Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Taiwan J Obstet Gynecol ; 60(4): 728-733, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34247815

RESUMO

OBJECTIVE: To examine the effects of cisplatin on uterine histology and implantation molecules and the possible protective role of recombinant Klotho administration on uterine histology and uterine receptivity in mice exposed to cisplatin. MATERIALS AND METHODS: This study was conducted using thirty-two adult female mice assigned to four groups with 8 mice in each group. Saline was given to the 1st group, cisplatin to the 2nd group, recombinant mouse Klotho to the 3rd group and recombinant mouse Klotho plus cisplatin to the 4th group. Uterine tissues were examined for damage histologically and immunobiologically for the uterine receptivity markers HOXA13 and alphaVBeta3 integrin. RESULTS: Apoptosis, degeneration, decrease in uterine thickness and uterine absence of gland scores were higher in the cisplatin group (3rd group) compared to the saline group (1st group) (cisplatin vs. saline p < 0.0001 for all parameters). In the recombinant Klotho plus cisplatin group (4th group), scores of apoptosis, degeneration, reduction in uterine thickness and uterine absence of gland were lower than the group receiving only cisplatin (cisplatin plus recombinant Klotho vs cisplatin, p = 0.006 for apoptosis; p = 0.017 for degeneration; p = 0.011 for the reduction in uterine thickness; p = 0.002 for the absence of gland). However, HOXA13 and alphaVBeta3 integrin staining levels were not different between the cisplatin group (group 3) and the cisplatin plus recombinant Klotho group (group 4) (p = 0.980 and p = 0.762, respectively.) CONCLUSION: Cisplatin has adverse effects on the uterus. Administration of recombinant Klotho was found to attenuate the cisplatin-induced damage but failed to preserve levels of the implantation molecules HOXA13 and alphaVbeta3. Further studies examining the effect of cisplatin toxicity using other implantation markers along with functional studies are needed.


Assuntos
Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Implantação do Embrião/efeitos dos fármacos , Proteínas de Homeodomínio/efeitos dos fármacos , Integrina alfaVbeta3/efeitos dos fármacos , Útero/metabolismo , Animais , Feminino , Glucuronidase/administração & dosagem , Proteínas Klotho , Camundongos , Modelos Animais , Substâncias Protetoras/administração & dosagem
2.
Mol Med Rep ; 22(2): 1035-1043, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32468073

RESUMO

Klotho is a type of single­pass transmembrane protein that is important for the proper function of numerous organs. The aim of the present study was to investigate the role of Klotho in sepsis­associated myocardial damage. In the present study, reverse transcription­quantitative PCR, western blotting and ELISA were conducted to examine the expression levels of function genes, and flow cytometry was performed to detect cell apoptosis and reactive oxygen species. The present study demonstrated that Klotho expression was significantly downregulated in septic mice and that the myocardial function of septic mice improved after treatment with exogenous Klotho protein. It further demonstrated that indoxyl sulfate inhibited the expression of Klotho protein. In addition, decreased Klotho protein further led to activation of the reactive oxygen species­p38 mitogen­activated protein kinase signaling pathway, finally resulting in myocardial damage. In conclusion, Klotho protein may be a key regulator in the myocardial damage of cardiorenal syndrome in sepsis. It also has a potential to be a therapeutic target for sepsis­associated myocardial damage in the future.


Assuntos
Síndrome Cardiorrenal/metabolismo , Glucuronidase/genética , Glucuronidase/metabolismo , Traumatismos Cardíacos/metabolismo , Sepse/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carbono/administração & dosagem , Síndrome Cardiorrenal/etiologia , Regulação para Baixo , Ecocardiografia , Glucuronidase/administração & dosagem , Glucuronidase/antagonistas & inibidores , Traumatismos Cardíacos/diagnóstico por imagem , Traumatismos Cardíacos/etiologia , Indicã/administração & dosagem , Indicã/sangue , Rim/metabolismo , Rim/patologia , Proteínas Klotho , Lipopolissacarídeos/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Miocárdio/patologia , Óxidos/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Sepse/induzido quimicamente , Sepse/complicações , Sepse/tratamento farmacológico , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Diabetes ; 69(7): 1368-1381, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32332158

RESUMO

α-Klotho is a circulating factor with well-documented antiaging properties. However, the central role of α-klotho in metabolism remains largely unexplored. The current study investigated the potential role of central α-klotho to modulate neuropeptide Y/agouti-related peptide (NPY/AgRP)-expressing neurons, energy balance, and glucose homeostasis. Intracerebroventricular administration of α-klotho suppressed food intake, improved glucose profiles, and reduced body weight in mouse models of type 1 and 2 diabetes. Furthermore, central α-klotho inhibition via an anti-α-klotho antibody impaired glucose tolerance. Ex vivo patch clamp electrophysiology and immunohistochemical analysis revealed that α-klotho suppresses NPY/AgRP neuron activity, at least in part, by enhancing miniature inhibitory postsynaptic currents. Experiments in hypothalamic GT1-7 cells observed that α-klotho induces phosphorylation of AKTser473, ERKthr202/tyr204, and FOXO1ser256 as well as blunts AgRP gene transcription. Mechanistically, fibroblast growth factor receptor 1 (FGFR1) inhibition abolished the downstream signaling of α-klotho, negated its ability to modulate NPY/AgRP neurons, and blunted its therapeutic effects. Phosphatidylinositol 3 kinase (PI3K) inhibition also abolished α-klotho's ability to suppress food intake and improve glucose clearance. These results indicate a prominent role of hypothalamic α-klotho/FGFR1/PI3K signaling in the modulation of NPY/AgRP neuron activity and maintenance of energy homeostasis, thus providing new insight into the pathophysiology of metabolic disease.


Assuntos
Proteína Relacionada com Agouti/fisiologia , Glucuronidase/fisiologia , Neurônios/fisiologia , Neuropeptídeo Y/fisiologia , Animais , Células Cultivadas , Diabetes Mellitus Experimental/tratamento farmacológico , Metabolismo Energético , Glucose/metabolismo , Glucuronidase/administração & dosagem , Infusões Intraventriculares , Proteínas Klotho , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/fisiologia
4.
Mol Genet Metab ; 130(1): 65-76, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32192868

RESUMO

Mucopolysaccharidosis VII (MPS VII) is a rare lysosomal storage disease characterized by a deficiency in the enzyme ß-glucuronidase that has previously been successfully treated in a mouse model with enzyme replacement therapy. Here, we present the generation of a novel, highly sialylated version of recombinant human ß-glucuronidase (rhGUS), vestronidase alfa, that has high uptake, resulting in an improved enzyme replacement therapy for the treatment of patients with MPS VII. In vitro, vestronidase alfa has 10-fold more sialic acid per mole of rhGUS monomer than a prior rhGUS version (referred to as GUS 43/44) and demonstrated very high affinity at ~1 nM half maximal uptake in human MPS VII fibroblasts. Vestronidase alfa has a longer enzymatic half-life after uptake into fibroblasts compared with other enzymes used as replacement therapy for MPS (40 days vs 3 to 4 days, respectively). In pharmacokinetic and tissue distribution experiments in Sprague-Dawley rats, intravenous administration of vestronidase alfa resulted in higher serum rhGUS levels and enhanced ß-glucuronidase activity distributed to target tissues. Weekly intravenous injections of vestronidase alfa (0.1 mg/kg to 20 mg/kg) in a murine model of MPS VII demonstrated efficient enzyme delivery to all tissues, including bone and brain, as well as reduced lysosomal storage of glycosaminoglycans (GAGs) in a dose-dependent manner, resulting in increased survival after 8 weeks of treatment. Vestronidase alfa was well-tolerated and demonstrated no toxicity at concentrations that reached 5-times the proposed clinical dose. In a first-in-human phase 1/2 clinical trial, a dose-dependent reduction in urine GAG levels was sustained over 38 weeks of treatment with vestronidase alfa. Together, these results support the therapeutic potential of vestronidase alfa as an enzyme replacement therapy for patients with MPS VII.


Assuntos
Terapia de Reposição de Enzimas/métodos , Glucuronidase/administração & dosagem , Glucuronidase/metabolismo , Lisossomos/enzimologia , Mucopolissacaridose VII/enzimologia , Mucopolissacaridose VII/terapia , Administração Intravenosa , Adulto , Animais , Células CHO , Criança , Cricetulus , Feminino , Fibroblastos/metabolismo , Glucuronidase/sangue , Glucuronidase/genética , Glucuronidase/farmacocinética , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/urina , Humanos , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Distribuição Tecidual/efeitos dos fármacos
5.
Mol Genet Metab ; 129(3): 219-227, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32063397

RESUMO

Vestronidase alfa (recombinant human beta-glucuronidase) is an enzyme replacement therapy (ERT) for Mucopolysaccharidosis (MPS) VII, a highly heterogeneous, ultra-rare disease. Twelve subjects, ages 8-25 years, completed a Phase 3, randomized, placebo-controlled, blind-start, single crossover study (UX003-CL301; NCT02377921), receiving 24-48 weeks of vestronidase alfa 4 mg/kg IV. All 12 subjects completed the blind-start study, which showed significantly reduced urinary glycosaminoglycans (GAG) and clinical improvement in a multi-domain responder index, and enrolled in a long-term, open-label, extension study (UX003-CL202; NCT02432144). Here, we report the final results of the extension study, up to an additional 144 weeks after completion of the blind-start study. Three subjects (25%) completed all 144 weeks of study, eight subjects (67%) ended study participation before Week 144 to switch to commercially available vestronidase alfa, and one subject discontinued due to non-compliance after receiving one infusion of vestronidase alfa in the extension study. The safety profile of vestronidase alfa in the extension study was consistent with observations in the preceding blind-start study, with most adverse events mild to moderate in severity. There were no treatment or study discontinuations due to AEs and no noteworthy changes in a standard safety chemistry panel. Out of the eleven subjects who tested positive for anti-drug antibodies at any time during the blind-start or extension study, including the baseline assessment in the blind-start study, seven subjects tested positive for neutralizing antibodies and all seven continued to demonstrate a reduction in urinary GAG levels. There was no association between antibody formation and infusion associated reactions. Subjects receiving continuous vestronidase alfa treatment showed a sustained urinary GAG reduction and clinical response evaluated using a multi-domain responder index that includes assessments in pulmonary function, motor function, range of motion, mobility, and visual acuity. Reduction in fatigue was also maintained in the overall population. As ERT is not expected to cross the blood brain barrier, limiting the impact on neurological signs of disease, and not all subjects presented with neurological symptoms, outcomes related to central nervous system pathology are not focused on in this report. Results from this study show the long-term safety and durability of clinical efficacy in subjects with MPS VII with long-term vestronidase alfa treatment.


Assuntos
Terapia de Reposição de Enzimas , Glucuronidase/uso terapêutico , Glicosaminoglicanos/urina , Mucopolissacaridose VII/tratamento farmacológico , Adolescente , Adulto , Anticorpos Neutralizantes , Barreira Hematoencefálica/efeitos dos fármacos , Criança , Estudos Cross-Over , Feminino , Glucuronidase/administração & dosagem , Glucuronidase/efeitos adversos , Glucuronidase/imunologia , Humanos , Masculino , Mucopolissacaridose VII/imunologia , Mucopolissacaridose VII/fisiopatologia , Doenças Raras/terapia , Resultado do Tratamento
6.
Am J Physiol Renal Physiol ; 318(3): F557-F564, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31928223

RESUMO

Klotho interacts with various membrane proteins such as receptors for transforming growth factor-ß (TGF-ß) and insulin-like growth factor (IGF). Renal expression of klotho is diminished in polycystic kidney disease (PKD). In the present study, the effects of klotho supplementation on PKD were assessed. Recombinant human klotho protein (10 µg·kg-1·day-1) or a vehicle was administered daily by subcutaneous injection to 6-wk-old mice with PKD (DBA/2-pcy). Blood pressure was measured using tail-cuff methods. After 2 mo, mice were killed, and the kidneys were harvested for analysis. Exogenous klotho protein supplementation reduced kidney weight, cystic area, systolic blood pressure, renal angiotensin II levels, and 8-epi-PGF2α excretion (P < 0.05). Klotho protein supplementation enhanced glomerular filtration rate, renal expression of superoxide dismutase, and klotho itself (P < 0.05). Klotho supplementation attenuated renal expressions of TGF-ß and collagen type I and diminished renal abundance of Twist, phosphorylated Akt, and mammalian target of rapamycin (P < 0.05). Pathological examination revealed that klotho decreased the fibrosis index and nuclear staining of Smad in PKD kidneys (P < 0.05). Our data indicate that klotho protein supplementation ameliorates the renin-angiotensin system, reducing blood pressure in PKD mice. Furthermore, the present results implicate klotho supplementation in the suppression of Akt/mammalian target of rapamycin signaling, slowing cystic expansion. Finally, our findings suggest that klotho protein supplementation attenuated fibrosis at least partly by inhibiting epithelial mesenchymal transition in PKD.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Glucuronidase/uso terapêutico , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/genética , Animais , Células Cultivadas , Feminino , Glucuronidase/administração & dosagem , Injeções Subcutâneas , Rim/fisiologia , Proteínas Klotho , Camundongos , Miofibroblastos/efeitos dos fármacos , Doenças Renais Policísticas/fisiopatologia , Proteínas Recombinantes
7.
Hum Mol Genet ; 28(21): 3610-3624, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31511867

RESUMO

Mucopolysaccharidosis (MPS) type VII is a lysosomal storage disease caused by ß-glucuronidase deficiency, prompting glycosaminoglycan accumulation in enlarged vesicles, leading to peripheral and neuronal dysfunction. Here, we present a gene therapy strategy using lumbar puncture of AAVrh10 encoding human ß-glucuronidase (AAVrh10-GUSB) to adult MPS VII mice. This minimally invasive technique efficiently delivers the recombinant vector to the cerebrospinal fluid (CSF) with a single intrathecal injection. We show that AAVrh10 delivery to the CSF allows global, stable transduction of CNS structures. In addition, drainage of AAVrh10-GUSB from the CSF to the bloodstream resulted in the transduction of somatic organs such as liver, which provided a systemic ß-glucuronidase source sufficient to achieve serum enzyme activity comparable to wild type mice. ß-glucuronidase levels were enough to correct biochemical and histopathological hallmarks of the disease in the CNS and somatic organs at short and long term. Moreover, the progression of the bone pathology was also reduced. Importantly, the biochemical correction led to a significant improvement in the physical, cognitive and emotional characteristics of MPS VII mice, and doubling their life span. Our strategy may have implications for gene therapy in patients with lysosomal storage diseases.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/genética , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/terapia , Animais , Comportamento Animal , Cognição , Dependovirus/metabolismo , Modelos Animais de Doenças , Emoções , Vetores Genéticos/metabolismo , Glucuronidase/administração & dosagem , Glucuronidase/genética , Glucuronidase/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Mucopolissacaridose VII/mortalidade , Mucopolissacaridose VII/psicologia , Sobrevida
8.
Am J Nephrol ; 49(5): 413-424, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30982029

RESUMO

BACKGROUND: Klotho treatment is a promising approach against kidney injury, but its clinical application is still undetermined. We developed a novel strategy to allow self-production of Klotho protein, using minicircle (MC) technology, and evaluated its feasibility in therapeutic Klotho delivery. METHODS: We engineered MC vectors to carry cassette sequences of Klotho and verified the self-production of Klotho protein from in HEK293T cells. We evaluated the location and persistence of delivered MC in vivo, and the duration of Klotho protein production from MCs by serial measurement of Klotho protein in blood. We subsequently evaluated the therapeutic potential of Klotho-encoding MCs in experimental model of renal injury. RESULTS: We confirmed the production of Klotho from MC by its significant availability in cells transfected with the MC, as well as in its conditioned medium, compared to that in cells transfected with parent vector. MCs were delivered in vivo by hydrodynamic injection via tail vein. After a single injection of MCs, red fluorescence protein was detected until 30 days in liver, and Klotho protein was maintained until 10 days in the blood, suggesting the production of Klotho protein from MCs via protein synthesis machinery in liver. Therapeutic effect of MC was confirmed by functional and histological improvement seen in mouse model of acute ischemia-reperfusion injury and unilateral ureteral obstruction. CONCLUSION: Together, these findings implied that self-generated Klotho protein, using MC technology, is functionally active and relevant as a therapeutic approach in renal injury.


Assuntos
Injúria Renal Aguda/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Glucuronidase/genética , Plasmídeos/administração & dosagem , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Animais , Meios de Cultivo Condicionados , Modelos Animais de Doenças , Estudos de Viabilidade , Vetores Genéticos/genética , Glucuronidase/administração & dosagem , Células HEK293 , Humanos , Microscopia Intravital , Rim/irrigação sanguínea , Rim/patologia , Proteínas Klotho , Masculino , Camundongos , Microscopia de Fluorescência , Plasmídeos/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Transfecção
9.
Neurobiol Aging ; 78: 18-28, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30851437

RESUMO

Alzheimer's disease (AD) is the most common type of senile dementia. The antiaging gene Klotho is reported to decline in the brain of patients and animals with AD. However, the role of Klotho in the progression of AD remains elusive. The present study explored the effects and underlying mechanism of Klotho in a mouse model of AD. The upregulation of cerebral Klotho expression was mediated by an intracerebroventricular injection of a lentiviral vector that encoded Klotho (LV-KL) in 7-month-old amyloid precursor protein/presenilin 1 transgenic mice. Three months later, LV-KL significantly induced Klotho overexpression in the brain and effectively ameliorated cognitive deficit and AD-like pathology in amyloid precursor protein/presenilin 1 mice. LV-KL induced autophagy activation and protein kinase B/mammalian target of rapamycin inhibition both in AD mice and BV2 murine microglia. These results suggest that the upregulation of Klotho expression in the brain may promote the autophagic clearance of amyloid beta and protect against cognitive deficits in AD mice. These findings highlight the preventive and therapeutic potential of Klotho for the treatment of AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Encéfalo/metabolismo , Disfunção Cognitiva/genética , Disfunção Cognitiva/terapia , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glucuronidase/administração & dosagem , Glucuronidase/genética , Lentivirus , Peptídeos beta-Amiloides/metabolismo , Animais , Autofagia , Modelos Animais de Doenças , Vetores Genéticos/farmacologia , Glucuronidase/metabolismo , Glucuronidase/farmacologia , Humanos , Injeções Intraventriculares , Proteínas Klotho , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Regulação para Cima
10.
BioDrugs ; 33(2): 233-240, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30848434

RESUMO

Mucopolysaccharidosis VII is an extremely rare, autosomal recessive lysosomal storage disorder characterized by a deficiency of ß-glucuronidase activity, resulting in partial degradation and accumulation of GAGs in numerous tissues throughout the body, with consequent cellular damage and organ dysfunction. Enzyme replacement therapy (ERT) with intravenous vestronidase alfa (Mepsevii™), a recombinant form of human ß-glucuronidase, is the first disease-specific therapy approved for the treatment of mucopolysaccharidosis VII in pediatric and adult patients. In the pivotal, blind start, phase 3 trial, 24 weeks of vestronidase alfa therapy significantly reduced urinary GAG (uGAG) excretion in patients with mucopolysaccharidosis VII. Based on a Multi-Domain Responder Index (MDRI; comprises six clinically important morbidity domains, with prespecified minimally important differences for each domain), most evaluable patients experienced an improvement in ≥ 1 domain during the 24-week primary assessment period (overall positive mean change of 0.5 domains). The clinical benefits of vestronidase alfa were sustained during longer-term treatment, as was the reduction in uGAG excretion. Vestronidase alfa has a manageable tolerability profile, with most adverse reactions of mild to moderate severity. Given the lack of treatment options and the clinical benefits it provides, intravenous vestronidase alfa is an important emerging ERT for patients with mucopolysaccharidosis VII.


Assuntos
Glucuronidase/administração & dosagem , Mucopolissacaridose VII/tratamento farmacológico , Adolescente , Adulto , Criança , Terapia de Reposição de Enzimas , Feminino , Glucuronidase/deficiência , Glucuronidase/farmacologia , Humanos , Lisossomos/metabolismo , Masculino , Mucopolissacaridose VII/enzimologia , Mucopolissacaridose VII/patologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Adulto Jovem
11.
Clin Pharmacokinet ; 58(5): 673-683, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30467742

RESUMO

INTRODUCTION: Mucopolysaccharidosis type VII (MPS VII, Sly Syndrome) is a progressive, debilitating, ultra-rare lysosomal storage disorder caused by the deficiency of ß-glucuronidase (GUS), an enzyme required for breakdown of glycosaminoglycans (GAGs). Vestronidase alfa, a recombinant human GUS, is an enzyme replacement therapy approved in the US and EU for the treatment of MPS VII. METHODS: The pharmacokinetics (PK) and pharmacodynamics (PD) of vestronidase alfa were evaluated in 23 adult and pediatric subjects with MPS VII enrolled in phase I-III clinical trials to optimize the clinical dosing regimen of vestronidase alfa. The serum concentration-time profiles were adequately described by a two-compartment population PK model incorporating subjects' body weight as the only significant covariate. RESULTS: Model-based simulations predicted a substantially decreased time duration of serum exposures exceeding the level of Kuptake (the in vitro determined vestronidase alfa concentration corresponding to 50% maximum rate of cellular uptake) for 4 or 8 mg/kg once every 4 weeks dosing, compared with 4 mg/kg once every other week (QOW) dosing by intravenous infusion, suggesting that given the same total monthly dose, the QOW dosing frequency should result in more efficient delivery to the GUS-deficient tissue cells, and therefore superior treatment efficacy. A standard inhibitory maximal effect model reasonably explained the observed pharmacological PD responses of reduction in urinary GAGs from pretreatment baseline, which appeared to have reached the plateau of maximal effect at the 4 mg/kg QOW dose. CONCLUSION: The modeling results, together with the clinical evidence of safety and efficacy, supported the recommended 4 mg/kg QOW dosing regimen of vestronidase alfa for pediatric and adult patients with MPS VII. CLINICAL TRIAL REGISTRATION: NCT01856218, NCT02418455, NCT02230566.


Assuntos
Glucuronidase/farmacocinética , Modelos Biológicos , Mucopolissacaridose VII/metabolismo , Adolescente , Adulto , Criança , Pré-Escolar , Simulação por Computador , Estudos Cross-Over , Terapia de Reposição de Enzimas , Feminino , Glucuronidase/administração & dosagem , Glucuronidase/sangue , Glicosaminoglicanos/urina , Humanos , Lactente , Masculino , Mucopolissacaridose VII/sangue , Mucopolissacaridose VII/tratamento farmacológico , Adulto Jovem
12.
Crit Care Med ; 46(12): e1196-e1203, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30239382

RESUMO

OBJECTIVES: To determine the applicability of recombinant Klotho to prevent inflammation and organ injury in sepsis in man and mice. DESIGN: Prospective, clinical laboratory study using "warm" human postmortem sepsis-acute kidney injury biopsies. Laboratory study using a mouse model of endotoxemia. SETTING: Research laboratory at a university teaching hospital. SUBJECTS: Adult patients who died of sepsis in the ICU and control patients undergoing total nephrectomy secondary to renal cancer; male C57BL/6 and Klotho haploinsufficient mice. INTERVENTIONS: Lipopolysaccharide (0.05 mg/kg) injection and kill after 4, 8, and 24 hours. Mice received recombinant Klotho (0.05 mg/kg) 30 minutes prior to lipopolysaccharide (1 mg/kg) injection. Mice treated with saline were included as controls. MEASUREMENTS AND MAIN RESULTS: Quantitative reverse transcription polymerase chain reaction and immunohistochemical staining were used to quantify Klotho messenger RNA and protein expression in the kidney of sepsis-acute kidney injury patients and the kidney and brain of mice. The messenger RNA and protein expression of damage markers, inflammatory cytokine, chemokines, and endothelial adhesion molecules were also determined in mice. Renal neutrophil influx was quantified. We found significantly lower renal Klotho messenger RNA and protein levels in sepsis-acute kidney injury biopsies than in control subjects. These findings were recapitulated in the kidney and brain of lipopolysaccharide-challenged mice. Decreased Klotho expression paralleled an increase in kidney damage markers neutrophil gelatinase-associated lipocalin and kidney injury molecule-1. Administration of recombinant Klotho prior to lipopolysaccharide injection attenuated organ damage, inflammation and endothelial activation in the kidney and brain of mice. Furthermore, less neutrophils infiltrated into the kidneys of recombinant Klotho mice compared with lipopolysaccharide only treated mice. CONCLUSIONS: Renal Klotho expression in human sepsis-acute kidney injury and in mouse models of sepsis was significantly decreased and correlated with renal damage. Recombinant Klotho intervention diminished organ damage, inflammation, and endothelial activation in the kidney and brain of lipopolysaccharide-challenged mice. Systemic Klotho replacement may potentially be an organ-protective therapy for septic patients to halt acute, inflammatory organ injury.


Assuntos
Glucuronidase/administração & dosagem , Glucuronidase/farmacologia , Insuficiência de Múltiplos Órgãos/prevenção & controle , Sepse/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Modelos Animais de Doenças , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Rim/fisiopatologia , Proteínas Klotho , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Estudos Prospectivos , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes
13.
Mol Genet Metab ; 123(4): 488-494, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29478819

RESUMO

BACKGROUND: Drug development for ultra-rare diseases is challenging because small sample sizes and heterogeneous study populations hamper the ability of randomized, placebo-controlled trials with a single primary endpoint to demonstrate valid treatment effects. METHODS: To overcome these challenges, a novel Blind Start design was utilized in a study of vestronidase alfa in mucopolysaccharidosis VII (Sly syndrome), an ultra-rare lysosomal disease, that demonstrates the strengths of this approach in a challenging drug-development setting. Twelve subjects were randomized to 1 of 4 blinded groups, each crossing over to active treatment in a blinded fashion at different timepoints with efficacy analysis comparing the last assessment before cross over to after 24 weeks of treatment. Study assessments included: Percentage change from baseline in urinary GAG (uGAG); a Multi-Domain Responder Index (MDRI) using prespecified minimal important differences (6-Minute Walk Test, Forced Vital Capacity, shoulder flexion, visual acuity, and Bruininks-Oseretsky Test of Motor Proficiency); fatigue as assessed by the Pediatric Quality of Life Inventory™ Multidimensional Fatigue Scale; and safety. RESULTS: Vestronidase alfa treatment for 24 weeks significantly reduced uGAG excretion (dermatan sulfate: 64.8%, p < 0.0001). Most subjects (10/12) had a clinically meaningful improvement in at least one MDRI domain with an overall mean change (±SD) of +0.5 (±0.8) at Treatment Week 24 (p = 0.0527). Exposure-adjusted incidence rates of adverse events were similar between groups. CONCLUSIONS: The Blind Start study and MDRI design improve statistical power that enhances detection of a positive treatment effect in this rare heterogeneous disease and could be utilized for other ultra-rare diseases.


Assuntos
Glucuronidase/administração & dosagem , Mucopolissacaridose VII/terapia , Proteínas Recombinantes/administração & dosagem , Adolescente , Adulto , Criança , Feminino , Seguimentos , Glucuronidase/deficiência , Humanos , Masculino , Mucopolissacaridose VII/enzimologia , Mucopolissacaridose VII/patologia , Prognóstico , Adulto Jovem
14.
Cell Rep ; 20(6): 1360-1371, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28793260

RESUMO

Cognitive dysfunction and decreased mobility from aging and neurodegenerative conditions, such as Parkinson and Alzheimer diseases, are major biomedical challenges in need of more effective therapies. Increasing brain resilience may represent a new treatment strategy. Klotho, a longevity factor, enhances cognition when genetically and broadly overexpressed in its full, wild-type form over the mouse lifespan. Whether acute klotho treatment can rapidly enhance cognitive and motor functions or induce resilience is a gap in our knowledge of its therapeutic potential. Here, we show that an α-klotho protein fragment (αKL-F), administered peripherally, surprisingly induced cognitive enhancement and neural resilience despite impermeability to the blood-brain barrier in young, aging, and transgenic α-synuclein mice. αKL-F treatment induced cleavage of the NMDAR subunit GluN2B and also enhanced NMDAR-dependent synaptic plasticity. GluN2B blockade abolished αKL-F-mediated effects. Peripheral αKL-F treatment is sufficient to induce neural enhancement and resilience in mice and may prove therapeutic in humans.


Assuntos
Adaptação Psicológica , Encéfalo/efeitos dos fármacos , Cognição , Glucuronidase/farmacologia , Fragmentos de Peptídeos/farmacologia , alfa-Sinucleína/genética , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/fisiologia , Feminino , Glucuronidase/administração & dosagem , Glucuronidase/química , Proteínas Klotho , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/química , Proteólise , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/metabolismo
15.
PLoS One ; 12(5): e0176817, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28463984

RESUMO

Endothelial cells (ECs) express fibroblast growth factor (FGF) receptors and are metabolically active after treatment with FGF-23. It is not known if this effect is α-Klotho independent or mediated by humoral or endogenous endothelial α-Klotho. In the present study, we aimed to characterize EC α-Klotho expression within the human vascular tree and to investigate the potential role of α-Klotho in determining FGF-23 mediated EC regulation. Human tissue and ECs from various organs were used for immunohistochemistry and Western blot. Primary cultures of human aortic endothelial cells (HAECs) and human brain microvascular endothelial cells (HBMECs) were used to generate in vitro cell models. We found endogenous α-Klotho expression in ECs from various organs except in microvascular ECs from human brain. Furthermore, FGF-23 stimulated endothelial nitric oxide synthase (eNOS) expression, nitric oxide (NO) production, and cell proliferation in HAECs. Interestingly, these effects were not observed in our HBMEC model in vitro. High phosphate treatment and endothelial α-Klotho knockdown mitigated FGF-23 mediated eNOS induction, NO production, and cell proliferation in HAECs. Rescue treatment with soluble α-Klotho did not reverse endothelial FGF-23 resistance caused by reduced or absent α-Klotho expression in HAECs. These novel observations provide evidence for differential α-Klotho functional expression in the human endothelium and its presence may play a role in determining the response to FGF-23 in the vascular tree. α-Klotho was not detected in cerebral microvascular ECs and its absence may render these cells nonresponsive to FGF-23.


Assuntos
Aorta/metabolismo , Células Endoteliais/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Aorta/citologia , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Encéfalo/metabolismo , Fármacos Cardiovasculares/administração & dosagem , Proliferação de Células/fisiologia , Células Cultivadas , Células Endoteliais/citologia , Fator de Crescimento de Fibroblastos 23 , Técnicas de Silenciamento de Genes , Glucuronidase/administração & dosagem , Glucuronidase/deficiência , Glucuronidase/genética , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imuno-Histoquímica , Proteínas Klotho , Microvasos/citologia , Microvasos/metabolismo , Óxido Nítrico/metabolismo , Fosfatos , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Blood ; 129(19): 2667-2679, 2017 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-28264799

RESUMO

Thrombosis is a common complication of chronic kidney disease (CKD), but the causes and mechanisms of CKD-associated thrombosis are not well clarified. Here, we show that platelet activity is remarkably enhanced in CKD mice, with increase of serum indoxyl sulfate (IS), a typical uremic toxin, which cannot be effectively cleared by routine dialysis. Ex vivo and in vitro experiments reveal that IS displays a distinct ability to enhance platelet activities, including elevated response to collagen and thrombin, increases in platelet-derived microparticles, and platelet-monocyte aggregates. The flow chamber assay and carotid artery thrombosis model demonstrate that IS-induced platelet hyperactivity contributes to thrombus formation. Further investigations disclose that reactive oxygen species (ROS)-mediated p38MAPK signaling plays a key role in IS-induced platelet hyperactivity. Moreover, we show that Klotho, which is expressed dominantly in the kidneys, has the capacity to counteract IS-induced platelet hyperactivity by inhibiting ROS/p38MAPK signaling, whereas Klotho reduction may aggravate the effect of IS on platelet activation in CKD and klotho+/- mice. Finally, we demonstrate that Klotho protein treatment can protect against IS-induced thrombosis and atherosclerosis in apoE-/- mice. Our findings uncover the mechanism of platelet hyperactivity induced by IS and provide new insights into the pathogenesis and treatment of CKD-associated thrombosis.


Assuntos
Plaquetas/efeitos dos fármacos , Indicã/efeitos adversos , Ativação Plaquetária/efeitos dos fármacos , Insuficiência Renal Crônica/induzido quimicamente , Trombose/induzido quimicamente , Animais , Plaquetas/patologia , Glucuronidase/administração & dosagem , Glucuronidase/metabolismo , Glucuronidase/uso terapêutico , Proteínas Klotho , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Insuficiência Renal Crônica/metabolismo , Trombose/tratamento farmacológico , Trombose/metabolismo
17.
Kidney Int ; 91(5): 1104-1114, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28131398

RESUMO

α-Klotho is highly expressed in the kidney, and its extracellular domain is cleaved and released into the circulation. Chronic kidney disease (CKD) is a state of α-Klotho deficiency, which exerts multiple negative systemic effects on numerous organs including the cardiovascular system. Since acute kidney injury (AKI) greatly escalates the risk of CKD development, we explored the effect of α-Klotho on prevention and treatment on post-AKI to CKD progression and cardiovascular disease. Therein, ischemia reperfusion injury-induced AKI was followed by early administration of recombinant α-Klotho or vehicle starting one day and continued for four days after kidney injury (CKD prevention protocol). A CKD model was generated by unilateral nephrectomy plus contralateral ischemia reperfusion injury. Late administration of α-Klotho in this model was started four weeks after injury and sustained for 12 weeks (CKD treatment protocol). The prevention protocol precluded AKI to CKD progression and protected the heart from cardiac remodeling in the post-AKI model. One important effect of exogenous α-Klotho therapy was the restoration of endogenous α-Klotho levels long after the cessation of exogenous α-Klotho therapy. The treatment protocol still effectively improved renal function and attenuated cardiac remodeling in CKD, although these parameters did not completely return to normal. In addition, α-Klotho administration also attenuated high phosphate diet-induced renal and cardiac fibrosis, and improved renal and cardiac function in the absence of pre-existing renal disease. Thus, recombinant α-Klotho protein is safe and efficacious, and might be a promising prophylactic or therapeutic option for prevention or retardation of AKI-to-CKD progression and uremic cardiomyopathy.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Cardiomiopatias/tratamento farmacológico , Glucuronidase/metabolismo , Glucuronidase/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Injúria Renal Aguda/sangue , Injúria Renal Aguda/complicações , Injúria Renal Aguda/metabolismo , Animais , Terapia Biológica/métodos , Cardiomiopatias/etiologia , Modelos Animais de Doenças , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Feminino , Fibrose , Glucuronidase/administração & dosagem , Humanos , Injeções Intraperitoneais , Rim/metabolismo , Rim/patologia , Proteínas Klotho , Masculino , Camundongos , Miocárdio/patologia , Proteínas Recombinantes/uso terapêutico , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo , Uremia/complicações
18.
J Am Soc Nephrol ; 28(4): 1162-1174, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27837149

RESUMO

αKlotho (αKL) regulates mineral metabolism, and diseases associated with αKL deficiency are characterized by hyperphosphatemia and vascular calcification (VC). αKL is expressed as a membrane-bound protein (mKL) and recognized as the coreceptor for fibroblast growth factor-23 (FGF23) and a circulating soluble form (cKL) created by endoproteolytic cleavage of mKL. The functions of cKL with regard to phosphate metabolism are unclear. We tested the ability of cKL to regulate pathways and phenotypes associated with hyperphosphatemia in a mouse model of CKD-mineral bone disorder and αKL-null mice. Stable delivery of adeno-associated virus (AAV) expressing cKL to diabetic endothelial nitric oxide synthase-deficient mice or αKL-null mice reduced serum phosphate levels. Acute injection of recombinant cKL downregulated the renal sodium-phosphate cotransporter Npt2a in αKL-null mice supporting direct actions of cKL in the absence of mKL. αKL-null mice with sustained AAV-cKL expression had a 74%-78% reduction in aorta mineral content and a 72%-77% reduction in mineral volume compared with control-treated counterparts (P<0.01). Treatment of UMR-106 osteoblastic cells with cKL + FGF23 increased the phosphorylation of extracellular signal-regulated kinase 1/2 and induced Fgf23 expression. CRISPR/Cas9-mediated deletion of fibroblast growth factor receptor 1 (FGFR1) or pretreatment with inhibitors of mitogen-activated kinase kinase 1 or FGFR ablated these responses. In summary, sustained cKL treatment reduced hyperphosphatemia in a mouse model of CKD-mineral bone disorder, and it reduced hyperphosphatemia and prevented VC in mice without endogenous αKL. Furthermore, cKL stimulated Fgf23 in an FGFR1-dependent manner in bone cells. Collectively, these findings indicate that cKL has mKL-independent activity and suggest the potential for enhancing cKL activity in diseases of hyperphosphatemia with associated VC.


Assuntos
Glucuronidase/uso terapêutico , Hiperfosfatemia/tratamento farmacológico , Calcificação Vascular/tratamento farmacológico , Animais , Osso e Ossos/metabolismo , Doença Crônica , Nefropatias Diabéticas/complicações , Modelos Animais de Doenças , Feminino , Fator de Crescimento de Fibroblastos 23 , Glucuronidase/administração & dosagem , Glucuronidase/fisiologia , Hiperfosfatemia/etiologia , Proteínas Klotho , Masculino , Camundongos , Camundongos Knockout
19.
Mol Genet Metab ; 119(3): 249-257, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27692945

RESUMO

Severe, progressive skeletal dysplasia is a major symptom of multiple mucopolysaccharidoses (MPS) types. While a gene therapy approach initiated at birth has been shown to prevent the development of bone pathology in different animal models of MPS, the capacity to correct developed bone disease is unknown. In this study, ex vivo micro-computed tomography was used to demonstrate that bone mass and architecture of murine MPS VII L5 vertebrae were within the normal range at 1month of age but by 2months of age were significantly different to normal. The difference between normal and MPS VII BV/TV increased with age reaching a maximal difference at approximately 4months of age. In mature MPS VII bone BV/TV is increased (51.5% versus 21.5% in normal mice) due to an increase in trabecular number (6.2permm versus 3.8permm in normal mice). The total number of osteoclasts in the metaphysis of MPS VII mice was decreased, as was the percentage of osteoclasts attached to bone. MPS VII osteoblasts produced significantly more osteoprotegerin (OPG) than normal osteoblasts and supported the production of fewer osteoclasts from spleen precursor cells than normal osteoblasts in a co-culture system. In contrast, the formation of osteoclasts from MPS VII spleen monocytes was similar to normal in vitro, when exogenous RANKL and m-CSF was added to the culture medium. Administration of murine ß-glucuronidase to MPS VII mice at 4months of age, when bone disease was fully manifested, using lentiviral gene delivery resulted in a doubling of osteoclast numbers and a significant increase in attachment capacity (68% versus 29.4% in untreated MPS VII animals). Bone mineral volume rapidly decreased by 39% after gene therapy and fell within the normal range by 6months of age. Collectively, these results indicate that lentiviral-mediated gene therapy is effective in reversing established skeletal pathology in murine MPS VII.


Assuntos
Densidade Óssea/genética , Terapia Genética , Glucuronidase/genética , Mucopolissacaridose VII/terapia , Animais , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Glucuronidase/administração & dosagem , Humanos , Lentivirus/genética , Camundongos , Mucopolissacaridose VII/diagnóstico por imagem , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/patologia , Osteoprotegerina/genética , Microtomografia por Raio-X
20.
J Cereb Blood Flow Metab ; 36(3): 476-86, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26661222

RESUMO

The adult blood-brain barrier, unlike the neonatal blood-brain barrier, does not transport lysosomal enzymes into brain, making enzyme replacement therapy ineffective in treating the central nervous system symptoms of lysosomal storage diseases. However, enzyme transport can be re-induced with alpha-adrenergics. Here, we examined agents that are known to alter the blood-brain barrier transport of large molecules or to induce lysosomal enzyme transport across the blood-brain barrier ((±)epinephrine, insulin, retinoic acid, and lipopolysaccharide) in 2-week-old and adult mice. In 2-week-old adolescent mice, all these pharmacologic agents increased brain and heart uptake of phosphorylated human ß-glucuronidase. In 8-week-old adult mice, manipulations with (±)epinephrine, insulin, and retinoic acid were significantly effective on uptake by brain and heart. The increased uptake of phosphorylated human ß-glucuronidase was inhibited by mannose 6-phosphate for the agents (±)epinephrine and retinoic acid and by L-NG-nitroarginine methyl ester for the agent lipopolysaccharide in neonatal and adult mice. An in situ brain perfusion study revealed that retinoic acid directly modulated the transport of phosphorylated human ß-glucuronidase across the blood-brain barrier. The present study indicates that there are multiple opportunities to at least transiently induce phosphorylated human ß-glucuronidase transport at the adult blood-brain barrier.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Glucuronidase/metabolismo , Lisossomos/enzimologia , Transporte Proteico/efeitos dos fármacos , Tretinoína/farmacologia , Animais , Glucuronidase/administração & dosagem , Humanos , Masculino , Camundongos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA