen , Neoplasias del Colon/patología , Estado de Conciencia , Modelos Animales de Enfermedad , Femenino , Glucosa-6-Fosfato Isomerasa/farmacología , Ketamina/farmacología , Ácido Láctico/metabolismo , Ratones , Ratones Endogámicos BALB C , Oxígeno/metabolismo , Tomografía de Emisión de Positrones , Xilazina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA