Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Kidney Int ; 101(6): 1232-1250, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35276205

RESUMO

The molecular circadian clock is an evolutionary adaptation to anticipate recurring changes in the environment and to coordinate variations in activity, metabolism and hormone secretion. Parathyroid hyperplasia in uremia is a significant clinical challenge. Here, we examined changes in the transcriptome of the murine parathyroid gland over 24 hours and found a rhythmic expression of parathyroid signature genes, such as Casr, Vdr, Fgfr1 and Gcm2. Overall, 1455 genes corresponding to 6.9% of all expressed genes had significant circadian rhythmicity. Biological pathway analysis indicated that the circadian clock system is essential for the regulation of parathyroid cell function. To study this, a novel mouse strain with parathyroid gland-specific knockdown of the core clock gene Bmal1 (PTHcre;Bmal1flox/flox) was created. Dampening of the parathyroid circadian clock rhythmicity was found in these knockdown mice, resulting in abrogated rhythmicity of regulators of parathyroid cell proliferation such as Sp1, Mafb, Gcm2 and Gata3, indicating circadian clock regulation of these genes. Furthermore, the knockdown resulted in downregulation of genes involved in mitochondrial function and synthesis of ATP. When superimposed by uremia, these PTHcre;Bmal1flox/flox mice had an increased parathyroid cell proliferative response, compared to wild type mice. Thus, our findings indicate a role of the internal parathyroid circadian clock in the development of parathyroid gland hyperplasia in uremia.


Assuntos
Relógios Circadianos , Uremia , Animais , Proliferação de Células , Relógios Circadianos/genética , Ritmo Circadiano/genética , Regulação da Expressão Gênica , Hiperplasia , Camundongos , Glândulas Paratireoides , Uremia/genética
2.
Int J Mol Sci ; 24(1)2022 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-36613582

RESUMO

Nicotinamide adenine dinucleotide (oxidized form, NAD+) is a critical coenzyme, with functions ranging from redox reactions and energy metabolism in mitochondrial respiration and oxidative phosphorylation to being a central player in multiple cellular signaling pathways, organ resilience, health, and longevity. Many of its cellular functions are executed via serving as a co-substrate for sirtuins (SIRTs), poly (ADP-ribose) polymerases (PARPs), and CD38. Kidney damage and diseases are common in the general population, especially in elderly persons and diabetic patients. While NAD+ is reduced in acute kidney injury (AKI) and chronic kidney disease (CKD), mounting evidence indicates that NAD+ augmentation is beneficial to AKI, although conflicting results exist for cases of CKD. Here, we review recent progress in the field of NAD+, mainly focusing on compromised NAD+ levels in AKI and its effect on essential cellular pathways, such as mitochondrial dysfunction, compromised autophagy, and low expression of the aging biomarker αKlotho (Klotho) in the kidney. We also review the compromised NAD+ levels in renal fibrosis and senescence cells in the case of CKD. As there is an urgent need for more effective treatments for patients with injured kidneys, further studies on NAD+ in relation to AKI/CKD may shed light on novel therapeutics.


Assuntos
Injúria Renal Aguda , Insuficiência Renal Crônica , Sirtuínas , Humanos , Metabolismo Energético , NAD/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Sirtuínas/metabolismo
3.
Kidney Int ; 98(6): 1461-1475, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32721445

RESUMO

Circadian rhythms in metabolism, hormone secretion, cell cycle and locomotor activity are regulated by a molecular circadian clock with the master clock in the suprachiasmatic nucleus of the central nervous system. However, an internal clock is also expressed in several peripheral tissues. Although about 10% of all genes are regulated by clock machinery an internal molecular circadian clock in the parathyroid glands has not previously been investigated. Parathyroid hormone secretion exhibits a diurnal variation and parathyroid hormone gene promoter contains an E-box like element, a known target of circadian clock proteins. Therefore, we examined whether an internal molecular circadian clock is operating in parathyroid glands, whether it is entrained by feeding and how it responds to chronic kidney disease. As uremia is associated with extreme parathyroid growth and since disturbed circadian rhythm is related to abnormal growth, we examined the expression of parathyroid clock and clock-regulated cell cycle genes in parathyroid glands of normal and uremic rats. Circadian clock genes were found to be rhythmically expressed in normal parathyroid glands and this clock was minimally entrained by feeding. Diurnal regulation of parathyroid glands was next examined. Significant rhythmicity of fibroblast-growth-factor-receptor-1, MafB and Gata3 was found. In uremic rats, deregulation of circadian clock genes and the cell cycle regulators, Cyclin D1, c-Myc, Wee1 and p27, which are influenced by the circadian clock, was found in parathyroid glands as well as the aorta. Thus, a circadian clock operates in parathyroid glands and this clock and downstream cell cycle regulators are disturbed in uremia and may contribute to dysregulated parathyroid proliferation in secondary hyperparathyroidism.


Assuntos
Relógios Circadianos , Ritmo Circadiano , Insuficiência Renal Crônica , Animais , Doenças Ósseas , Relógios Circadianos/genética , Ritmo Circadiano/genética , Minerais , Glândulas Paratireoides , Ratos , Insuficiência Renal Crônica/genética
4.
Pflugers Arch ; 471(8): 1079-1094, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31236663

RESUMO

Activin A is a new fascinating player in chronic kidney disease-mineral and bone disorder (CKD-MBD), which is implicated in progressive renal disease, vascular calcification, and osteodystrophy. Plasma activin A rises early in the progression of renal disease. Disruption of circadian rhythms is related to increased risk of several diseases and circadian rhythms are observed in mineral homeostasis, bone parameters, and plasma levels of phosphate and PTH. Therefore, we examined the circadian rhythm of activin A and CKD-MBD-related parameters (phosphate, PTH, FGF23, and klotho) in healthy controls and CKD rats (5/6 nephrectomy) on high-, standard- and low-dietary phosphate contents as well as during fasting conditions. Plasma activin A exhibited circadian rhythmicity in healthy control rats with fourfold higher values at acrophase compared with nadir. The rhythm was obliterated in CKD. Activin A was higher in CKD rats compared with controls when measured at daytime but not significantly when measured at evening/nighttime, stressing the importance of time-specific reference intervals when interpreting plasma values. Plasma phosphate, PTH, and FGF23 all showed circadian rhythms in control rats, which were abolished or disrupted in CKD. Plasma klotho did not show circadian rhythm. Thus, the present investigation shows, for the first time, circadian rhythm of plasma activin A. The rhythmicity is severely disturbed by CKD and is associated with disturbed rhythms of phosphate and phosphate-regulating hormones PTH and FGF23, indicating that disturbed circadian rhythmicity is an important feature of CKD-MBD.


Assuntos
Ativinas/sangue , Doenças Ósseas Metabólicas/sangue , Ritmo Circadiano , Fosfatos/sangue , Insuficiência Renal Crônica/sangue , Uremia/sangue , Animais , Doenças Ósseas Metabólicas/etiologia , Fatores de Crescimento de Fibroblastos/sangue , Glucuronidase/sangue , Proteínas Klotho , Masculino , Hormônio Paratireóideo/sangue , Ratos , Ratos Wistar , Insuficiência Renal Crônica/complicações , Uremia/etiologia
5.
Am J Physiol Renal Physiol ; 314(5): F753-F762, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29187373

RESUMO

In a new paradigm of etiology related to chronic kidney disease-mineral and bone disorder (CKD-MBD), kidney injury may cause induction of factors in the injured kidney that are released into the circulation and thereby initiate and maintain renal fibrosis and CKD-MBD. Klotho is believed to ameliorate renal fibrosis and CKD-MBD, while activin A might have detrimental effects. The unilateral ureter obstruction (UUO) model is used here to examine this concept by investigating early changes related to renal fibrosis in the obstructed kidney, untouched contralateral kidney, and vasculature which might be affected by secreted factors from the obstructed kidney, and comparing with unilateral nephrectomized controls (UNX). Obstructed kidneys showed early Klotho gene and protein depletion, whereas plasma Klotho increased in both UUO and UNX rats, indicating an altered metabolism of Klotho. Contralateral kidneys had no compensatory upregulation of Klotho and maintained normal expression of the examined fibrosis-related genes, as did remnant UNX kidneys. UUO caused upregulation of transforming growth factor-ß and induction of periostin and activin A in obstructed kidneys without changes in the contralateral kidneys. Plasma activin A doubled in UUO rats after 10 days while no changes were seen in UNX rats, suggesting secretion of activin A from the obstructed kidney with potentially systemic effects on CKD-MBD. As such, increased aortic sclerostin was observed in UUO rats compared with UNX and normal controls. The present results are in line with the new paradigm and show very early vascular effects of unilateral kidney fibrosis, supporting the existence of a new kidney-vasculature axis.


Assuntos
Injúria Renal Aguda/sangue , Distúrbio Mineral e Ósseo na Doença Renal Crônica/sangue , Glucuronidase/sangue , Subunidades beta de Inibinas/sangue , Rim/metabolismo , Obstrução Ureteral/sangue , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Aorta/metabolismo , Biomarcadores/sangue , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Distúrbio Mineral e Ósseo na Doença Renal Crônica/patologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/fisiopatologia , Modelos Animais de Doenças , Fibrose , Regulação da Expressão Gênica , Marcadores Genéticos/genética , Glucuronidase/genética , Subunidades beta de Inibinas/genética , Rim/patologia , Rim/fisiopatologia , Proteínas Klotho , Masculino , Ratos Wistar , Transdução de Sinais , Fatores de Tempo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Obstrução Ureteral/patologia , Obstrução Ureteral/fisiopatologia
6.
Calcif Tissue Int ; 102(1): 85-92, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29063159

RESUMO

The calcium and phosphate homeostasis is regulated by a complex interplay between parathyroid hormone (PTH), fibroblast growth factor 23 (FGF23), and calcitriol. Experimental studies have demonstrated an inhibitory effect of FG23 on PTH production and secretion; the physiological role of this regulation is however not well understood. Surprisingly, in uremia, concomitantly elevated FGF23 and PTH levels are observed. The parathyroid gland rapidly loses its responsiveness to extracellular calcium in vitro and a functional parathyroid cell line has currently not been established. Therefore, the aim of the present investigation was to study the impact of FGF23 on the Ca2+/PTH relationship in vivo under conditions of normocalcemia and hypocalcemia. Wistar rats were allocated to treatment with intravenous recombinant FGF23 and inhibition of the FGF receptor in the setting of normocalcemia and acute hypocalcemia. We demonstrated that FGF23 rapidly inhibited PTH secretion and that this effect was completely blocked by inhibition of the FGF receptor. Furthermore, inhibition of the FGF receptor by itself significantly increased PTH levels, indicating that FGF23 has a suppressive tonus on the parathyroid gland's PTH secretion. In acute hypocalcemia, there was no effect of either recombinant FGF23 or FGF receptor inhibition on the physiological response to the low ionized calcium levels. In conclusion, FGF23 has an inhibitory tonus on PTH secretion in normocalcemia and signals through the FGF receptor. In acute hypocalcemia, when increased PTH secretion is needed to restore the calcium homeostasis, this inhibitory effect of FGF23 is abolished.


Assuntos
Calcitriol/sangue , Fatores de Crescimento de Fibroblastos/metabolismo , Hipocalcemia/sangue , Hormônio Paratireóideo/sangue , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Fator de Crescimento de Fibroblastos 23 , Homeostase/fisiologia , Ratos Wistar , Uremia/metabolismo
7.
Kidney Int ; 92(1): 165-178, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28341272

RESUMO

Fibroblast growth factor 23 (FGF23) secreted by osteocytes is a circulating factor essential for phosphate homeostasis. High plasma FGF23 levels are associated with cardiovascular complications and mortality. Increases of plasma FGF23 in uremia antedate high levels of phosphate, suggesting a disrupted feedback regulatory loop or an extra-skeletal source of this phosphatonin. Since induction of FGF23 expression in injured organs has been reported we decided to examine the regulation of FGF23 gene and protein expressions in the kidney and whether kidney-derived FGF23 contributes to the high plasma levels of FGF23 in uremia. FGF23 mRNA was not detected in normal kidneys, but was clearly demonstrated in injured kidneys, already after four hours in obstructive nephropathy and at 8 weeks in the remnant kidney of 5/6 nephrectomized rats. No renal extraction was found in uremic rats in contrast to normal rats. Removal of the remnant kidney had no effect on plasma FGF23 levels. Well-known regulators of FGF23 expression in bone, such as parathyroid hormone, calcitriol, and inhibition of the FGF receptor by PD173074, had no impact on kidney expression of FGF23. Thus, the only direct contribution of the injured kidney to circulating FGF23 levels in uremia appears to be reduced renal extraction of bone-derived FGF23. Kidney-derived FGF23 does not generate high plasma FGF23 levels in uremia and is regulated differently than the corresponding regulation of FGF23 gene expression in bone.


Assuntos
Fatores de Crescimento de Fibroblastos/sangue , Fibroblastos/metabolismo , Rim/metabolismo , Insuficiência Renal Crônica/sangue , Uremia/sangue , Animais , Biomarcadores/sangue , Osso e Ossos/metabolismo , Modelos Animais de Doenças , Fatores de Crescimento de Fibroblastos/genética , Fibrose , Rim/patologia , Rim/fisiopatologia , Masculino , Hormônio Paratireóideo/sangue , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/fisiopatologia , Fatores de Tempo , Regulação para Cima , Uremia/genética , Uremia/fisiopatologia
8.
BMC Nephrol ; 16: 29, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25885328

RESUMO

BACKGROUND: Understanding the regulation of mineral homeostasis and function of the skeleton as buffer for Calcium and Phosphate has regained new interest with introduction of the syndrome "Chronic Kidney Disease-Mineral and Bone Disorder"(CKD-MBD). The very rapid minute-to-minute regulation of plasma-Ca(2+) (p-Ca(2+)) takes place via an exchange mechanism of Ca(2+) between plasma and bone. A labile Ca storage pool exists on bone surfaces storing excess or supplying Ca when blood Ca is lowered. Aim was to examine minute-to-minute regulation of p-Ca(2+) in the very early phase of acute uremia, as induced by total bilateral nephrectomy and to study the effect of absence of kidneys on the rapid recovery of p-Ca(2+) from a brief induction of acute hypocalcemia. METHODS: The rapid regulation of p-Ca(2+) was examined in sham-operated rats, acute nephrectomized rats (NX), acute thyroparathyrectomized(TPTX) rats and NX-TPTX rats. RESULTS: The results clearly showed that p-Ca(2+) falls rapidly and significantly very early after acute NX, from 1.23 ± 0.02 to 1.06 ± 0.04 mM (p < 0.001). Further hypocalcemia was induced by a 30 min iv infusion of EGTA. Control groups had saline. After discontinuing EGTA a rapid increase in p-Ca(2+) took place, but with a lower level in NX rats (p < 0.05). NX-TPTX model excluded potential effect of accumulation of Calcitonin and C-terminal PTH, both having potential hypocalcemic actions. Acute TPTX resulted in hypercalcemia, 1.44 ± 0.02 mM and less in NX-TPTX rats,1.41 ± 0.02 mM (p < 0.05). Recovery of p-Ca(2+) from hypocalcemia resulted in lower levels in NX-TPTX than in TPTX rats, 1.20 ± 0.02 vs.1.30 ± 0.02 (p < 0.05) demonstrating that absence of kidneys significantly affected the rapid regulation of p-Ca(2+) independent of PTH, C-PTH and CT. CONCLUSIONS: P-Ca(2+) on a minute-to-minute basis is influenced by presence of kidneys. Hypocalcemia developed rapidly in acute uremia. Levels of p-Ca(2+), obtained during recovery from hypocalcemia resulted in lower levels in acutely nephrectomized rats. This indicates that kidneys are of significant importance for the 'set-point' of p-Ca(2+) on bone surface, independently of PTH and calcitonin. Our results point toward existence of an as yet unknown factor/mechanism, which mediates the axis between kidney and bone, and which is involved in the very rapid regulation of p-Ca(2+).


Assuntos
Osso e Ossos/metabolismo , Cálcio/metabolismo , Hipercalcemia/sangue , Hipocalcemia/sangue , Uremia/fisiopatologia , Animais , Densidade Óssea/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Homeostase/fisiologia , Hipercalcemia/diagnóstico , Hipocalcemia/diagnóstico , Masculino , Nefrectomia/métodos , Paratireoidectomia/métodos , Distribuição Aleatória , Ratos , Ratos Wistar , Valores de Referência , Tireoidectomia/métodos , Fatores de Tempo , Uremia/etiologia
9.
Scand J Clin Lab Invest ; 74(3): 206-12, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24456420

RESUMO

BACKGROUND: Uremic patients develop hyperplasia of the parathyroid glands due to disturbances in the mineral metabolism. The hyperplastic parathyroids are associated with significant expression of parathyroid hormone (PTH)-related peptide (PTHrP). PTHrP has been shown to have an autocrine/paracrine function in the parathyroids, but it is still uncertain if PTHrP is a secretory product of the gland and thereby possess endocrine actions. In cells of severe adenomatous secondary hyperparathyroidism PTHrP and PTH have been found to be co-localized in the same secretory granules. PTH and PTHrP act through the same receptor, the PTH1R, and it has been shown experimentally that PTHrP enhances the PTH secretory response to hypocalcemia, indicating a link between the two hormones. METHODS: Together with a number of parameters involved in mineral homeostasis plasma PTHrP was measured before hemodialysis in 90 patients and in 15 healthy subjects. Plasma PTH was determined in order to examine the possible relationship between the two peptides. RESULTS: In hemodialysis patients mean plasma PTHrP, 4.2 ± 2.1, was significantly lower than that of healthy subjects, 8.3 ± 1.1 pmol/L, p < 0.0001. No relationship was found between plasma PTHrP and PTH in hemodialysis patients. Gender, PTX, specific treatments and diagnoses had no impact on PTHrP concentrations. CONCLUSION: Thus PTHrP is measurable in hemodialysis patients, but its secretion might not be part of a regulated mineral homeostatic process and may not derive from the uremic hyperplastic parathyroid glands.


Assuntos
Glândulas Paratireoides/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/sangue , Hormônio Paratireóideo/sangue , Diálise Renal , Uremia/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Estudos Transversais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Glândulas Paratireoides/fisiopatologia , Uremia/fisiopatologia , Uremia/terapia
10.
Clin Kidney J ; 16(12): 2712-2720, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38046005

RESUMO

Background: Plasma (p-)activin A is elevated in chronic kidney disease-mineral and bone disorder (CKD-MBD). Activin A inhibition ameliorates CKD-MBD complications (vascular calcification and bone disease) in rodent CKD models. We examined whether p-activin A was associated with major adverse cardiovascular events (MACE), all-cause mortality and CKD-MBD complications in CKD patients. Methods: The study included 916 participants (741 patients and 175 controls) from the prospective Copenhagen CKD cohort. Comparisons of p-activin A with estimated glomerular filtration rate (eGFR), coronary and thoracic aorta Agatston scores, and bone mineral density (BMD) were evaluated by univariable linear regression using Spearman's rank correlation, analysis of covariance and ordinal logistic regression with adjustments. Association of p-activin A with rates of MACE and all-cause mortality was evaluated by the Aalen-Johansen or Kaplan-Meier estimator, with subsequent multiple Cox regression analyses. Results: P-activin A was increased by CKD stage 3 (124-225 pg/mL, P < .001) and correlated inversely with eGFR (r = -0.53, P < 0.01). P-activin A was associated with all-cause mortality [97 events, hazard ratio 1.55 (95% confidence interval 1.04; 2.32), P < 0.05] after adjusting for age, sex, diabetes mellitus (DM) and eGFR. Median follow-up was 4.36 (interquartile range 3.64-4.75) years. The association with MACE was not significant after eGFR adjustment. Agatston scores and BMD were not associated with p-activin A. Conclusion: P-activin A increased with declining kidney function and was associated with all-cause mortality independently of age, sex, DM and eGFR. No association with MACE, vascular calcification or BMD was demonstrated.

11.
JBMR Plus ; 6(4): e10610, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35434452

RESUMO

Vascular calcification and bone disorder progress simultaneously in chronic kidney disease (CKD). Still, how the complex pathological mechanisms are linked is only sparsely understood. Up to now, the focus has been on the disturbed bone metabolism in developing vascular calcification. However, our group has recently demonstrated that vascular calcification has negative effects on bone formation and mineralization as shown in the bone of normal recipient rats transplanted with the calcified aorta from CKD rats. In the present in vitro study, the hypothesis of a direct crosstalk between the vasculature and bone was examined. Calcified aortas from 5/6 nephrectomized rats and normal aortas from control rats were excised and incubated ex vivo. The calcified aorta secreted large amounts of sclerostin, dickkopf-1 (Dkk1), and activin A. Both normal and calcified aortas secreted frizzle-related protein 4 (SFRP4). Aorta rings were co-incubated with the osteoblast-like cell line UMR-106. The calcified aorta strongly inhibited calcium crystal formation in UMR-106 cells, together with a significant upregulation of the mineralization inhibitors osteopontin and progressive ankylosis protein homolog (ANKH). The strong stimulation of osteopontin was blocked by lithium chloride, indicating involvement of Wnt/ß-catenin signaling. The present in vitro study shows detrimental effects of the calcified aorta on bone cell mineralization. These findings support the hypothesis of an active role of the calcified vasculature in the systemic CKD-mineral and bone disorder (CKD-MBD), resulting in a pathological vascular-bone tissue crosstalk. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

12.
PLoS One ; 16(6): e0252554, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34061900

RESUMO

Acute kidney injury (AKI) is associated with a very high mortality and an increased risk for progression to chronic kidney disease (CKD). Ischemia-reperfusion injury (IRI) is a model for AKI, which results in tubular damage, dysfunction of the mitochondria and autophagy, and in decreased cellular nicotinamide adenine dinucleotide (NAD+) with progressing fibrosis resulting in CKD. NAD+ is a co-enzyme for several proteins, including the NAD+ dependent sirtuins. NAD+ augmentation, e.g. by use of its precursor nicotinamide riboside (NR), improves mitochondrial homeostasis and organismal metabolism in many species. In the present investigation the effects of prophylactic administration of NR on IRI-induced AKI were studied in the rat. Bilateral IRI reduced kidney tissue NAD+, caused tubular damage, reduced α-Klotho (klotho), and altered autophagy flux. AKI initiated progression to CKD, as shown by induced profibrotic Periostin (postn) and Inhibin subunit beta-A, (activin A / Inhba), both 24 hours and 14 days after surgery. NR restored tissue NAD+ to that of the sham group, increased autophagy (reduced p62) and sirtuin1 (Sirt1) but did not ameliorate renal tubular damage and profibrotic genes in the 24 hours and 14 days IRI models. AKI induced NAD+ depletion and impaired autophagy, while augmentation of NAD+ by NR restored tissue NAD+ and increased autophagy, possibly serving as a protective response. However, prophylactic administration of NR did not ameliorate tubular damage of the IRI rats nor rescued the initiation of fibrosis in the long-term AKI to CKD model, which is a pivotal event in CKD pathogenesis.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/etiologia , NAD/metabolismo , Niacinamida/análogos & derivados , Substâncias Protetoras/administração & dosagem , Compostos de Piridínio/administração & dosagem , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Injúria Renal Aguda/complicações , Injúria Renal Aguda/metabolismo , Animais , Autofagia/efeitos dos fármacos , Progressão da Doença , Fibrose , Glucuronidase/metabolismo , Rim/metabolismo , Rim/patologia , Proteínas Klotho , Masculino , Mitocôndrias/metabolismo , Niacinamida/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Wistar , Insuficiência Renal Crônica/etiologia , Sirtuína 1/metabolismo , Resultado do Tratamento
13.
Bone Rep ; 15: 101130, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34584906

RESUMO

Increasing levels of magnesium in blood are associated with reduced risk of cardiovascular disease in chronic kidney disease (CKD). Magnesium supplementation may reduce the progression of vascular calcification in CKD. The diurnal pattern and effect of fasting on magnesium in blood and urine in CKD is unknown, and knowledge of this may influence management of magnesium supplementation. We included ten patients with CKD stage four without diabetes mellitus and ten healthy controls. Participants were admitted to our hospital ward for a 24-h study period. Blood and urine samples were collected in a non-fasting state at 8 o'clock in the morning and every third hour hereafter until the final samples in a fasting state at 8 o'clock the following morning. We found no diurnal variation in plasma magnesium (p = 0.097) in either group, but a significant diurnal variation in urinary excretion of magnesium (p = 0.044) in both CKD and healthy controls with no significant interaction between the two groups, and thus no suggestion that CKD affects diurnal variation of plasma magnesium or urinary magnesium excretion. The levels of plasma magnesium were not significantly different in fasting and non-fasting conditions. Magnesium in plasma does not display a significant diurnal variation and can be measured at any time of day and in both fasting and non-fasting conditions. Urinary magnesium excretion displays diurnal variation, which is likely related to increased uptake of magnesium during meals and helps maintain a stable concentration of magnesium in blood.

14.
J Bone Miner Res ; 36(3): 510-522, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33301619

RESUMO

An association between lower bone mineral density (BMD) and presence of vascular calcification (VC) has been reported in several studies. Chronic kidney disease (CKD) causes detrimental disturbances in the mineral balance, bone turnover, and development of severe VC. Our group has previously demonstrated expression of Wnt inhibitors in calcified arteries of CKD rats. Therefore, we hypothesized that the CKD-induced VC via this pathway signals to bone and induces bone loss. To address this novel hypothesis, we developed a new animal model using isogenic aorta transplantation (ATx). Severely calcified aortas from uremic rats were transplanted into healthy rats (uremic ATx). Transplantation of normal aortas into healthy rats (normal ATx) and age-matched rats (control) served as control groups. Trabecular tissue mineral density, as measured by µCT, was significantly lower in uremic ATx rats compared with both control groups. Uremic ATx rats showed a significant upregulation of the mineralization inhibitors osteopontin and progressive ankylosis protein homolog in bone. In addition, we found significant changes in bone mRNA levels of several genes related to extracellular matrix, bone turnover, and Wnt signaling in uremic ATx rats, with no difference between normal ATx and control. The bone histomorphometry analysis showed significant lower osteoid area in uremic ATx compared with normal ATx along with a trend toward fewer osteoblasts as well as more osteoclasts in the erosion lacunae. Uremic ATx and normal ATx had similar trabecular number and thickness. The bone formation rate did not differ between the three groups. Plasma biochemistry, including sclerostin, kidney, and mineral parameters, were similar between all three groups. ex vivo cultures of aorta from uremic rats showed high secretion of the Wnt inhibitor sclerostin. In conclusion, the presence of VC lowers BMD, impairs bone metabolism, and affects several pathways in bone. The present results prove the existence of a vasculature to bone tissue cross-talk. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Assuntos
Insuficiência Renal Crônica , Calcificação Vascular , Animais , Osso e Ossos , Rim , Ratos , Via de Sinalização Wnt
15.
PLoS One ; 13(8): e0201936, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30075015

RESUMO

Uremic vascular calcification is a regulated cell-mediated process wherein cells in the arterial wall transdifferentiate to actively calcifying cells resulting in a process resembling bone formation. Wnt signalling is established as a major driver for vessel formation and maturation and for embryonic bone formation, and disturbed Wnt signalling might play a role in vascular calcification. ICG-001 is a small molecule Wnt inhibitor that specifically targets the coactivator CREB binding protein (CBP)/ß-catenin-mediated signalling. In the present investigation we examined the effect of ICG-001 on vascular calcification in uremic rats. Uremic vascular calcification was induced in adult male rats by 5/6-nephrectomy, high phosphate diet and alfacalcidol. The presence of uremic vascular calcification in the aorta was associated with induction of gene expression of the Wnt target gene and marker of proliferation, cyclinD1; the mediator of canonical Wnt signalling, ß-catenin and the matricellular proteins, fibronectin and periostin. Furthermore, genes from fibrosis-related pathways, TGF-ß and activin A, as well as factors related to epithelial-mesenchymal transition, snail1 and vimentin were induced. ICG-001 treatment had significant effects on gene expression in kidney and aorta from healthy rats. These effects were however limited in uremic rats, and treatment with ICG-001 did not reduce the Ca-content of the uremic vasculature.


Assuntos
Proteína de Ligação a CREB/metabolismo , Uremia/metabolismo , Uremia/patologia , Calcificação Vascular/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Biomarcadores , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Falência Renal Crônica/sangue , Falência Renal Crônica/etiologia , Falência Renal Crônica/metabolismo , Falência Renal Crônica/patologia , Masculino , Minerais/metabolismo , Especificidade de Órgãos/genética , Pirimidinonas/farmacologia , Ratos , Uremia/sangue , Via de Sinalização Wnt/efeitos dos fármacos , Microtomografia por Raio-X
16.
PLoS One ; 13(1): e0190820, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29304096

RESUMO

Hyperphosphatemia and vascular calcification are frequent complications of chronic renal failure and bone morphogenetic protein 7 (BMP7) has been shown to protect against development of vascular calcification in uremia. The present investigation examined the potential reversibility of established uremic vascular calcification by treatment of uremic rats with BMP7. A control model of isogenic transplantation of a calcified aorta from uremic rats into healthy littermates examined whether normalization of the uremic environment reversed vascular calcification. Uremia and vascular calcification were induced in rats by 5/6 nephrectomy, high phosphate diet and alfacalcidol treatment. After 14 weeks severe vascular calcification was present and rats were allocated to BMP7, vehicle or aorta transplantation. BMP7 treatment caused a significant decrease of plasma phosphate to 1.56 ± 0.17 mmol/L vs 2.06 ± 0.34 mmol/L in the vehicle group even in the setting of uremia and high phosphate diet. Uremia and alfacalcidol resulted in an increase in aortic expression of genes related to fibrosis, osteogenic transformation and extracellular matrix calcification, and the BMP7 treatment resulted in a decrease in the expression of profibrotic genes. The total Ca-content of the aorta was however unchanged both in the abdominal aorta: 1.9 ± 0.6 µg/mg tissue in the vehicle group vs 2.2 ± 0.6 µg/mg tissue in the BMP7 group and in the thoracic aorta: 71 ± 27 µg/mg tissue in the vehicle group vs 54 ± 18 µg/mg tissue in the BMP7 group. Likewise, normalization of the uremic environment by aorta transplantation had no effect on the Ca-content of the calcified aorta: 16.3 ± 0.6 µg/mg tissue pre-transplantation vs 15.9 ± 2.3 µg/mg tissue post-transplantation. Aortic expression of genes directly linked to extracellular matrix calcification was not affected by BMP7 treatment, which hypothetically might explain persistent high Ca-content in established vascular calcification. The present results highlight the importance of preventing the development of vascular calcification in chronic kidney disease. Once established, vascular calcification persists even in the setting when hyperphosphatemia or the uremic milieu is abolished.


Assuntos
Proteína Morfogenética Óssea 7/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Uremia/tratamento farmacológico , Calcificação Vascular/tratamento farmacológico , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Proteína Morfogenética Óssea 7/uso terapêutico , Doença Crônica , Fibrose , Masculino , Fosfatos/sangue , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Uremia/genética , Microtomografia por Raio-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA