Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Kidney Blood Press Res ; 44(4): 715-726, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31430745

RESUMEN

BACKGROUND: Chronic kidney disease (CKD) reduces both Klotho expression and its shedding into circulation, an effect that accelerates progression and cardiovascular complications. However, the mechanisms that regulate Klotho release by the human kidney are still unknown. METHODS: We measured plasma Klotho across the kidney, splanchnic organs and lung in 22 patients (71 ± 2 years, estimated glomerular filtration rate [eGFR] 60 ± 5.4 mL/min 1.73 m2) during elective diagnostic cardiac catheterizations. RESULTS: Although the Klotho average renal vein concentrations were remarkably higher (by ∼9%) than arterial values, the kidney removed Klotho (or was at zero balance) in 7 subjects, indicating that the kidney contribution to systemic Klotho is not constant. Klotho fractional enrichment across the kidney was inversely related to plasma sodium (r = 0.43, p = 0.045) and acid uric acid levels (r = 0.38, p = 0.084) and directly, to renal oxygen extraction (r = 0.56, p = 0.006). In multivariate analysis, renal oxygen extraction was the only predictor of the enrichment of Klotho across the kidney, suggesting the dependence of renal Klotho release on tubular hypoxia or oxidative metabolism. Klotho balance was neutral across the lung. In patients with eGFR <60 mL/min, Klotho was also removed by splanchnic organs (single pass fractional extraction ∼11%). CONCLUSIONS: The present study identifies kidney oxygen uptake as a predictor of Klotho release, and splanchnic organs as a site for Klotho removal. This study provides new understanding of kidney Klotho release and suggests that modulating kidney oxygen metabolism could increase Klotho delivery, as an option to slow disease progression and blunt organ damage.


Asunto(s)
Glucuronidasa/metabolismo , Riñón/metabolismo , Oxígeno/metabolismo , Anciano , Femenino , Glucuronidasa/sangre , Humanos , Riñón/irrigación sanguínea , Proteínas Klotho , Masculino , Oxígeno/sangre , Sodio , Solubilidad , Circulación Esplácnica , Ácido Úrico
2.
Am J Physiol Renal Physiol ; 312(2): F254-F258, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27558560

RESUMEN

Fibroblast growth factor-23 (FGF-23) accumulates in blood of patients with chronic kidney disease (CKD) and is associated both with cardiovascular complications and disease progression. However, our knowledge of the sites and mechanisms that regulate plasma FGF-23 is still incomplete. We measured plasma intact FGF-23 across the kidney, splanchnic organs, and lung in 11 patients [estimated glomerular filtration rate (eGFR) 60 ± 6 ml/min] during elective diagnostic cardiac catheterizations. In these patients FGF-23 was removed by the kidney, with a fractional extraction (FE) of ∼22%. The FE of FGF-23 across the kidney was similar to that of creatinine (∼17%, P = NS). In addition, the FGF-23 FE by the kidney was significantly directly related to eGFR (r = 0.709 P = 0.018) and to kidney creatinine FE (r = 0.736 P = 0.013) but only as a trend to plasma phosphate levels (r = 0.55, P = 0.18). There was no difference in FGF-23 levels in blood perfusing splanchnic organs and cardiopulmonary bed. However, the arterial-venous difference of FGF-23 across the lung was directly related to FGF-23 pulmonary artery levels, suggesting that the lung, and possibly the heart, participate in the homeostasis of plasma FGF-23 when its systemic levels are increased. Our data show that the human kidney is the only site for FGF-23 removal from blood and suggest that FGF-23 is predominantly removed by glomerular filtration. The kidney ability to remove FGF-23 from the circulation likely accounts for the early increase in blood of FGF-23 in patients with CKD.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Riñón/metabolismo , Pulmón/metabolismo , Insuficiencia Renal Crónica/metabolismo , Circulación Esplácnica/fisiología , Anciano , Anciano de 80 o más Años , Cateterismo Cardíaco , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Humanos , Masculino , Persona de Mediana Edad , Insuficiencia Renal Crónica/sangre
3.
Kidney Int ; 88(6): 1419-1426, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26308671

RESUMEN

An emergent hypothesis is that a resistance to the anabolic drive by insulin may contribute to loss of strength and muscle mass in patients with chronic kidney disease (CKD). We tested whether insulin resistance extends to protein metabolism using the forearm perfusion method with arterial insulin infusion in 7 patients with CKD and metabolic acidosis (bicarbonate 19 mmol/l) and 7 control individuals. Forearm glucose balance and protein turnover (2H-phenylalanine kinetics) were measured basally and in response to insulin infused at different rates for 2 h to increase local forearm plasma insulin concentration by approximately 20 and 50 µU/ml. In response to insulin, forearm glucose uptake was significantly increased to a lesser extent (-40%) in patients with CKD than controls. In addition, whereas in the controls net muscle protein balance and protein degradation were decreased by both insulin infusion rates, in patients with CKD net protein balance and protein degradation were sensitive to the high (0.035 mU/kg per min) but not the low (0.01 mU/kg per min) insulin infusion. Besides blunting muscle glucose uptake, CKD and acidosis interfere with the normal suppression of protein degradation in response to a moderate rise in plasma insulin. Thus, alteration of protein metabolism by insulin may lead to changes in body tissue composition which may become clinically evident in conditions characterized by low insulinemia.

4.
J Clin Med ; 13(2)2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38256456

RESUMEN

Cellular senescence has emerged as an important driver of aging and age-related disease in the kidney. The activity of ß-galactosidase at pH 6 (SA-ß-Gal) is a classic maker of senescence in cellular biology; however, the predictive role of kidney tissue SA-ß-Gal on eGFR loss in chronic kidney disease (CKD) is still not understood. We retrospectively studied the expression of SA-ß-Gal in kidney biopsies obtained in a cohort [n = 22] of incident patients who were followed up for 3 years as standard of care. SA-ß-Gal staining was approximately fourfold higher in the tubular compartment of patients with CKD vs. controls [26.0 ± 9 vs. 7.4 ± 6% positive tubuli in patients vs. controls; p < 0.025]. Tubular expressions of SA-ß-Gal, but not proteinuria, at the time of biopsy correlated with eGFR loss at the follow up; moreover, SA-ß-Gal expression in more than 30% of kidney tubules was associated with fast progressive kidney disease. In conclusion, our study shows that SA-ß-Gal is upregulated in the kidney tubular compartment of adult patients affected by CKD and suggests that tubular SA-ß-Gal is associated with accelerated loss of renal function.

5.
Physiol Rep ; 11(6): e15573, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36945836

RESUMEN

Sites and mechanisms regulating the supply of homocysteine (Hcy) to the circulation are unexplored in humans. We studied the exchange of Hcy across the forearm in CKD patients (n = 17, eGFR 20 ± 2 ml/min), in hemodialysis (HD)-treated patients (n = 14) and controls (n = 9). Arterial Hcy was ~ 2.5 folds increased in CKD and HD patients (p < 0.05-0.03 vs. controls). Both in controls and in patients Hcy levels in the deep forearm vein were consistently greater (+~7%, p < 0.05-0.01) than the corresponding arterial levels, indicating the occurrence of Hcy release from muscle. The release of Hcy from the forearm was similar among groups. In all groups arterial Hcy varied with its release from muscle (p < 0.03-0.02), suggesting that muscle plays an important role on plasma Hcy levels. Forearm Hcy release was inversely related to folate plasma level in all study groups but neither to vitamin B12 and IL-6 levels nor to muscle protein net balance. These data indicate that the release of Hcy from peripheral tissue metabolism plays a major role in influencing its Hcy plasma levels in humans and patients with CKD, and that folate is a major determinant of Hcy release.


Asunto(s)
Insuficiencia Renal Crónica , Humanos , Insuficiencia Renal Crónica/terapia , Diálisis Renal , Ácido Fólico , Vitamina B 12 , Músculo Esquelético , Homocisteína
6.
J Basic Clin Physiol Pharmacol ; 34(6): 805-809, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37843253

RESUMEN

OBJECTIVES: Graft-versus-host disease (GVHD) of central nervous system is an atypical and rare manifestation of chronic GVHD, presenting with a heterogeneous spectrum of signs and symptoms. Diagnosis of neurological manifestations of GVHD can be highly challenging and remain associated with dismal prognosis, significant morbidity, and reduced quality of life. CASE PRESENTATION: In this report, we describe a 39-year-old woman developing neurological signs and symptoms 8 months after allogeneic HSCT magnetic resonance imaging showed multifocal hyperintense lesions involving the periventricular region and frontal subcortical white matter. There was no laboratory evidence of infective or malignant etiology, and the case was diagnosed as CNS-GVHD. The patient was treated with intravenous methylprednisolone pulse therapy and the clinical conditions gradually improved. After few months, patient symptoms progressed despite the addition of high-dose intravenous immunoglobulin, tacrolimus, and a new course of high dose steroids. To engage targeted therapy, the patient underwent brain biopsy that revealed a loss of myelin fibers, perivascular and diffuse infiltration of T cells, and macrophages associated with reactive gliosis, representing a demyelinating disease. We intensified treatment with cyclophosphamide and subsequently introduced ibrutinib as salvage strategy. Despite a magnetic resonance imaging showing great regression of the demyelinating lesions, patient's conditions deteriorated and she died 16 months after HSCT. CONCLUSIONS: CNS-GVHD is a rare complication of HSCT that is difficult to diagnose. Based on our experience, brain biopsy may represent a useful diagnostic tool when the clinical features of neurological symptoms are ambiguous or in patients without evidence of preceding chronic GVHD.


Asunto(s)
Síndrome de Bronquiolitis Obliterante , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Femenino , Humanos , Adulto , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Calidad de Vida , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Sistema Nervioso Central/patología , Tacrolimus/uso terapéutico
7.
Cancers (Basel) ; 15(22)2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-38001657

RESUMEN

We are currently witnessing a dramatic shift in our approach to the treatment of B-cell non-Hodgkin lymphoma (B-NHL). In the evolving clinical landscape, novel treatments for this clinically heterogeneous disease span a wide range of interventions, encompassing targeted agents, cell therapy approaches, and novel monoclonal antibodies (NMABs). Among these, the latter are likely to exert the most profound impact due to their distinctive high efficacy and versatile applicability. NMABs represent a heterogeneous group of agents, including naked antibodies, immunotoxins, and T-cell-engaging molecules. In recent times, several NMABs have either gained regulatory approval or are on the verge of introduction into clinical practice, addressing multiple therapeutic indications and treatment regimens. Their anticipated impact is expected to be broad, initially in the context of relapsed/refractory (R/R) disease and subsequently extending to early treatment lines. The scope of this review is to provide a comprehensive overview of the biological characteristics, clinical properties, efficacy, and toxicity profiles of NMABs that have recently been introduced or are nearing integration into clinical practice.

8.
Kidney Int ; 79(7): 773-82, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21228768

RESUMEN

Apoptosis and myostatin are major mediators of muscle atrophy and might therefore be involved in the wasting of uremia. To examine whether they are expressed in the skeletal muscle of patients with chronic kidney disease (CKD), we measured muscle apoptosis and myostatin mRNA and their related intracellular signal pathways in rectus abdominis biopsies obtained from 22 consecutive patients with stage 5 CKD scheduled for peritoneal dialysis. Apoptotic loss of myonuclei, determined by anti-single-stranded DNA antibody and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assays, was significantly increased three to fivefold, respectively. Additionally, myostatin and interleukin (IL)-6 gene expressions were significantly upregulated, whereas insulin-like growth factor-I mRNA was significantly lower than in controls. Phosphorylated JNK (c-Jun amino-terminal kinase) and its downstream effector, phospho-c-Jun, were significantly upregulated, whereas phospho-Akt was markedly downregulated. Multivariate analysis models showed that phospho-Akt and IL-6 contributed individually and significantly to the prediction of apoptosis and myostatin gene expression, respectively. Thus, our study found activation of multiple pathways that promote muscle atrophy in the skeletal muscle of patients with CKD. These pathways appear to be associated with different intracellular signals, and are likely differently regulated in patients with CKD.


Asunto(s)
Apoptosis , Enfermedades Renales/complicaciones , Atrofia Muscular/etiología , Miostatina/genética , ARN Mensajero/análisis , Recto del Abdomen/química , Recto del Abdomen/patología , Anciano , Análisis de Varianza , Biopsia , Western Blotting , Estudios de Casos y Controles , Enfermedad Crónica , Femenino , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Factor I del Crecimiento Similar a la Insulina/genética , Interleucina-6/genética , Italia , Proteínas Quinasas JNK Activadas por Mitógenos/análisis , Enfermedades Renales/genética , Enfermedades Renales/patología , Masculino , Persona de Mediana Edad , Atrofia Muscular/genética , Atrofia Muscular/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/análisis , Proteínas Proto-Oncogénicas c-jun/análisis , Análisis de Regresión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Regulación hacia Arriba
9.
Gastroenterology ; 138(4): 1557-65, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20026114

RESUMEN

BACKGROUND & AIMS: Although metabolic acidosis stimulates protein catabolism, its effects on splanchnic protein turnover and energy expenditure have not been measured in human beings. We investigated the effects of chronic metabolic acidosis (CMA) on splanchnic protein dynamics and oxygen consumption in human beings by using a leucine tracer and mass-balance techniques. METHODS: Five subjects were studied after 6 days of HCl-, CaCl(2)-, and NH(4)Cl-induced acidosis; 8 subjects served as controls. Blood samples were collected from the radial artery and the hepatic veins. Measurements were performed on plasma and whole-blood samples. RESULTS: Based on plasma measurements, subjects who had undergone CMA had lower rates of splanchnic proteolysis (-35%) and protein synthesis (-50%; P < .05) than controls, as well as a negative leucine kinetic balance (-6.81 +/- 2.48 micromol/kg/min/1.73 m(2) body surface [BS](-1)), compared with the neutral balance in control plasma samples (0.76 +/- 2.11 micromol/kg/min/1.73; P < .05 between groups). Based on measurements from whole blood, splanchnic proteolysis and protein synthesis did not differ significantly between CMA and control samples, and the net leucine kinetic balance was neutral in both groups (CMA, -0.69 +/- 1.57; controls, -0.74 +/- 3.45 micromol/kg/min/1.73). In CMA whole-blood measurements, splanchnic oxygen consumption (44.8 +/- 4.3 mL/min/1.73 m(2) BS) was slightly lower than in controls (57.5 +/- 8.4 mL/min/1.73 m(2) BS; P = NS). Splanchnic protein synthesis correlated with oxygen consumption (r = 0.82; P < .001). CONCLUSIONS: CMA reduces splanchnic protein turnover and results in a negative leucine balance--an effect that apparently is offset by the contribution of blood cells to organ leucine (and protein) dynamics. Protein synthesis is a major contributor (about 67%) to energy expenditure in splanchnic organs.


Asunto(s)
Acidosis/metabolismo , Mesenterio/metabolismo , Consumo de Oxígeno , Proteínas/metabolismo , Adulto , Amoníaco/metabolismo , Enfermedad Crónica , Femenino , Humanos , Leucina/metabolismo , Masculino , Persona de Mediana Edad
10.
Kidney Int ; 74(7): 937-45, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18633341

RESUMEN

Resistance to the anabolic action of growth hormone may contribute to the loss of strength and muscle mass in adult patients with chronic kidney disease. We tested this hypothesis by infusing growth hormone in patients to levels necessary to saturate hormone receptors. This led to a significant decrease of plasma potassium and amino acid levels in control and hyperkalemic patients with chronic kidney disease. These effects were completely or partially blunted in patients with elevated C-reactive protein levels. In forearm perfusion studies, growth hormone caused a further decrease in the negative potassium and protein balance of hemodialysis patients without inflammation but no effect was seen in patients with inflammation. Only IL-6 levels and age were found to be independent correlates in these growth hormone-induced variations in plasma potassium and blood amino acids. This shows that although a resistance to pharmacologic doses of growth hormone is not a general feature of patients with chronic kidney disease, there is a subgroup characterized by blunted growth hormone action. Our results support the hypothesis that uremia with inflammation, but not uremia per se, inhibits downstream growth hormone signaling contributing to muscle atrophy.


Asunto(s)
Resistencia a Medicamentos , Hormona de Crecimiento Humana/farmacocinética , Inflamación , Enfermedades Renales/patología , Uremia , Factores de Edad , Anciano , Aminoácidos/sangre , Estudios de Casos y Controles , Enfermedad Crónica , Femenino , Hormona de Crecimiento Humana/administración & dosificación , Humanos , Interleucina-6/sangre , Enfermedades Renales/complicaciones , Masculino , Persona de Mediana Edad , Atrofia Muscular/etiología , Potasio/sangre
11.
Kidney Int Rep ; 3(3): 701-710, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29854979

RESUMEN

INTRODUCTION: Early studies have shown that patients with chronic kidney disease (CKD) are able to maintain nitrogen balance despite significantly lower protein intake, but how and to what extent muscle protein metabolism adapts to a low-protein diet (LPD) or to a supplemented very LPD (sVLPD) is still unexplored. METHODS: We studied muscle protein turnover by the forearm perfusion method associated with the kinetics of 2H-phenylalanine in patients with CKD: (i) in a parallel study in subjects randomized to usual diet (1.1 g protein/kg, n = 5) or LPD (0.55 g protein/kg, n = 6) (Protocol 1); (ii) in a crossover, self-controlled study in subjects on a 0.55 g/kg LPD followed by a sVLPD (0.45 g/kg + amino/ketoacids 0.1 g/kg, n = 6) (Protocol 2). RESULTS: As compared with a 1.1 g/kg containing diet, a 0.55 g/kg LPD induced the following: (i) a 17% to 40% decrease in muscle protein degradation and net protein balance, respectively, (ii) no change in muscle protein synthesis, (iii) a slight (by approximately 7%, P < 0.06) decrease in whole-body protein degradation, and (iv) an increase in the efficiency of muscle protein turnover. As compared with an LPD, an sVLPD induced the following: (i) no change in muscle protein degradation, and (ii) an approximately 50% decrease in the negative net protein balance, and an increase in the efficiency of muscle protein turnover. CONCLUSION: The results of these studies indicate that in patients with CKD the adaptation of muscle protein metabolism to restrained protein intake can be obtained via combined responses of protein degradation and the efficiency of recycling of amino acids deriving from protein breakdown.

12.
G Ital Nefrol ; 35(4)2018 Jul.
Artículo en Italiano | MEDLINE | ID: mdl-30035443

RESUMEN

A changing paradigm of treatment of kidney transplant recipients is a new, wider approach to immunosuppression, which should take into account both antiviral and anticancer effects, in addition to cardiovascular protection. Recent observations suggest that the early introduction of mammalian target of rapamycin inhibitors (mTORi) in association with low dose CNI may offer many of these effects. The present manuscript summarizes benefits and contraindications of combinations with mTORi in kidney transplant immunosuppressive strategies.


Asunto(s)
Terapia de Inmunosupresión/métodos , Trasplante de Riñón , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Humanos , Complicaciones Posoperatorias/prevención & control , Virosis/prevención & control
13.
J Cachexia Sarcopenia Muscle ; 8(1): 131-144, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27897392

RESUMEN

BACKGROUND: Inflammation in skeletal muscle is implicated in the pathogenesis of insulin resistance and cachexia but why uremia up-regulates pro-inflammatory cytokines is unknown. Toll-like receptors (TLRs) regulate locally the innate immune responses, but it is unknown whether in chronic kidney disease (CKD) TLR4 muscle signalling is altered. The aim of the study is to investigate whether in CKD muscle, TLRs had abnormal function and may be involved in transcription of pro-inflammatory cytokine. METHODS: TLR4, phospho-p65, phospho-ikBα, tumour necrosis factor (TNF)-α, phospho p38, Murf 1, and atrogin were studied in skeletal muscle from nondiabetic CKD stage 5 patients (n = 29) and controls (n = 14) by immunohistochemistry, western blot, and RT-PCR. Muscle cell cultures (C2C12) exposed to uremic serum were employed to study TLR4 expression (western blot and RT-PCR) and TLR-driven signalling. TLR4 signalling was abrogated by a small molecule chemical inhibitor or TLR4 siRNA. Phospho AKT and phospho p38 were evaluated by western blot. RESULTS: CKD subjects had elevated TLR4 gene and protein expression. Also expression of NFkB, p38 MAPK and the NFkB-regulated gene TNF-α was increased. At multivariate analysis, TLR4 protein content was predicted by eGFR and Subjective Global Assessment, suggesting that the progressive decline in renal function and wasting mediate TLR4 activation. In C2C12, uremic serum increased TLR4 as well as TNF-α and down-regulated pAkt. These effects were prevented by blockade of TLR4. CONCLUSIONS: CKD promotes muscle inflammation through an up-regulation of TLR4, which may activate downward inflammatory signals such as TNF-α and NFkB-regulated genes.


Asunto(s)
Recto del Abdomen/metabolismo , Insuficiencia Renal Crónica/metabolismo , Receptor Toll-Like 4/metabolismo , Adiponectina/sangre , Adulto , Anciano , Anciano de 80 o más Años , Animales , Proteína C-Reactiva/análisis , Línea Celular , Citocinas/sangre , Citocinas/genética , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Leptina/sangre , Masculino , Ratones , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Insuficiencia Renal Crónica/genética , Resistina/sangre , Transducción de Señal , Receptor Toll-Like 4/genética , Factor de Transcripción ReIA/metabolismo , Uremia/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Semin Nephrol ; 26(1): 3-7, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16412817

RESUMEN

Plasma homocysteine (Hcy) levels are increased significantly in patients with moderate renal failure and increase markedly in patients with end-stage renal disease. An increase in plasma Hcy level theoretically could be caused by an increased production rate (ie, transmethylation), a decreased rate of removal by transsulfuration or remethylation, or a decrease in the excretion of Hcy. Current evidence indicates that the major mechanism for hyperhomocysteinemia in renal failure is a decrease in Hcy removal from the body. However, it is debated whether this effect is the result of a decrease in the renal metabolic clearance or a result of extrarenal metabolic changes. The human kidney plays a major role in the removal of several aminothiols or Hcy-related compounds from the circulation (eg, cysteine-glycine, glutathione, AdoMet, and AdoHcy). However, the glomerular filtration of Hcy seems to be restricted because of protein binding. Besides glomerular filtration, the normal kidney can remove Hcy by plasma flow and peritubular uptake. Although in the low normal range in absolute terms, the flow through the transsulfuration pathway is reduced if related to Hcy levels in uremia; in addition, the remethylation pathway also is impaired. Besides the potential effect of the reduced renal mass on Hcy removal, available evidence suggests the occurrence of a generalized down-regulation of the methionine cycle and catabolism in uremia. AdoHcy, sulfate, and dimethylglycine currently are being investigated as retained solutes that can inhibit 1 or more pathways of Hcy metabolism. In addition, the high Hcy levels decrease in malnourished end-stage renal disease patients and change according to nutrient intake and several other nutritional parameters, indicating that circulating Hcy levels become an expression of nutritional status.


Asunto(s)
Hiperhomocisteinemia/etiología , Enfermedades Renales/complicaciones , Enfermedad Crónica , Humanos
15.
Int J Environ Res Public Health ; 8(5): 1631-54, 2011 05.
Artículo en Inglés | MEDLINE | ID: mdl-21655142

RESUMEN

Protein-energy wasting (PEW) is common in patients with chronic kidney disease (CKD) and is associated with an increased death risk from cardiovascular diseases. However, while even minor renal dysfunction is an independent predictor of adverse cardiovascular prognosis, PEW becomes clinically manifest at an advanced stage, early before or during the dialytic stage. Mechanisms causing loss of muscle protein and fat are complex and not always associated with anorexia, but are linked to several abnormalities that stimulate protein degradation and/or decrease protein synthesis. In addition, data from experimental CKD indicate that uremia specifically blunts the regenerative potential in skeletal muscle, by acting on muscle stem cells. In this discussion recent findings regarding the mechanisms responsible for malnutrition and the increase in cardiovascular risk in CKD patients are discussed. During the course of CKD, the loss of kidney excretory and metabolic functions proceed together with the activation of pathways of endothelial damage, inflammation, acidosis, alterations in insulin signaling and anorexia which are likely to orchestrate net protein catabolism and the PEW syndrome.


Asunto(s)
Desnutrición Proteico-Calórica/etiología , Insuficiencia Renal Crónica/complicaciones , Envejecimiento/metabolismo , Biomarcadores , Composición Corporal , Enfermedades Cardiovasculares/etiología , Proteínas en la Dieta/metabolismo , Humanos , Infecciones/etiología , Músculo Esquelético/fisiopatología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/mortalidad
16.
Clin Nutr ; 29(4): 424-33, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20207454

RESUMEN

The progressive loss of kidney function in patients with chronic kidney disease (CKD) is associated with a number of complications, including cardiovascular diseases, anemia, hyperparathyroidism, inflammation, metabolic acidosis, malnutrition and protein-energy wasting. The excess cardiovascular risk related to CKD is due in part to a higher prevalence of traditional atherosclerotic risk factors, in part to non-traditional, emerging risk factors peculiar to CKD. While even minor renal dysfunction is an independent predictor of adverse cardiovascular prognosis, nutritional changes are more often observed in an advanced setting. In addition, factors related to renal-replacement treatment may be implicated in the pathogenesis of heart disease and protein-energy wasting in dialysis-treated patients. Progressive alterations in kidney metabolism may cause progressive effects on cardiovascular status and nutrition. Altered kidney amino acid/protein metabolism and or excretion may be a key factor in the homeostasis of several vasoactive compounds and hormones in patients with more advanced disease. In this discussion recent research regarding the kidney handling of amino acids and protein turnover and their potential link with cardiovascular disease, progressive kidney dysfunction and nutritional status are reviewed.


Asunto(s)
Aminoácidos/metabolismo , Proteínas en la Dieta/metabolismo , Fallo Renal Crónico/metabolismo , Riñón/metabolismo , Proteínas/metabolismo , Animales , Proteínas en la Dieta/efectos adversos , Progresión de la Enfermedad , Humanos , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/fisiopatología , Fallo Renal Crónico/terapia , Diálisis Renal/efectos adversos , Diálisis Renal/métodos
17.
Metab Brain Dis ; 24(1): 159-67, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19083087

RESUMEN

Renal synthesis and excretion of ammonia are critical for efficient removal of acids from the body. Besides the rate of ammonia production, the intrarenal distribution of produced ammonia is a crucial step in the renal regulation of acid-base balance. Various acid-base disorders are associated not only with changes in ammonia production but also with its distribution between the urine and the renal veins. The final effect of ammonia production on acid-base balance largely depends on the events that determine the distribution of ammonia produced between urine and blood. Several factors, among which urine pH, urine flow, total ammonia production "per se" and renal blood flow may affect the percent of ammonia excreted into urines in humans with different acid-base disturbances. Among these factors, urine pH is the most important. An additional effect of stimulated ammoniagenesis is kidney hypertrophy. In tubule epithelial cells, the associated increase in ammonia production, rather than the acidosis per se, is responsible for favoring tubular hypertrophy. This effect is related to the inhibition of protein degradation, owing to changes in lysosomal pH and cathepsin activity, without effects on cell cycle. Both changes of PI-3 kinase pathway and the suppression of chaperone-mediated autophagy are candidate mechanism for ammonia-mediated inhibition of protein degradation in tubule cells. Available data in humans indicate that the response of kidney to metabolic acidosis includes both changes in amino acid uptake and suppression of protein degradation. The latter effect is associated with the increase in ammonia excretion and partition into the urine.


Asunto(s)
Equilibrio Ácido-Base/fisiología , Amoníaco/metabolismo , Riñón/metabolismo , Proteínas/metabolismo , Desequilibrio Ácido-Base/metabolismo , Desequilibrio Ácido-Base/fisiopatología , Animales , Humanos , Concentración de Iones de Hidrógeno , Túbulos Renales/metabolismo , Circulación Renal/fisiología , Transducción de Señal/fisiología , Orina/fisiología
18.
Cytokine ; 37(1): 51-4, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17420140

RESUMEN

Chronic elevation of circulating Interleukin-6 (IL-6) is observed in elderly individuals as well as in several illnesses, including chronic kidney diseases. A number of cells and tissues possess the ability to metabolize significant amounts of IL-6 in vitro. However, information on signals and mechanisms by which IL-6 is removed from blood in humans is still incomplete. To assess the individual role of splanchnic organs and kidney on IL-6 inter-organ exchange we used the IL-6 mass-balance technique across the hepato-splanchnic bed and kidney in six subjects with normal renal and liver function undergoing diagnostic venous catheterizations. Both in the hepatic and renal veins IL-6 levels were significantly lower (p=0.041 and 0.038, respectively), than in the artery. The fractional extraction of IL-6, i.e., the percentage of arterial IL-6 extracted after a single pass, was greater across the splanchnic organs (18%) than across the kidney (8%). Accordingly, IL-6 plasma clearance across splanchnic organs was greater than across the kidney and the sum of kidney and splanchnic removal accounted for as much as 63% of the estimated adipocyte IL-6 release. Our data demonstrate that, although the individual contribution to removal is different, both splanchnic organs and kidneys affect in a significant way the disposal of IL-6 in humans. According, both liver and kidney dysfunction could affect the handling of this proinflammatory cytokine and favour a chronic inflammatory response.


Asunto(s)
Interleucina-6/metabolismo , Riñón/metabolismo , Circulación Esplácnica , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad
19.
Am J Physiol Renal Physiol ; 283(6): F1430-7, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12388393

RESUMEN

To evaluate the role of blood cells in interorgan amino acid transport and in the estimates of regional protein turnover, we studied the effects of plasma vs. whole blood sampling on regional leucine kinetics in postabsorptive humans. Studies were carried out by combining the arteriovenous difference technique with the measurement of [14C]- and [15N]leucine isotope exchange across the human kidney, the splanchnic area, and the leg. In the kidney, whole blood-derived rates of leucine-carbon appearance, disappearance, and net balance (NB) were greater (by 3-15 times; P < 0.035) than those calculated in plasma. In addition, the net leucine-carbon (i.e., protein) balance across the kidney was negative in whole blood (-5.6 +/- 1.3 micromol/min x 1.73 m2, P < 0.01 vs. 0) but neutral in plasma [-0.24 +/- 1.33, P = not significant from 0; P < 0.01 vs. whole blood]. A net leucine transport out of renal cells was shown in blood but not in plasma. In contrast, rates of leucine-carbon appearance, disappearance, NB, and net transport, in both the splanchnic area and the leg, were similar in whole blood and plasma. These data suggest that blood cells play a key role in leucine transport out of the kidney and, consequently, in the leucine-derived estimates of renal protein degradation and NB, which is at variance with what is observed across the splanchnic organs or the leg. These data also emphasize the need for complete whole blood arteriovenous measurements to accurately estimate protein turnover across the kidney.


Asunto(s)
Células Sanguíneas/fisiología , Riñón/metabolismo , Leucina/farmacocinética , Adulto , Transporte Biológico/fisiología , Femenino , Arteria Femoral , Vena Femoral , Humanos , Cetoácidos/sangre , Cinética , Leucina/sangre , Masculino , Proteínas/metabolismo , Circulación Esplácnica
20.
J Am Soc Nephrol ; 11(11): 2106-2113, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11053487

RESUMEN

The relationships among growth hormone (GH), leptin, and resting energy expenditure (REE) are not understood. It has been reported that in malnourished hemodialysis patients, GH increases muscle protein synthesis, a process that requires energy. The present study evaluated the arterial levels and the forearm exchange of leptin, as well as the REE of the same patients during their participation in the same study, in four sequential 6-wk periods: I, baseline; II, GH treatment; III, washout; and IV, GH + intradialytic parenteral nutrition. During periods II and IV, patients received GH (5 mg three times per week). REE rose by 5% in period II, declined during period III, and rose by 7% during period IV. Basal leptin levels were low (2.0 +/- 0.19 ng/L). Insulin and leptin levels, as well as leptin release from the forearm, were unchanged during periods I through III but rose (+ 36%; P: < 0.05) during period IV. Changes in arterial leptin were directly related to changes in forearm leptin release (P: < 0.002), indicating a role of leptin production by peripheral tissues on leptinemia. Changes in leptin release were directly related to insulin (P: < 0.001) and, less consistently, to insulin-like growth factor-binding protein-1 levels (P: < 0.02). Similarly, variations in leptin levels were directly related to insulin (P: < 0.01). Variations in REE were not related to variations in leptin or insulin levels but to changes in muscle protein synthesis (P: < 0.025). The data show that in malnourished hemodialysis patients, treatment with GH is not invariably associated with an increase in leptin production. An increase in leptin release by peripheral tissues and leptin levels occurs only in the setting of hyperinsulinemia. The increase in REE that is induced by treatment with GH is not dependent on changes in leptin but is largely accounted for by the energy cost of the stimulation of muscle protein synthesis.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Hormona de Crecimiento Humana/uso terapéutico , Leptina/metabolismo , Desnutrición Proteico-Calórica/tratamiento farmacológico , Desnutrición Proteico-Calórica/metabolismo , Diálisis Renal , Estudios Cruzados , Femenino , Humanos , Insulina/sangre , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Leptina/sangre , Masculino , Persona de Mediana Edad , Nutrición Parenteral , Estudios Prospectivos , Desnutrición Proteico-Calórica/terapia , Descanso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA