Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
1.
Sci Rep ; 13(1): 4898, 2023 03 25.
Article de Anglais | MEDLINE | ID: mdl-36966182

RÉSUMÉ

An elevation in serum phosphate-also called hyperphosphatemia-is associated with reduced kidney function in chronic kidney disease (CKD). Reports show CKD patients are more likely to develop lung disease and have poorer kidney function that positively correlates with pulmonary obstruction. However, the underlying mechanisms are not well understood. Here, we report that two murine models of CKD, which both exhibit increased serum levels of phosphate and fibroblast growth factor (FGF) 23, a regulator of phosphate homeostasis, develop concomitant airway inflammation. Our in vitro studies point towards a similar increase of phosphate-induced inflammatory markers in human bronchial epithelial cells. FGF23 stimulation alone does not induce a proinflammatory response in the non-COPD bronchial epithelium and phosphate does not cause endogenous FGF23 release. Upregulation of the phosphate-induced proinflammatory cytokines is accompanied by activation of the extracellular-signal regulated kinase (ERK) pathway. Moreover, the addition of cigarette smoke extract (CSE) during phosphate treatments exacerbates inflammation as well as ERK activation, whereas co-treatment with FGF23 attenuates both the phosphate as well as the combined phosphate- and CS-induced inflammatory response, independent of ERK activation. Together, these data demonstrate a novel pathway that potentially explains pathological kidney-lung crosstalk with phosphate as a key mediator.


Sujet(s)
Fumer des cigarettes , Broncho-pneumopathie chronique obstructive , Insuffisance rénale chronique , Humains , Animaux , Souris , Phosphates/métabolisme , Fumer des cigarettes/effets indésirables , Inflammation/anatomopathologie , Extracellular Signal-Regulated MAP Kinases/métabolisme , Insuffisance rénale chronique/complications , Épithélium/métabolisme , Broncho-pneumopathie chronique obstructive/anatomopathologie , Cellules épithéliales/métabolisme
2.
Kidney Int ; 102(2): 261-279, 2022 08.
Article de Anglais | MEDLINE | ID: mdl-35513125

RÉSUMÉ

Fibroblast growth factor (FGF) 23 is a phosphate-regulating hormone that is elevated in patients with chronic kidney disease and associated with cardiovascular mortality. Experimental studies showed that elevated FGF23 levels induce cardiac hypertrophy by targeting cardiac myocytes via FGF receptor isoform 4 (FGFR4). A recent structural analysis revealed that the complex of FGF23 and FGFR1, the physiologic FGF23 receptor in the kidney, includes soluble α-klotho (klotho) and heparin, which both act as co-factors for FGF23/FGFR1 signaling. Here, we investigated whether soluble klotho, a circulating protein with cardio-protective properties, and heparin, a factor that is routinely infused into patients with kidney failure during the hemodialysis procedure, regulate FGF23/FGFR4 signaling and effects in cardiac myocytes. We developed a plate-based binding assay to quantify affinities of specific FGF23/FGFR interactions and found that soluble klotho and heparin mediate FGF23 binding to distinct FGFR isoforms. Heparin specifically mediated FGF23 binding to FGFR4 and increased FGF23 stimulatory effects on hypertrophic growth and contractility in isolated cardiac myocytes. When repetitively injected into two different mouse models with elevated serum FGF23 levels, heparin aggravated cardiac hypertrophy. We also developed a novel procedure for the synthesis and purification of recombinant soluble klotho, which showed anti-hypertrophic effects in FGF23-treated cardiac myocytes. Thus, soluble klotho and heparin act as independent FGF23 co-receptors with opposite effects on the pathologic actions of FGF23, with soluble klotho reducing and heparin increasing FGF23-induced cardiac hypertrophy. Hence, whether heparin injections during hemodialysis in patients with extremely high serum FGF23 levels contribute to their high rates of cardiovascular events and mortality remains to be studied.


Sujet(s)
Facteur-23 de croissance des fibroblastes , Héparine , Protéines Klotho , Insuffisance rénale chronique , Animaux , Cardiomégalie , Glucuronidase/métabolisme , Héparine/métabolisme , Humains , Protéines Klotho/métabolisme , Souris , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/thérapie
3.
Sci Rep ; 12(1): 7326, 2022 05 05.
Article de Anglais | MEDLINE | ID: mdl-35513431

RÉSUMÉ

Fibroblast growth factor (FGF) 21, a hormone that increases insulin sensitivity, has shown promise as a therapeutic agent to improve metabolic dysregulation. Here we report that FGF21 directly targets cardiac myocytes by binding ß-klotho and FGF receptor (FGFR) 4. In combination with high glucose, FGF21 induces cardiac myocyte growth in width mediated by extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. While short-term FGF21 elevation can be cardio-protective, we find that in type 2 diabetes (T2D) in mice, where serum FGF21 levels are elevated, FGFR4 activation induces concentric cardiac hypertrophy. As T2D patients are at risk for heart failure with preserved ejection fraction (HFpEF), we propose that induction of concentric hypertrophy by elevated FGF21-FGFR4 signaling may constitute a novel mechanism promoting T2D-associated HFpEF such that FGFR4 blockade might serve as a cardio-protective therapy in T2D. In addition, potential adverse cardiac effects of FGF21 mimetics currently in clinical trials should be investigated.


Sujet(s)
Diabète de type 2 , Défaillance cardiaque , Animaux , Cardiomégalie/métabolisme , Diabète de type 2/complications , Diabète de type 2/métabolisme , Modèles animaux de maladie humaine , Facteurs de croissance fibroblastique/métabolisme , Défaillance cardiaque/métabolisme , Humains , Souris , Myocytes cardiaques/métabolisme , Récepteur FGFR4/métabolisme , Débit systolique
4.
Sci Rep ; 12(1): 6695, 2022 04 23.
Article de Anglais | MEDLINE | ID: mdl-35461329

RÉSUMÉ

In chronic kidney disease, ferric citrate has been shown to be an effective phosphate binder and source of enteral iron; however, the effects of ferric citrate on the kidney have been less well-studied. Here, in Col4α3 knockout mice-a murine model of progressive chronic kidney disease, we evaluated the effects of five weeks of 1% ferric citrate dietary supplementation. As expected, ferric citrate lowered serum phosphate concentrations and increased serum iron levels in the Col4α3 knockout mice. Consistent with decreased enteral phosphate absorption and possibly improved iron status, ferric citrate greatly reduced circulating fibroblast growth factor 23 levels. Interestingly, ferric citrate also lessened systemic inflammation, improved kidney function, reduced albuminuria, and decreased kidney inflammation and fibrosis, suggesting renoprotective effects of ferric citrate in the setting of chronic kidney disease. The factors mediating possible ferric citrate renoprotection, the mechanisms by which they may act, and whether ferric citrate affects chronic kidney disease progression in humans deserves further study.


Sujet(s)
Composés du fer III , Insuffisance rénale chronique , Animaux , Modèles animaux de maladie humaine , Femelle , Composés du fer III/pharmacologie , Humains , Inflammation , Fer , Mâle , Souris , Souris knockout , Phosphates
5.
Elife ; 112022 03 18.
Article de Anglais | MEDLINE | ID: mdl-35302487

RÉSUMÉ

Elevations in plasma phosphate concentrations (hyperphosphatemia) occur in chronic kidney disease (CKD), in certain genetic disorders, and following the intake of a phosphate-rich diet. Whether hyperphosphatemia and/or associated changes in metabolic regulators, including elevations of fibroblast growth factor 23 (FGF23) directly contribute to specific complications of CKD is uncertain. Here, we report that similar to patients with CKD, mice with adenine-induced CKD develop inflammation, anemia, and skeletal muscle wasting. These complications are also observed in mice fed high phosphate diet even without CKD. Ablation of pathologic FGF23-FGFR4 signaling did not protect mice on an increased phosphate diet or mice with adenine-induced CKD from these sequelae. However, low phosphate diet ameliorated anemia and skeletal muscle wasting in a genetic mouse model of CKD. Our mechanistic in vitro studies indicate that phosphate elevations induce inflammatory signaling and increase hepcidin expression in hepatocytes, a potential causative link between hyperphosphatemia, anemia, and skeletal muscle dysfunction. Our study suggests that high phosphate intake, as caused by the consumption of processed food, may have harmful effects irrespective of pre-existing kidney injury, supporting not only the clinical utility of treating hyperphosphatemia in CKD patients but also arguing for limiting phosphate intake in healthy individuals.


Sujet(s)
Anémie , Hyperphosphatémie , Anémie/complications , Animaux , Facteur-23 de croissance des fibroblastes/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Humains , Hyperphosphatémie/complications , Inflammation , Souris , Muscles squelettiques/métabolisme , Récepteur FGFR4
6.
Am J Physiol Lung Cell Mol Physiol ; 322(5): L625-L640, 2022 05 01.
Article de Anglais | MEDLINE | ID: mdl-35272496

RÉSUMÉ

Chronic illnesses rarely present in a vacuum, devoid of other complications, and chronic kidney disease is hardly an exception. Comorbidities associated with chronic kidney disease lead to faster disease progression, expedited dialysis dependency, and a higher mortality rate. Although chronic kidney disease is most commonly accompanied by cardiovascular diseases and diabetes, there is clear cross talk between the lungs and kidneys pH balance, phosphate metabolism, and immune system regulation. Our present understanding of the exact underlying mechanisms that contribute to chronic kidney disease-related pulmonary disease is poor. This review summarizes the current research on kidney-pulmonary interorgan cross talk in the context of chronic kidney disease, highlighting various acute and chronic pulmonary diseases that lead to further complications in patient care. Treatment options for patients presenting with chronic kidney disease and lung disease are explored by assessing activated molecular pathways and the body's compensatory response mechanisms following homeostatic imbalance. Understanding the link between the lungs and kidneys will potentially improve health outcomes for patients and guide healthcare professionals to better understand how and when to treat each of the pulmonary comorbidities that can present with chronic kidney disease.


Sujet(s)
Maladies pulmonaires , Insuffisance rénale chronique , Maladie chronique , Femelle , Humains , Rein/métabolisme , Poumon , Maladies pulmonaires/métabolisme , Mâle , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/métabolisme
7.
Kidney Int ; 101(4): 711-719, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-34838540

RÉSUMÉ

Ferric citrate is approved as an iron replacement product in patients with non-dialysis chronic kidney disease and iron deficiency anemia. Ferric citrate-delivered iron is enterally absorbed, but the specific mechanisms involved have not been evaluated, including the possibilities of conventional, transcellular ferroportin-mediated absorption and/or citrate-mediated paracellular absorption. Here, we first demonstrate the efficacy of ferric citrate in high hepcidin models, including Tmprss6 knockout mice (characterized by iron-refractory iron deficiency anemia) with and without adenine diet-induced chronic kidney disease. Next, to assess whether or not enteral ferric citrate absorption is dependent on ferroportin, we evaluated the effects of ferric citrate in a tamoxifen-inducible, enterocyte-specific ferroportin knockout murine model (Villin-Cre-ERT2, Fpnflox/flox). In this model, ferroportin deletion was efficient, as tamoxifen injection induced a 4000-fold decrease in duodenum ferroportin mRNA expression, with undetectable ferroportin protein on Western blot of duodenal enterocytes, resulting in a severe iron deficiency anemia phenotype. In ferroportin-deficient mice, three weeks of 1% ferric citrate dietary supplementation, a dose that prevented iron deficiency in control mice, did not improve iron status or rescue the iron deficiency anemia phenotype. We repeated the conditional ferroportin knockout experiment in the setting of uremia, using an adenine nephropathy model, where three weeks of 1% ferric citrate dietary supplementation again failed to improve iron status or rescue the iron deficiency anemia phenotype. Thus, our data suggest that enteral ferric citrate absorption is dependent on conventional enterocyte iron transport by ferroportin and that, in these models, significant paracellular absorption does not occur.


Sujet(s)
Anémie par carence en fer , Transporteurs de cations , Anémie par carence en fer/traitement médicamenteux , Animaux , Transporteurs de cations/génétique , Composés du fer III/pharmacologie , Hepcidines/métabolisme , Humains , Fer/métabolisme , Souris
8.
Kidney Int ; 96(4): 813-815, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31543148

RÉSUMÉ

High serum concentrations of the phosphaturic hormone, fibroblast growth factor 23 (FGF23), contribute to various tissue injuries. In chronic kidney disease, the sources of FGF23 and the stimuli that control FGF23 production differ from those in the physiologic scenario. Mediators of inflammation are intensively studied as potential factors that contribute to FGF23 elevations and thereby might function as drug targets to lower FGF23 levels. The present study focuses on tumor necrosis factor.


Sujet(s)
Hypophosphatémie familiale , Insuffisance rénale chronique , Facteur-23 de croissance des fibroblastes , Facteurs de croissance fibroblastique , Humains , Inflammation
9.
Int J Mol Sci ; 20(17)2019 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-31461904

RÉSUMÉ

In patients with chronic kidney disease (CKD), adverse outcomes such as systemic inflammation and anemia are contributing pathologies which increase the risks for cardiovascular mortality. Amongst these complications, abnormalities in mineral metabolism and the metabolic milieu are associated with chronic inflammation and iron dysregulation, and fibroblast growth factor 23 (FGF23) is a risk factor in this context. FGF23 is a bone-derived hormone that is essential for regulating vitamin D and phosphate homeostasis. In the early stages of CKD, serum FGF23 levels rise 1000-fold above normal values in an attempt to maintain normal phosphate levels. Despite this compensatory action, clinical CKD studies have demonstrated powerful and dose-dependent associations between FGF23 levels and higher risks for mortality. A prospective pathomechanism coupling elevated serum FGF23 levels with CKD-associated anemia and cardiovascular injury is its strong association with chronic inflammation. In this review, we will examine the current experimental and clinical evidence regarding the role of FGF23 in renal physiology as well as in the pathophysiology of CKD with an emphasis on chronic inflammation and anemia.


Sujet(s)
Anémie par carence en fer/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Insuffisance rénale chronique/métabolisme , Anémie par carence en fer/étiologie , Anémie par carence en fer/anatomopathologie , Animaux , Facteur-23 de croissance des fibroblastes , Hepcidines/métabolisme , Humains , Inflammation/métabolisme , Fer/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/anatomopathologie
10.
Am J Hypertens ; 32(1): 34-44, 2019 01 01.
Article de Anglais | MEDLINE | ID: mdl-30329020

RÉSUMÉ

BACKGROUND: In uremic animals, vitamin D receptor (VDR) agonists like paricalcitol (Pc) attenuate cardiac hypertrophy, but this effect has not been replicated consistently in humans with chronic kidney disease. Elevated fibroblast growth factor 23 (FGF23) levels cause cardiac hypertrophy with activation of the myocardial calcineurin/nuclear factor of activated T cell (NFAT) axis and may antagonize the cardioprotective effects of VDR agonist therapy. We hypothesized that the effectiveness of Pc may depend on the prevailing circulating levels of FGF23 and could be potentiated by the combined administration of a pan-FGF23 receptor (FGFR) blocker agent (PD173074). METHODS: In rats with 5/6 nephrectomy treated with Pc or PD173074 or both agents concurrently, myocardial mRNA expression of renin-angiotensin system, VDR, FGFR4, and calcineurin/NFAT target genes was determined. In adolescents on hemodialysis, we analyzed sequential echocardiograms, blood pressures and serial FGF23 measurements, and their relations to the cumulative administered dose of parenteral Pc. RESULTS: The ratio of Pc dose/plasma levels of FGF23 correlated inversely (P < 0.005) with the cardiac mass in uremic rats and in hemodialysis patients, independently of hypertension. Despite persistently elevated FGF23 levels and myocardial FGFR4 activation, Pc suppressed upregulated myocardial calcineurin/NFAT target genes, and the effects were amplified by coadministration of PD173074. CONCLUSIONS: The beneficial effects of Pc on uremic cardiac hypertrophy are counterbalanced by the increased FGF23 levels. Blockade of FGF23-mediated signaling increased the Pc-induced suppression of the myocardial calcineurin/NFAT system. Higher doses of Pc should be considered in the treatment of patients with uremic cardiomyopathy.


Sujet(s)
Syndrome cardiorénal/prévention et contrôle , Cardiomyopathies/prévention et contrôle , Ergocalciférol/pharmacologie , Facteurs de croissance fibroblastique/sang , Ventricules cardiaques/effets des médicaments et des substances chimiques , Défaillance rénale chronique/traitement médicamenteux , Pyrimidines/pharmacologie , Récepteur FGFR4/antagonistes et inhibiteurs , Récepteur calcitriol/agonistes , Adolescent , Animaux , Syndrome cardiorénal/métabolisme , Syndrome cardiorénal/physiopathologie , Cardiomyopathies/métabolisme , Cardiomyopathies/physiopathologie , Enfant , Modèles animaux de maladie humaine , Association de médicaments , Femelle , Facteur-23 de croissance des fibroblastes , Ventricules cardiaques/métabolisme , Ventricules cardiaques/physiopathologie , Humains , Défaillance rénale chronique/métabolisme , Défaillance rénale chronique/physiopathologie , Mâle , Rat Sprague-Dawley , Récepteur FGFR4/métabolisme , Récepteur calcitriol/métabolisme , Études rétrospectives , Transduction du signal , Urémie/traitement médicamenteux , Urémie/métabolisme , Urémie/anatomopathologie , Urémie/physiopathologie , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Remodelage ventriculaire/effets des médicaments et des substances chimiques
11.
Sci Rep ; 7(1): 1993, 2017 05 16.
Article de Anglais | MEDLINE | ID: mdl-28512310

RÉSUMÉ

Fibroblast growth factor (FGF) 23 is a phosphaturic hormone that directly targets cardiac myocytes via FGF receptor (FGFR) 4 thereby inducing hypertrophic myocyte growth and the development of left ventricular hypertrophy (LVH) in rodents. Serum FGF23 levels are highly elevated in patients with chronic kidney disease (CKD), and it is likely that FGF23 directly contributes to the high rates of LVH and cardiac death in CKD. It is currently unknown if the cardiac effects of FGF23 are solely pathological, or if they potentially can be reversed. Here, we report that FGF23-induced cardiac hypertrophy is reversible in vitro and in vivo upon removal of the hypertrophic stimulus. Specific blockade of FGFR4 attenuates established LVH in the 5/6 nephrectomy rat model of CKD. Since CKD mimics a form of accelerated cardiovascular aging, we also studied age-related cardiac remodeling. We show that aging mice lacking FGFR4 are protected from LVH. Finally, FGF23 increases cardiac contractility via FGFR4, while known effects of FGF23 on aortic relaxation do not require FGFR4. Taken together, our data highlight a role of FGF23/FGFR4 signaling in the regulation of cardiac remodeling and function, and indicate that pharmacological interference with cardiac FGF23/FGFR4 signaling might protect from CKD- and age-related LVH.


Sujet(s)
Facteurs de croissance fibroblastique/métabolisme , Hypertrophie ventriculaire gauche/étiologie , Hypertrophie ventriculaire gauche/métabolisme , Récepteur FGFR4/métabolisme , Transduction du signal , Animaux , Biopsie , Régime alimentaire , Modèles animaux de maladie humaine , Facteur-23 de croissance des fibroblastes , Facteurs de croissance fibroblastique/sang , Hypertrophie ventriculaire gauche/anatomopathologie , Souris , Souris knockout , Contraction myocardique/génétique , Myocytes cardiaques/métabolisme , Rats , Récepteur FGFR4/antagonistes et inhibiteurs
12.
J Vis Exp ; (121)2017 03 10.
Article de Anglais | MEDLINE | ID: mdl-28362385

RÉSUMÉ

The liver plays a decisive role in the regulation of systemic inflammation. In chronic kidney disease in particular, the liver reacts in response to the uremic milieu, oxidative stress, endotoxemia and the decreased clearance of circulating proinflammatory cytokines by producing a large number of acute-phase reactants. Experimental tools to study inflammation and the underlying role of hepatocytes are crucial to understand the regulation and contribution of hepatic cytokines to a systemic acute phase response and a prolonged pro-inflammatory scenario, especially in an intricate setting such as chronic kidney disease. Since studying complex mechanisms of inflammation in vivo remains challenging, resource-intensive and usually requires the usage of transgenic animals, primary isolated hepatocytes provide a robust tool to gain mechanistic insights into the hepatic acute-phase response. Since this in vitro technique features moderate costs, high reproducibility and common technical knowledge, primary isolated hepatocytes can also be easily used as a screening approach. Here, we describe an enzymatic-based method to isolate primary murine hepatocytes, and we describe the assessment of an inflammatory response in these cells using ELISA and quantitative real-time PCR.


Sujet(s)
Hépatocytes/cytologie , Inflammation/anatomopathologie , Foie/cytologie , Réaction de polymérisation en chaine en temps réel/méthodes , Animaux , Techniques de culture cellulaire/méthodes , Cellules cultivées , Cytokines/métabolisme , Test ELISA/méthodes , Inflammation/induit chimiquement , Inflammation/métabolisme , Médiateurs de l'inflammation/métabolisme , Interleukine-6/effets indésirables , Lipopolysaccharides/toxicité , Souris , Perfusion , Reproductibilité des résultats
13.
Kidney Int ; 90(5): 985-996, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27457912

RÉSUMÉ

Patients with chronic kidney disease (CKD) develop increased levels of the phosphate-regulating hormone, fibroblast growth factor (FGF) 23, that are associated with a higher risk of mortality. Increases in inflammatory markers are another common feature that predicts poor clinical outcomes. Elevated FGF23 is associated with higher circulating levels of inflammatory cytokines in CKD, which can stimulate osteocyte production of FGF23. Here, we studied whether FGF23 can directly stimulate hepatic production of inflammatory cytokines in the absence of α-klotho, an FGF23 coreceptor in the kidney that is not expressed by hepatocytes. By activating FGF receptor isoform 4 (FGFR4), FGF23 stimulated calcineurin signaling in cultured hepatocytes, which increased the expression and secretion of inflammatory cytokines, including C-reactive protein. Elevating serum FGF23 levels increased hepatic and circulating levels of C-reactive protein in wild-type mice, but not in FGFR4 knockout mice. Administration of an isoform-specific FGFR4 blocking antibody reduced hepatic and circulating levels of C-reactive protein in the 5/6 nephrectomy rat model of CKD. Thus, FGF23 can directly stimulate hepatic secretion of inflammatory cytokines. Our findings indicate a novel mechanism of chronic inflammation in patients with CKD and suggest that FGFR4 blockade might have therapeutic anti-inflammatory effects in CKD.


Sujet(s)
Cytokines/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Hépatocytes/métabolisme , Inflammation/métabolisme , Insuffisance rénale chronique/métabolisme , Animaux , Calcineurine/métabolisme , Facteur-23 de croissance des fibroblastes , Glucuronidase/métabolisme , Humains , Protéines Klotho , Souris , Facteurs de transcription NFATC/métabolisme , Phospholipase C gamma/métabolisme , Culture de cellules primaires , Rats , Récepteur FGFR4/métabolisme , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE