Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Appl Physiol (1985) ; 136(4): 1007-1014, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38482570

RESUMO

Highly bioavailable inorganic phosphate (Pi) is present in large quantities in the typical Western diet and represents a large fraction of total phosphate intake. Dietary Pi excess induces exercise intolerance and skeletal muscle mitochondrial dysfunction in normal mice. However, the relevance of this to humans remains unknown. The study was conducted on 13 individuals without a history of cardiopulmonary disease (46% female, 15% Black participants) enrolled in the pilot-phase of the Dallas Heart and Mind Study. Total dietary phosphate was estimated from 24-h dietary recall (ASA24). Muscle ATP synthesis was measured at rest, and phosphocreatinine (PCr) dynamics was measured during plantar flexion exercise using 7-T 31P magnetic resonance (MR) spectroscopy in the calf muscle. Correlation was assessed between dietary phosphate intake normalized to total caloric intake, resting ATP synthesis, and PCr depletion during exercise. Higher dietary phosphate intake was associated with lower resting ATP synthesis (r = -0.62, P = 0.03), and with higher levels of PCr depletion during plantar flexion exercise relative to the resting period (r = -0.72; P = 0.004). These associations remain significant after adjustment for age and estimated glomerular filtration rate (both P < 0.05). High dietary phosphate intake was also associated with lower serum Klotho levels, and Klotho levels are in turn associated with PCr depletion and higher ADP accumulation post exercise. Our study suggests that higher dietary phosphate is associated with reduced skeletal muscle mitochondrial function at rest and exercise in humans providing new insight into potential mechanisms linking the Western diet to impaired energy metabolism.NEW & NOTEWORTHY This is the first translational research study directly demonstrating the adverse effects of dietary phosphate on muscle energy metabolism in humans. Importantly, our data show that dietary phosphate is associated with impaired muscle ATP synthesis at rest and during exercise, independent of age and renal function. This is a new biologic paradigm with significant clinical dietary implications.


Assuntos
Doenças Cardiovasculares , Fosfatos , Adulto , Humanos , Feminino , Animais , Camundongos , Masculino , Doenças Cardiovasculares/metabolismo , Músculo Esquelético/fisiologia , Metabolismo Energético/fisiologia , Trifosfato de Adenosina/metabolismo , Fosfocreatina/metabolismo
2.
J Am Soc Nephrol ; 32(12): 3051-3065, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34551996

RESUMO

BACKGROUND: The kidney is the source of sKlotho and kidney-specific loss of Klotho leads to a phenotype resembling the premature multiorgan failure phenotype in Klotho-hypomorphic mice ( kl/kl mice). Klotho and the Ca-sensing receptor (CaSR) are highly expressed in the distal convoluted tubule (DCT). The physiologic mechanisms that regulate sKlotho levels are unknown. METHODS: We measured sKlotho in WT and tubule-specific CaSR -/- (TS-CaSR -/- ) mice treated with calcimimetics, alkali, or acid, and Klotho shed from minced mouse kidneys, and from HEK-293 cells expressing the CaSR and Klotho, in response to calcimimetics, calcilytics, alkalotic and acidic pH, and ADAM protease inhibitors. The CaSR, Klotho, and ADAM10 were imaged in mouse kidneys and cell expression systems using confocal microscopy. RESULTS: The CaSR, Klotho, and ADAM10 colocalize on the basolateral membrane of the DCT. Calcimimetics and HCO 3 increase serum sKlotho levels in WT but not in CaSR -/- mice, and acidic pH suppresses sKlotho levels in WT mice. In minced kidneys and cultured cells, CaSR activation with high Ca, calcimimetics, or alkali increase shed Klotho levels via ADAM10, as demonstrated using the ADAM10 inhibitor GI254023X and siRNA. In cultured cells, the CaSR, Klotho, and ADAM10 form cell surface aggregates that disperse after CaSR activation. CONCLUSIONS: We identify a novel physiologic mechanism for regulation of sKlotho levels by the renal CaSR-ADAM10-Klotho pathway. We show that CaSR activators, including alkali, increase renal CaSR-stimulated Klotho shedding and predict that this mechanism is relevant to the effects of acidosis and alkali therapy on CKD progression.


Assuntos
Glucuronidase , Receptores de Detecção de Cálcio , Humanos , Camundongos , Animais , Receptores de Detecção de Cálcio/genética , Glucuronidase/metabolismo , Células HEK293 , Rim/metabolismo , Proteína ADAM10 , Concentração de Íons de Hidrogênio
3.
Am J Physiol Lung Cell Mol Physiol ; 321(4): L736-L749, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34346778

RESUMO

Normal lungs do not express α-Klotho (Klotho) protein but derive cytoprotection from circulating soluble Klotho. It is unclear whether chronic supranormal Klotho levels confer additional benefit. To address this, we tested the age-related effects of modest Klotho overexpression on acute lung injury (ALI) and recovery. Transgenic Klotho-overexpressing (Tg-Kl) and wild-type (WT) mice (2 and 6 mo old) were exposed to hyperoxia (95% O2; 72 h; injury; Hx) then returned to normoxia (21% O2; 24 h; recovery; Hx-R). Control mice were kept in normoxia. Renal and serum Klotho, lung histology, and bronchoalveolar lavage fluid oxidative damage markers were assessed. Effects of hyperoxia on Klotho release were tested in human embryonic kidney cells stably expressing Klotho. A549 lung epithelial cells transfected with Klotho cDNA or vector were exposed to cigarette smoke; lactate dehydrogenase and double-strand DNA breaks were measured. Serum Klotho decreased with age. Hyperoxia suppressed renal Klotho at both ages and serum Klotho at 2 mo of age. Tg-Kl mice at both ages and 2-mo-old WT mice survived Hx-R; 6-mo-old Tg-Kl mice showed lower lung damage than age-matched WT mice. Hyperoxia directly inhibited Klotho expression and release in vitro; Klotho transfection attenuated cigarette smoke-induced cytotoxicity and DNA double-strand breaks in lung epithelial cells. Young animals with chronic high baseline Klotho expression were more resistant to ALI. Chronic constitutive Klotho overexpression in older Tg-Kl animals attenuated hyperoxia-induced lung damage and improves survival and short-term recovery despite an acute reduction in serum Klotho during injury. We conclude that chronic enhancement of Klotho expression increases resilience to ALI.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Glucuronidase/sangue , Glucuronidase/metabolismo , Fumaça/efeitos adversos , Lesão Pulmonar Aguda/patologia , Animais , Linhagem Celular , Citoproteção/genética , Citoproteção/fisiologia , Quebras de DNA de Cadeia Dupla , Dano ao DNA/genética , Feminino , Glucuronidase/genética , Células HEK293 , Humanos , Hiperóxia , Proteínas Klotho , L-Lactato Desidrogenase/análise , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Transgênicos
4.
FASEB Bioadv ; 3(7): 531-540, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34258522

RESUMO

Alpha-Klotho is a multi-functional protein essential for maintenance of a myriad of cell functions. αKlotho is a single transmembrane protein with a large extracellular segment consisting of two domains (termed Kl1 and Kl2) which is shed into the extracellular fluid by proteolytic cleavage to furnish circulating soluble αKlotho. Based on cDNA sequence, an alternatively spliced mRNA is predicted to translate to a putative soluble αKlotho protein in mouse and human with only the Kl1 domain that represents a "spliced αKlotho Kl1" (spKl1) and is released from the cell without membrane targeting or cleavage. The existence of this protein remains in silico for two decades. We generated a novel antibody (anti-spE15) against the 15 amino acid epitope (E15; VSPLTKPSVGLLLPH) which is not present in Kl1 or full-length αKlotho and validated its specific reactivity against spKl1 in vitro. Using anti-spE15 and two well-established anti-αKlotho monoclonal antibodies, we performed immunoblots, immunoprecipitation, and immunohistochemistry to investigate for expression of spKl1 in the mouse brain. We found anti-spE15 labeling in mouse brain but were not able to see co-labelling of Kl1 and spE15 epitopes on the same protein, which is the pre-requisite for the existence of a spKl1 polypeptide, indicating that anti-spE15 likely binds to another protein other than the putative spKl1. In isolated choroid plexus from mouse brain, we found strong staining with anti-spE15, but did not find the spliced αKlotho transcript. We conclude that using reliable reagents and inclusion of proper controls, there is no evidence of the spKl1 protein in the mouse brain.

6.
Clin Kidney J ; 13(2): 235-244, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32297879

RESUMO

BACKGROUND: Soluble Klotho has multiple systemic salutary effects. In animals, both acute and chronic kidney disease models display systemic Klotho deficiency. As such, there is considerable interest in investigating soluble Klotho as a biomarker in patients with different types and severity of kidney diseases. Unfortunately, there remains uncertainty regarding the best method to measure soluble Klotho in human serum samples. METHODS: Using human serum samples obtained from several clinical cohorts with a wide range of kidney function, we measured soluble Klotho using a commercial enzyme-linked immunosorbent assay (ELISA) as well as with an immunoprecipitation-immunoblot (IP-IB) assay utilizing a synthetic antibody with high affinity and specificity for Klotho. Recovery of spiking with a known amount of exogenous Klotho was tested. A subset of samples was analyzed with and without the addition of a protease inhibitor cocktail at the time of collection or after the first freeze-thaw cycle to determine if these maneuvers influenced performance. RESULTS: The IP-IB assay was superior to the ELISA at recovery of exogenous Klotho (81-115% versus 60-81%) across the spectrum of kidney function. Klotho measurements by IP-IB were highly correlated with estimated glomerular filtration rate (eGFR) (R = 0.80, P < 0.001) in comparison with the commercial ELISA, which exhibited minimal correlation with eGFR (R = 0.18, P = 0.12). Use of a protease inhibitor cocktail neither improved nor impaired performance of the IP-IB assay; however, subsequent freeze-thaw cycle resulted in a significant reduction in Klotho recovery and dissipated the correlation between Klotho levels and eGFR. With the ELISA, the use of protease inhibitor cocktail resulted in an increase in intrasubject variability. CONCLUSIONS: The IP-IB assay is preferable to the commercial ELISA to measure soluble Klotho concentrations in never-thawed serum samples of humans with varying severity of kidney disease. However, due to the labor-intensive nature of the IP-IB assay, further research is needed to secure an assay suitable for high-throughput work.

7.
J Clin Endocrinol Metab ; 105(4)2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31774122

RESUMO

CONTEXT: α-klotho is an integral membrane protein that serves as a coreceptor for fibroblast growth factor 23 (FGF23) in conjunction with cognate fibroblast growth factor receptors. Proteolytic cleavage sheds the ectodomain of α-klotho (soluble α-klotho) as an endocrine substance into blood, urine, and cerebrospinal fluid. OBJECTIVE: To study the relationship of soluble α-klotho to mineral metabolism in the general population with mainly preserved kidney function. DESIGN: Cross-sectional analysis of the associations between soluble α-klotho with laboratory markers of markers of mineral metabolism in a population-based cohort. SETTING: Three centers in Switzerland including 1128 participants. MEASURES: Soluble full-length α-klotho levels by a specific immunoassay and markers of mineral metabolism. RESULTS: The median serum level of soluble α-klotho was 15.0 pmol/L. Multivariable analyses using α-klotho as the outcome variable revealed a sex-by-PTH interaction: In men, PTH was positively associated with α-klotho levels, whereas this association was negative in women. Plasma phosphate associated with soluble α-klotho levels in an age-dependent manner, changing from a positive association in young adults gradually to a negative association in the elderly. The decline of 1,25 (OH)2 vitamin D3 levels in parallel to the gradual impairment of kidney function was greatly attenuated in the setting of high circulating soluble α-klotho levels. CONCLUSIONS: Soluble α-klotho level is associated with plasma phosphate in an age-dependent manner and with PTH in a sex-dependent manner. Furthermore, our data reveal soluble α-klotho as a modulator of 1,25 (OH)2 vitamin D3 levels in individuals with preserved renal function.


Assuntos
Biomarcadores/sangue , Glucuronidase/sangue , Minerais/metabolismo , Hormônio Paratireóideo/sangue , Fosfatos/sangue , População Branca/estatística & dados numéricos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos Transversais , Feminino , Fator de Crescimento de Fibroblastos 23 , Seguimentos , Humanos , Proteínas Klotho , Masculino , Pessoa de Meia-Idade , Prognóstico , Adulto Jovem
8.
Am J Nephrol ; 49(5): 410-412, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30982040
9.
FASEB Bioadv ; 1(11): 675-687, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32123814

RESUMO

Alpha-Klotho (αKlotho), produced by the kidney and selected organs, is essential for tissue maintenance and protection. Homozygous αKlotho-deficiency leads to premature multi-organ degeneration and death; heterozygous insufficiency leads to apoptosis, oxidative stress, and increased injury susceptibility. There is inconsistent data in the literature regarding whether αKlotho is produced locally in the lung or derived from circulation. We probed murine and human lung by immunohistochemistry (IHC) and immunoblot (IB) using two monoclonal (anti-αKlotho Kl1 and Kl2 domains) and three other common commercial antibodies. Monoclonal anti-Kl1 and anti-Kl2 yielded no labeling in lung on IHC or IB; specific labeling was observed in kidney (positive control) and also murine lungs following tracheal delivery of αKlotho cDNA, demonstrating specificity and ability to detect artificial pulmonary expression. Other commercial antibodies labeled numerous lung structures (IHC) and multiple bands (IB) incompatible with known αKlotho mobility; labeling was not abolished by blocking with purified αKlotho or using lungs from hypomorphic αKlotho-deficient mice, indicating nonspecificity. Results highlight the need for rigorous validation of reagents. The lung lacks native αKlotho expression and derives full-length αKlotho from circulation; findings could explain susceptibility to lung injury in extrapulmonary pathology associated with reduced circulating αKlotho levels, for example, renal failure. Conversely, αKlotho may be artificially expressed in the lung, suggesting therapeutic opportunities.

10.
Crit Care Explor ; 1(6)2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32123869

RESUMO

OBJECTIVE: Klotho and Fibroblast Growth Factor (FGF)-23 were recently postulated as candidate biomarkers and/or therapeutic targets in acute kidney injury (AKI). We examined whether urine Klotho and serum intact FGF23 levels were differentially and independently associated with major adverse kidney events (MAKE) in critically ill patients with and without AKI. DESIGN: Single-center, prospective, case-control study. SETTING: ICU in a tertiary medical center. PATIENTS: 54 AKI patients and 52 controls without AKI admitted to the ICU. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: AKI was defined by KDIGO criteria and included only AKI stage ≥2. Controls were matched by age, gender, and baseline eGFR. Paired serum and urine samples were obtained 24-48h after AKI diagnosis (cases) or ICU admission (controls). The primary outcome was 90-day MAKE, which was the composite of all-cause death, dependence on renal replacement therapy or a 50% or higher decrease in eGFR from baseline. Forty-four (41.5%) patients developed MAKE-90. Patients who developed MAKE-90 had more comorbidity, higher acuity of illness scores and more prevalent AKI. Levels of urine Klotho adjusted by creatinine (Cr) were lower and serum intact FGF23 levels were higher in AKI patients vs. ICU controls. In adjusted models, the highest vs. lowest tertile of urine Klotho/Cr was independently associated with an overall 95% lower risk of MAKE-90 (81% lower risk in patients with AKI). The highest vs. lowest tertile of serum intact FGF23 was associated with >300% higher risk of MAKE-90. CONCLUSIONS: Urine Klotho/Cr levels were significantly lower and serum intact FGF23 levels significantly higher in critically ill patients with AKI vs. matched-controls without AKI. When measured in the first 48h of ICU admission or AKI diagnosis, urine Klotho/Cr independently associated with major adverse kidney events, particularly in patients with AKI. These results show promise for testing these biomarkers -individually or in combination- as part of novel risk-prediction models of renal outcomes in the ICU.

11.
Kidney Int ; 94(1): 49-59, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29735309

RESUMO

High circulating fibroblast growth factor 23 (FGF23) levels are probably a major risk factor for cardiovascular disease in chronic kidney disease. FGF23 interacts with the receptor FGFR4 in cardiomyocytes inducing left ventricular hypertrophy. Moreover, in the liver FGF23 via FGFR4 increases the risk of inflammation which is also found in chronic kidney disease. In contrast, X-linked hypophosphatemia is characterized by high FGF23 circulating levels due to loss of function mutations of the phosphate-regulating gene with homologies to an endopeptidase on the X chromosome (PHEX), but is not characterized by high cardiovascular morbidity. Here we used a novel murine X-linked hypophosphatemia model, the PhexC733RMhda mouse line, bearing an amino acid substitution (p.Cys733Arg) to test whether high circulating FGF23 in the absence of renal injury would trigger cardiovascular disease. As X-linked hypophosphatemia patient mimics, these mice show high FGF23 levels, hypophosphatemia, normocalcemia, and low/normal vitamin D levels. Moreover, these mice show hyperparathyroidism and low circulating soluble αKlotho levels. At the age of 27 weeks we found no left ventricular hypertrophy and no alteration of cardiac function as assessed by echocardiography. These mice also showed no activation of the calcineurin/NFAT pathway in heart and liver and no tissue and systemic signs of inflammation. Importantly, blood pressure, glomerular filtration rate and urea clearance were similar between genotypes. Thus, the presence of high circulating FGF23 levels alone in the absence of renal impairment and normal/high phosphate levels is not sufficient to cause cardiovascular disease.


Assuntos
Raquitismo Hipofosfatêmico Familiar/sangue , Fatores de Crescimento de Fibroblastos/sangue , Hipertrofia Ventricular Esquerda/epidemiologia , Animais , Modelos Animais de Doenças , Ecocardiografia , Raquitismo Hipofosfatêmico Familiar/genética , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Coração/diagnóstico por imagem , Humanos , Hipertrofia Ventricular Esquerda/sangue , Hipertrofia Ventricular Esquerda/diagnóstico , Hipertrofia Ventricular Esquerda/etiologia , Mutação com Perda de Função , Masculino , Camundongos , Camundongos Transgênicos , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Endopeptidase Neutra Reguladora de Fosfato PHEX/metabolismo , Fosfatos/sangue , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/complicações , Fatores de Risco , Microtomografia por Raio-X
12.
Stem Cells ; 36(4): 616-625, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29226550

RESUMO

Induced pluripotent stem cells (iPSCs) have been reported to alleviate organ injury, although the mechanisms of action remain unclear and administration of intact cells faces many limitations. We hypothesized that cell-free conditioned media (CM) containing the secretome of iPSCs possess antioxidative constituents that can alleviate pulmonary oxidant stress damage. We derived iPSCs from human dermal fibroblasts and harvested the CM. Addition of iPSC CM to cultured human alveolar type-1 epithelial cells mitigated hyperoxia-induced depletion of endogenous total antioxidant capacity while tracheal instillation of iPSC CM into adult rat lungs enhanced hyperoxia-induced increase in TAC. In both the in vitro and in vivo models, iPSC CM ameliorated oxidative damage to DNA, lipid, and protein, and activated the nuclear factor (erythroid 2)-related factor 2 (Nrf2) network of endogenous antioxidant proteins. Compared with control fibroblast-conditioned or cell-free media, iPSC CM is highly enriched with αKlotho at a concentration up to more than 10-fold of that in normal serum. αKlotho is an essential antioxidative cell maintenance and protective factor and an activator of the Nrf2 network. Immunodepletion of αKlotho reduced iPSC CM-mediated cytoprotection by ∼50%. Thus, the abundant αKlotho content significantly contributes to iPSC-mediated antioxidation and cytoprotection. Results uncover a major mechanism of iPSC action, suggest a fundamental role of αKlotho in iPSC maintenance, and support the translational potential of airway delivery of cell-free iPSC secretome for protection against lung injury. The targeted cell-free secretome-based approach may also be applicable to the amelioration of injury in other organs. Stem Cells 2018;36:616-625.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Células Epiteliais Alveolares/metabolismo , Antioxidantes/metabolismo , Glucuronidase/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Lesão Pulmonar Aguda/patologia , Células Epiteliais Alveolares/patologia , Animais , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Proteínas Klotho , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Ratos Sprague-Dawley
13.
Kidney Int ; 91(2): 324-337, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27914707

RESUMO

The kidney controls systemic calcium and phosphate levels and disturbances of its control mechanisms can lead to a variety of diseases. The insulin-sensitizing adipokine adiponectin is renoprotective and accelerates functional recovery following renal injury. However, unlike other adipokines, adiponectin is reduced in obesity. High adiponectin levels are also correlated with bone loss, suggestive of an additional action in mineral metabolism. Using knockout, wild-type, and adiponectin-overexpressing transgenic mice, we sought to identify the mechanistic basis for adiponectin's ability to regulate calcium and phosphate balance at the level of the kidney. Adiponectin knockout mice exhibited lower serum calcium, lower urinary calcium excretion, and markedly lower serum fibroblast growth factor 23 (FGF23) levels, although circulating klotho concentrations were significantly higher than in wild-type littermates. The transgenic mice exhibited lower bone mass and strength, particularly compared to adiponectin knockout mice. The transgenic mice were hyper-responsive to a 2% phosphate-enriched diet, exhibiting 2-fold higher serum FGF23 and concomitantly higher fractional phosphate excretion. These mice also excreted more calcium with calcium-enriched diet and had less renal klotho protein expression. In contrast, the knockout mice exhibited a smaller increase in FGF23 and maintained elevated klotho levels on both mineral challenges. Kidney-specific adiponectin expression in doxycycline-inducible adiponectin mice and adiponectin addition in vitro confirmed adiponectin's ability to reduce tubular epithelial cell klotho secretion. Thus, adiponectin alters calcium and phosphate balance and renal mineral excretion, in part, through klotho. This work highlights the profound effects of adipose tissue on renal function and has identified a new mechanism by which adiponectin may regulate bone mass.


Assuntos
Adiponectina/metabolismo , Cálcio da Dieta/metabolismo , Glucuronidase/sangue , Rim/metabolismo , Fosfatos/metabolismo , Fósforo na Dieta/metabolismo , Eliminação Renal , Adiponectina/deficiência , Adiponectina/genética , Animais , Fenômenos Biomecânicos , Cálcio da Dieta/sangue , Cálcio da Dieta/urina , Colágeno/metabolismo , Cães , Fêmur/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fibrose , Genótipo , Glucuronidase/genética , Homeostase , Hormônios/sangue , Rim/patologia , Túbulos Renais/metabolismo , Proteínas Klotho , Células Madin Darby de Rim Canino , Masculino , Camundongos Knockout , Osteogênese , Fenótipo , Fosfatos/sangue , Fosfatos/urina , Fósforo na Dieta/sangue , Fósforo na Dieta/urina , Coluna Vertebral/metabolismo , Transfecção
14.
PLoS One ; 10(10): e0139914, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26452228

RESUMO

Klotho transgenic mice exhibit resistance to oxidative stress as measured by their urinal levels of 8-hydroxy-2-deoxyguanosine, albeit this anti-oxidant defense mechanism has not been locally investigated in the brain. Here, we tested the hypothesis that the reactive oxygen species (ROS)-sensitive apoptosis signal-regulating kinase 1 (ASK1)/p38 MAPK pathway regulates stress levels in the brain of these mice and showed that: 1) the ratio of free ASK1 to thioredoxin (Trx)-bound ASK1 is relatively lower in the transgenic brain whereas the reverse is true for the Klotho knockout mice; 2) the reduced p38 activation level in the transgene corresponds to higher level of ASK1-bound Trx, while the KO mice showed elevated p38 activation and lower level of-bound Trx; and 3) that 14-3-3ζ is hyper phosphorylated (Ser-58) in the transgene which correlated with increased monomer forms. In addition, we evaluated the in vivo robustness of the protection by challenging the brains of Klotho transgenic mice with a neurotoxin, MPTP and analyzed for residual neuron numbers and integrity in the substantia nigra pars compacta. Our results show that Klotho overexpression significantly protects dopaminergic neurons against oxidative damage, partly by modulating p38 MAPK activation level. Our data highlight the importance of ASK1/p38 MAPK pathway in the brain and identify Klotho as a possible anti-oxidant effector.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Glucuronidase/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Sistema de Sinalização das MAP Quinases , Estresse Oxidativo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neurônios Dopaminérgicos/patologia , Ativação Enzimática , Glucuronidase/genética , Proteínas Klotho , MAP Quinase Quinase Quinase 5/genética , Camundongos , Camundongos Knockout , Oxirredução , Proteínas Quinases p38 Ativadas por Mitógeno/genética
15.
Nephrol Dial Transplant ; 30(2): 223-33, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25324355

RESUMO

BACKGROUND: αKlotho is the prototypic member of the Klotho family and is most highly expressed in the kidney. αKlotho has pleiotropic biologic effects, and in the kidney, its actions include regulation of ion transport, cytoprotection, anti-oxidation and anti-fibrosis. In rodent models of chronic kidney disease (CKD), αKlotho deficiency has been shown to be an early biomarker as well as a pathogenic factor. The database for αKlotho in human CKD remains controversial even after years of study. METHODS: We used a synthetic antibody library to identify a high-affinity human antigen-binding fragment that recognizes human, rat and mouse αKlotho primarily in its native, rather than denatured, form. RESULTS: Using an immunoprecipitation-immunoblot (IP-IB) assay, we measured both serum and urinary levels of full-length soluble αKlotho in humans and established that human CKD is associated with αKlotho deficiency in serum and urine. αKlotho levels were detectably lower in early CKD preceding disturbances in other parameters of mineral metabolism and progressively declined with CKD stages. We also found that exogenously added αKlotho is inherently unstable in the CKD milieu suggesting that decreased production may not be the sole reason for αKlotho deficiency. CONCLUSION: Synthetic antibody libraries harbor tremendous potential for a variety of biomedical and clinical applications. Using such a reagent, we furnish data in support of αKlotho deficiency in human CKD, and we set the foundation for the development of diagnostic and therapeutic applications of anti-αKlotho antibodies.


Assuntos
Anticorpos/sangue , Glucuronidase/deficiência , Insuficiência Renal Crônica/enzimologia , Animais , Biomarcadores/sangue , Estudos de Casos e Controles , Ensaio de Imunoadsorção Enzimática , Fatores de Crescimento de Fibroblastos/sangue , Glucuronidase/imunologia , Voluntários Saudáveis , Humanos , Immunoblotting , Fragmentos Fab das Imunoglobulinas/sangue , Imunoprecipitação , Proteínas Klotho , Camundongos , Biblioteca de Peptídeos , Ratos
16.
PLoS One ; 8(5): e63565, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23675491

RESUMO

Bone is constantly formed and resorbed throughout life by coordinated actions of osteoblasts and osteoclasts. However, the molecular mechanisms involved in osteoblast function remain incompletely understood. Here we show, for the first time, that the peptidyl-prolyl isomerase PIN1 controls the osteogenic activity of osteoblasts. Pin1 null mice exhibited an age-dependent decrease in bone mineral density and trabecular bone formation without alteration in cortical bone. Further analysis identified a defect in BMP signaling in Pin1 null osteoblasts but normal osteoclast function. PIN1 interacted with SMAD5 and was required for the expression by primary osteoblasts of osteoblast specific transcription factors (CBFA1 and OSX), ECM (collagen I and OCN) and the formation of bone nodules. Our results thus uncover a novel aspect of the molecular underpinning of osteoblast function and identify a new therapeutic target for bone diseases.


Assuntos
Densidade Óssea/genética , Desenvolvimento Ósseo/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Osso e Ossos/metabolismo , Peptidilprolil Isomerase/genética , Transdução de Sinais , Animais , Proteína Morfogenética Óssea 2/metabolismo , Osso e Ossos/diagnóstico por imagem , Cálcio/sangue , Diferenciação Celular , Colecalciferol/sangue , Expressão Gênica , Camundongos , Camundongos Knockout , Peptidilprolil Isomerase de Interação com NIMA , Osteoclastos/citologia , Osteoclastos/metabolismo , Peptidilprolil Isomerase/metabolismo , Ligação Proteica , Radiografia , Proteína Smad5/metabolismo
17.
J Biol Chem ; 287(34): 29134-46, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22733815

RESUMO

FGFs 19, 21, and 23 are hormones that regulate in a Klotho co-receptor-dependent fashion major metabolic processes such as glucose and lipid metabolism (FGF21) and phosphate and vitamin D homeostasis (FGF23). The role of heparan sulfate glycosaminoglycan in the formation of the cell surface signaling complex of endocrine FGFs has remained unclear. Here we show that heparan sulfate is not a component of the signal transduction unit of FGF19 and FGF23. In support of our model, we convert a paracrine FGF into an endocrine ligand by diminishing heparan sulfate-binding affinity of the paracrine FGF and substituting its C-terminal tail for that of an endocrine FGF containing the Klotho co-receptor-binding site to home the ligand into the target tissue. In addition to serving as a proof of concept, the ligand conversion provides a novel strategy for engineering endocrine FGF-like molecules for the treatment of metabolic disorders, including global epidemics such as type 2 diabetes and obesity.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Heparitina Sulfato/metabolismo , Modelos Biológicos , Comunicação Parácrina , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Sistema Endócrino/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Heparitina Sulfato/genética , Humanos , Camundongos , Camundongos Mutantes , Obesidade/genética , Obesidade/metabolismo , Obesidade/terapia
18.
Clin Exp Nephrol ; 16(3): 442-7, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22350461

RESUMO

BACKGROUND: Klotho has been investigated as an anti-aging protein that is predominantly expressed in the distal convoluted tubules in the kidneys and in the choroid plexus of the brain. The purpose of the present study was to determine the relationship between the soluble form of Klotho and renal function in chronic peritoneal dialysis (PD) patients, a relationship which remains poorly understood. METHODS: The soluble Klotho levels in the serum, urine, and peritoneal dialysate obtained from thirty-six PD patients were determined by a sandwich enzyme-linked immunosorbent assay system. RESULTS: The amount of urinary excreted soluble Klotho over 24 h ranged from 1.54 to 1774.4 ng/day (median 303.2 ng/day; interquartile range [IR] 84.1-498.5), while the serum soluble Klotho concentration ranged from 194.4 to 990.4 pg/ml (mean 553.7 ± 210.4 pg/ml). The amount of urinary Klotho excretion was significantly correlated with residual renal function. However, there was no apparent correlation between the serum soluble Klotho levels and the residual renal function. Klotho was also detected in the 24-h dialysate collections. There was a significant correlation between the peritoneal Klotho excretion and the amount of albumin contained in the dialysate collections (r = 0.798, p < 0.01). CONCLUSIONS: The total amount of urinary excreted Klotho, but not the serum level of soluble Klotho, may be a potential biomarker for assessing the residual renal function among PD patients. Whether our findings are also valid for chronic kidney disease patients overall should therefore be evaluated in greater detail.


Assuntos
Glucuronidase/metabolismo , Falência Renal Crônica/fisiopatologia , Rim/fisiopatologia , Biomarcadores/metabolismo , Glucuronidase/sangue , Glucuronidase/urina , Humanos , Proteínas Klotho , Diálise Peritoneal , Solubilidade
19.
Neurobiol Aging ; 33(7): 1483.e25-30, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22245317

RESUMO

Klotho is a putative age-suppressing gene whose overexpression in mice results in extension of life span. The Klotho gene encodes a single-pass transmembrane protein whose extracellular domain is shed and released into blood, urine, and cerebrospinal fluid, potentially functioning as a humoral factor. The extracellular domain of Klotho has an activity that increases the expression of antioxidant enzymes and confers resistance to oxidative stress in cultured cells and in whole animals. The transmembrane form of the Klotho protein directly binds to multiple fibroblast growth factor receptors and modifies their ligand affinity and specificity. The purpose of the present study was to determine the precise cellular localization of Klotho in the mouse brain. Using light microscopic immunohistochemical methods, we found the highest levels of Klotho immunoreactivity in 2 brain regions: the choroid plexus, and cerebellar Purkinje cells. In the choroid plexus cells, Klotho was found not only on the plasma membrane but also in large amounts near the nuclear membrane. Likewise, in the Purkinje cell Klotho was found throughout the cell including dendrites, axon and soma with large amounts near the nuclear membrane. Using immunoelectron microscopy, we found Klotho in the cell membrane, but the highest concentration was localized in the peripheral portion of the nucleus and the nucleolus in both cell types. This new finding suggests that in addition to Klotho being secreted from cells in brain, it also has a nuclear function.


Assuntos
Envelhecimento/metabolismo , Química Encefálica/genética , Nucléolo Celular/química , Glucuronidase/genética , Proteínas Nucleares/química , Animais , Nucléolo Celular/genética , Núcleo Celular/química , Núcleo Celular/genética , Glucuronidase/química , Proteínas Klotho , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Proteínas Nucleares/genética
20.
Proteome Sci ; 9: 53, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21917185

RESUMO

An improved version of quantitative protein array platform utilizing linear Quantum dot signaling for systematically measuring protein levels and phosphorylation states is presented. The signals are amplified linearly by a confocal laser Quantum dot scanner resulting in ~1000-fold more sensitivity than traditional Western blots, but are not linear by the enzyme-based amplification. Software is developed to facilitate the quantitative readouts of signaling network activities. Kinetics of EGFRvIII mutant signaling was analyzed to quantify cross-talks between EGFR and other signaling pathways.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...