Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Exp Nephrol ; 27(11): 961-971, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37578637

RESUMEN

BACKGROUND: Dialysis patients often take multiple oral medications, leading to a high pill burden. Phosphate binders (PBs) account for a large proportion of this daily pill burden (DPB). The relationship between DPB and mortality risk remains unclear, and we hypothesized that this relationship might be influenced by the proportion of PBs to all medications. METHODS: We divided DPB into those derived from PBs and non-PB drugs and analyzed the association with mortality risk over a 7-year period in 513 chronic hemodialysis patients using a baseline model. RESULTS: The median (interquartile range) DPB from all drugs was 15.8 (11.2-21.0) pills/day/patient, and the median ratio of PB pills to all drug pills was 29.3 (13.7-45.9)% at baseline. During a median observation period of 5.2 years, 161 patients (31.4%) died. Kaplan-Meier analysis showed no significant difference in all-cause mortality between PB users and non-users. However, a significant survival advantage was observed in the highest tertile of DPB from PBs compared to the lowest tertile. Conversely, the highest tertile of DPB from non-PB drugs was associated with worse survival. Consequently, the highest tertile of the ratio of PBs to all pills was associated with better survival. This association remained significant even after adjusting for patient characteristics in the Cox proportional hazards model. However, when serum nutritional parameters were included as covariates, the significant association disappeared. CONCLUSIONS: Dialysis patients prescribed a higher rate of PB pills to all medications exhibited a lower mortality risk, possibly due to their better nutritional status.


Asunto(s)
Fallo Renal Crónico , Diálisis Renal , Humanos , Diálisis Renal/efectos adversos , Estudios de Cohortes , Estudios de Seguimiento , Fósforo , Fosfatos
2.
Biomed Res Int ; 2014: 376191, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24883308

RESUMEN

Activin, a member of the TGF-ß superfamily, regulates cell growth and differentiation in various cell types. Activin A acts as a negative regulator of renal development as well as tubular regeneration after renal injury. However, it remains unknown whether activin A is involved in renal fibrosis. To clarify this issue, we utilized a rat model of unilateral ureteral obstruction (UUO). The expression of activin A was significantly increased in the UUO kidneys compared to that in contralateral kidneys. Activin A was detected in glomerular mesangial cells and interstitial fibroblasts in normal kidneys. In UUO kidneys, activin A was abundantly expressed by interstitial α-SMA-positive myofibroblasts. Administration of recombinant follistatin, an activin antagonist, reduced the fibrotic area in the UUO kidneys. The number of proliferating cells in the interstitium, but not in the tubules, was significantly lower in the follistatin-treated kidneys. Expression of α-SMA, deposition of type I collagen and fibronectin, and CD68-positive macrophage infiltration were significantly suppressed in the follistatin-treated kidneys. These data suggest that activin A produced by interstitial fibroblasts acts as a potent profibrotic factor during renal fibrosis. Blockade of activin A action may be a novel approach for the prevention of renal fibrosis progression.


Asunto(s)
Fibrosis/tratamiento farmacológico , Folistatina/administración & dosificación , Subunidades beta de Inhibinas/biosíntesis , Obstrucción Ureteral/tratamiento farmacológico , Animales , Proliferación Celular/efectos de los fármacos , Fibrosis/genética , Fibrosis/patología , Humanos , Subunidades beta de Inhibinas/antagonistas & inhibidores , Subunidades beta de Inhibinas/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Ratas , Factor de Crecimiento Transformador beta/genética , Obstrucción Ureteral/genética , Obstrucción Ureteral/patología
3.
Am J Physiol Renal Physiol ; 304(6): F665-73, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23324177

RESUMEN

N-type Ca(2+) channels are densely distributed in sympathetic nerves that innervate renal tubules. However, the role of N-type Ca(2+) channels in renal fibrosis remains unknown. To address this issue, we examined the difference between the effects of amlodipine (an L-type Ca(2+) channel blocker) and cilnidipine (a dual L/N-type Ca(2+) channel blocker) on fibrotic changes using a rat unilateral ureteral obstruction (UUO) model. The expression of both L-type and N-type Ca(2+) channels was significantly upregulated in UUO kidneys compared with that in contralateral kidneys. There were no significant differences in mean blood pressure among the rats tested. Both amlodipine and cilnidipine significantly attenuated fibrotic changes in UUO kidneys. The antifibrotic effect of cilnidipine was more potent than that of amlodipine. Amlodipine as well as cilnidipine reduced type III collagen deposition, α-smooth muscle actin (α-SMA) expression, and interstitial cell proliferation. In addition, cilnidipine significantly reduced deposition of type I collagen and macrophage infiltration in UUO kidneys. With the use of in vivo bromodeoxyuridine labeling, label-retaining cells (LRCs) were identified as a population of tubular cells that participate in epithelial-mesenchymal transition after UUO. Some LRCs migrated into the interstitium, expressed α-SMA and vimentin, and produced several extracellular matrixes in UUO kidneys. The number of interstitial LRCs was significantly decreased by cilnidipine but not amlodipine. These data suggest that N-type Ca(2+) channels contribute to multiple steps of renal fibrosis, and its blockade may thus be a useful therapeutic approach for prevention of renal fibrosis.


Asunto(s)
Amlodipino/uso terapéutico , Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo N/metabolismo , Dihidropiridinas/uso terapéutico , Obstrucción Ureteral/metabolismo , Actinas/metabolismo , Amlodipino/farmacología , Animales , Cadherinas/metabolismo , Canales de Calcio Tipo L/metabolismo , Proliferación Celular/efectos de los fármacos , Dihidropiridinas/farmacología , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Riñón/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Obstrucción Ureteral/tratamiento farmacológico
4.
Am J Physiol Renal Physiol ; 302(6): F694-702, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22169012

RESUMEN

Recovery after acute kidney injury is impaired in the elderly, but the precise mechanism for such age-related incompetence remains unclear. By in vivo bromodeoxyuridine (BrdU) labeling, renal progenitor cells (label-retaining cells; LRCs) were identified in tubules of normal rat kidney and were shown to be the origin of proliferating cells after injury. In the present study, the involvement of LRCs in the age-related decline of tubular recovery after injury was examined. After 1 wk of BrdU labeling followed by a 2-wk chase period, ischemia-reperfusion injury was induced in 7-wk-, 7-mo-, and 12-mo-old rats. Age-related decreases in DNA synthesis and cell proliferation in renal tubules after injury were found. The number of LRCs also significantly declined with age. At 24 h after reperfusion, the number of LRCs significantly increased in all ages of rats tested. There was no significant difference in the ratio of LRC division among rats of different ages. The area of the rat endothelial cell antigen (RECA)-1-positive capillary network declined with age. When renal tubules isolated from rats treated with BrdU label were cocultured with human umbilical vein endothelial cells (HUVEC), the number of LRCs significantly increased compared with tubules cultured without HUVEC. These data suggest that the reduced capacity of tubular regeneration in the aging kidney is partly explained by the shortage of LRC reserves. The size of the LRC pool might be regulated by the surrounding peritubular capillary network.


Asunto(s)
Envejecimiento/fisiología , Riñón/citología , Riñón/fisiología , Regeneración/fisiología , Células Madre/fisiología , Animales , Bromodesoxiuridina , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales , Regulación de la Expresión Génica , Humanos , Riñón/lesiones , Masculino , Ratas , Ratas Wistar , Daño por Reperfusión , Coloración y Etiquetado
5.
Am J Physiol Renal Physiol ; 301(2): F387-95, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21561997

RESUMEN

Renal proximal tubular epithelium can regenerate after various insults. To examine whether the tubular repair process is regulated by surrounding peritubular capillaries, we established an in vitro human tubulogenesis model that mimics in vivo tubular regeneration after injury. In this model, HGF, a potent renotropic factor, dose dependently induced tubular structures in human renal proximal tubular epithelial cells cultured in gels. Consistent with regenerating tubular cells after injury, HGF-induced tubular structures expressed a developmental gene, Pax-2, and a mesenchymal marker, vimentin, and formed a lumen with aquaporin-1 expression. Electron microscopic analysis showed the presence of microvilli on the apical site of the lumen, suggesting that these structures are morphologically equivalent to renal tubules in vivo. When cocultured with human umbilical vein endothelial cells (HUVEC), HGF-induced tubular formation was significantly enhanced. This could not be reproduced by the addition of VEGF, basic FGF, or PDGF. Protein array revealed that HUVEC produced various matrix metalloproteinases (MMPs). The stimulatory effects of coculture with HUVEC or HUVEC-derived conditional medium were almost completely abolished by addition of the tissue inhibitor of metalloproteinase (TIMP)-1 or TIMP-2. These data suggest that endothelial cell-derived factors including MMPs play a critical role in tubulogenesis and imply a potential role of peritubular capillary endothelium as a source of factor(s) required for tubular recovery after injury.


Asunto(s)
Factor de Crecimiento de Hepatocito/fisiología , Túbulos Renales Proximales/fisiología , Regeneración , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Isquemia/patología , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/patología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratas , Ratas Wistar
6.
Endocr J ; 55(1): 1-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17827789

RESUMEN

The research described in this review suggests a novel and important role for activin A in the developmental and repair processes of the kidney (Table 1). The results obtained in these studies indicate that activin A is a negative regulator of kidney development and plays an essential part in kidney diseases, such as acute renal failure or renal fibrosis. It is also possible that activin A is a key player in the pathophysiological processes of other kidney diseases, such as congenital urogenital abnormalities, renal cystic disease and renal cell carcinoma. Activin A is thus a potential target for therapeutic interventions in kidney diseases. To address this issue, more detailed analysis on the regulation of activin production, modulation of activin activity and activin target genes is required.


Asunto(s)
Activinas/fisiología , Riñón/embriología , Riñón/fisiología , Regeneración/fisiología , Receptores de Activinas/fisiología , Animales , Comunicación Autocrina/fisiología , Folistatina/fisiología , Humanos , Enfermedades Renales/etiología , Modelos Biológicos , Organogénesis/genética , Organogénesis/fisiología , Uréter/embriología , Conductos Mesonéfricos/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...