Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
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
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.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Mol Genet Metab ; 114(2): 203-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25468648

RESUMO

Mucopolysaccharidosis type VII (MPS VII, Sly syndrome) is a very rare lysosomal storage disease caused by a deficiency of the enzyme ß-glucuronidase (GUS), which is required for the degradation of three glycosaminoglycans (GAGs): dermatan sulfate, heparan sulfate, and chondroitin sulfate. Progressive accumulation of these GAGs in lysosomes leads to increasing dysfunction in numerous tissues and organs. Enzyme replacement therapy (ERT) has been used successfully for other MPS disorders, but there is no approved treatment for MPS VII. Here we describe the first human treatment with recombinant human GUS (rhGUS), an investigational therapy for MPS VII, in a 12-year old boy with advanced stage MPS VII. Despite a tracheostomy, nocturnal continuous positive airway pressure, and oxygen therapy, significant pulmonary restriction and obstruction led to oxygen dependence and end-tidal carbon dioxide (ETCO2) levels in the 60-80mmHg range, eventually approaching respiratory failure (ETCO2 of 100mmHg) and the need for full-time ventilation. Since no additional medical measures could improve his function, we implemented experimental ERT by infusing rhGUS at 2mg/kg over 4h every 2 weeks for 24 weeks. Safety was evaluated by standard assessments and observance for any infusion associated reactions (IARs). Urinary GAG (uGAG) levels, pulmonary function, oxygen dependence, CO2 levels, cardiac valve function, liver and spleen size, and growth velocity were assessed to evaluate response to therapy. rhGUS infusions were well tolerated. No serious adverse events (SAEs) or IARs were observed. After initiation of rhGUS infusions, the patient's uGAG excretion decreased by more than 50%. Liver and spleen size were reduced within 2 weeks of the first infusion and reached normal size by 24 weeks. Pulmonary function appeared to improve during the course of treatment based on reduced changes in ETCO2 after off-ventilator challenges and a reduced oxygen requirement. The patient regained the ability to eat orally, gained weight, and his energy and activity levels increased. Over 24 weeks, treatment with every-other-week infusions of rhGUS was well tolerated with no SAEs, IARs, or hypersensitivity reactions and was associated with measurable improvement in objective clinical measures and quality of life.


Assuntos
Terapia de Reposição de Enzimas , Glucuronidase/administração & dosagem , Glucuronidase/uso terapêutico , Mucopolissacaridose VII/tratamento farmacológico , Administração Intravenosa , Peso Corporal/efeitos dos fármacos , Criança , Dermatan Sulfato/urina , Glucuronidase/efeitos adversos , Glucuronidase/genética , Glicosaminoglicanos/urina , Heparitina Sulfato/urina , Hepatomegalia/tratamento farmacológico , Hepatomegalia/patologia , Humanos , Masculino , Mucopolissacaridose VII/patologia , Mucopolissacaridose VII/fisiopatologia , Qualidade de Vida , Esplenomegalia/tratamento farmacológico , Esplenomegalia/patologia , Terapias em Estudo
13.
Poult Sci ; 103(11): 104166, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39214054

RESUMO

This novel study investigated the effects of intracerebroventricular (ICV) injection α- klotho and its interaction with neuropeptide Y (NPY) receptors on food intake in broiler chicken. This study included 4 experiments with 4 groups in each with 11 replicates per group. Birds were feed deprived 3 h prior injection, following injection returned to their cage and food provided. In experiment 1, group 1 received ICV injection of the saline and groups 2 to 4 received ICV injection of the α-klotho (1, 2, and 4 µg), respectively. In experiment 2, chicken received ICV injection of the saline, B5063 (NPY1 receptor antagonist, 1.25 µg), α-klotho (4 µg) and co-injection of the B5063 + α-klotho. In experiments 3 and 4, SF22 (NPY2 receptor antagonist, 1.25 µg), and SML0891 (NPY5 receptor antagonist, 1.25 µg) were injected instead of the B5063. Then consumed food was measured at 30, 60, and 120 min post the injection. Based on results, ICV injection of the α-klotho (2 and 4 µg) significantly decreased food intake (P < 0.05). Co-injection of the B5063 + α-klotho significantly amplified hypophagic effect of the α-klotho (P < 0.05). α-klotho-induced hypophagia was not influenced by SF22 or SML0891. These results suggest that α-klotho-induced hypophagia is mediated via NPY1 receptors in broiler chicken.


Assuntos
Galinhas , Ingestão de Alimentos , Glucuronidase , Proteínas Klotho , Animais , Masculino , Proteínas Aviárias/metabolismo , Proteínas Aviárias/administração & dosagem , Galinhas/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Glucuronidase/metabolismo , Glucuronidase/administração & dosagem , Injeções Intraventriculares/veterinária , Proteínas Klotho/administração & dosagem , Receptores de Neuropeptídeo Y/metabolismo
14.
Nat Genet ; 4(2): 154-9, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8348154

RESUMO

Genetic defects of lysosomal hydrolases result in severe storage diseases and treatments based on enzyme replacement have been proposed. In mice lacking beta-glucuronidase, which develop a disease homologous to human mucopolysaccharidosis type VII (Sly syndrome), we have used autologous implants of genetically-modified skin fibroblasts for the continuous in vivo production of the enzyme. The human beta-glucuronidase cDNA was introduced with a retroviral vector into mutant mice skin fibroblasts grown in primary culture. Fourteen mutant mice were implanted intraperitoneally with these modified cells embedded into collagen lattices. All animals expressed beta-glucuronidase from the vascularized neo-organs that developed after implantation and accumulated the enzyme in their tissues. A complete disappearance of the lysosomal storage lesions was observed in their liver and spleen.


Assuntos
Modelos Animais de Doenças , Fibroblastos/transplante , Terapia Genética/métodos , Glucuronidase/deficiência , Fígado/enzimologia , Lisossomos/enzimologia , Camundongos Mutantes/genética , Mucopolissacaridose VII/terapia , Próteses e Implantes , Proteínas Recombinantes de Fusão/uso terapêutico , Baço/enzimologia , Animais , Células Cultivadas , Fibroblastos/enzimologia , Vetores Genéticos , Glucuronidase/administração & dosagem , Glucuronidase/genética , Glucuronidase/uso terapêutico , Glicosaminoglicanos/metabolismo , Fígado/patologia , Camundongos , Mucopolissacaridose VII/enzimologia , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/patologia , Especificidade de Órgãos , Cavidade Peritoneal , Fenótipo , Proteínas Recombinantes de Fusão/administração & dosagem , Retroviridae/genética , Baço/patologia , Transfecção
15.
Molecules ; 18(6): 7179-93, 2013 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-23783456

RESUMO

Stichopus japonicus acid mucopolysaccharide (SJAMP) is an important biologically active compound that can be extracted from the body wall of the sea cucumber. The present study investigated the anti-tumor and immunomodulatory effects of SJAMP in an experimental hepatocellular carcinoma (HCC) model in rats. Three doses of SJAMP (17.5 mg/kg, 35 mg/kg, and 70 mg/kg administered 5 days/week via oral gavage) were given to rats with diethylnitrosamine (DEN)-induced HCC. SJAMP treatment significantly inhibited DEN-induced HCC by reducing both the number and mean volume of nodules, decreasing serum a-fetoprotein (AFP) levels and proliferating cell nuclear antigen (PCNA) expression in liver, and increasing p21 expression. Furthermore, SJAMP decreased the serum levels of ALT, AST, GGT and TNF-α and increased serum IL-2. SJAMP administration also improved indices of spleen and thymus function and improved both macrophage phagocytosis and NK cell-mediated tumoricidal activity. Moreover, CD3+ and CD4+ T lymphocyte levels recovered significantly and the CD4+/CD8+ T cell ratio normalized in a dose-dependent manner. In conclusion, SJAMP effectively inhibited the growth of HCC through the stimulation of immune organs and tissue proliferation, leading to the enhancement of cellular immunity pathways in rats.


Assuntos
Carcinoma Hepatocelular/imunologia , Glucuronidase/farmacologia , Fatores Imunológicos/farmacologia , Imunomodulação/efeitos dos fármacos , Neoplasias Hepáticas Experimentais/imunologia , Neoplasias Hepáticas/imunologia , Liases/farmacologia , Stichopus/química , Animais , Biomarcadores Tumorais/metabolismo , Biópsia , Peso Corporal/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Citocinas/sangue , Citotoxicidade Imunológica/efeitos dos fármacos , Glucuronidase/administração & dosagem , Fatores Imunológicos/administração & dosagem , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Liases/administração & dosagem , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Baço/efeitos dos fármacos , Baço/imunologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Timo/efeitos dos fármacos , Timo/imunologia , Carga Tumoral/efeitos dos fármacos
16.
Mol Genet Metab ; 107(1-2): 161-72, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22902520

RESUMO

Mucopolysaccharidosis (MPS) type VII is a lysosomal storage disease caused by deficiency of the lysosomal enzyme ß-glucuronidase (GUS), leading to accumulation of glycosaminoglycans (GAGs). Enzyme replacement therapy (ERT) effectively clears GAG storage in the viscera. Recent studies showed that a chemically modified form of GUS (PerT-GUS), which escaped clearance by mannose 6-phosphate and mannose receptors and showed prolonged circulation, reduced CNS storage more effectively than native GUS. Clearance of storage in bone has been limited due to the avascularity of the growth plate. To evaluate the effectiveness of long-circulating PerT-GUS in reducing the skeletal pathology, we treated MPS VII mice for 12 weeks beginning at 5 weeks of age with PerT-GUS or native GUS and used micro-CT, radiographs, and quantitative histopathological analysis for assessment of bones. Micro-CT findings showed PerT-GUS treated mice had a significantly lower BMD. Histopathological analysis also showed reduced storage material and a more organized growth plate in PerT-GUS treated mice compared with native GUS treated mice. Long term treatment with PerT-GUS from birth up to 57 weeks also significantly improved bone lesions demonstrated by micro-CT, radiographs and quantitative histopathological assay. In conclusion, long-circulating PerT-GUS provides a significant impact to rescue of bone lesions and CNS involvement.


Assuntos
Doenças Ósseas/etiologia , Doenças Ósseas/terapia , Terapia de Reposição de Enzimas , Glucuronidase/uso terapêutico , Mucopolissacaridose VII/complicações , Mucopolissacaridose VII/terapia , Proteínas Recombinantes/uso terapêutico , Animais , Doenças Ósseas/diagnóstico , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Vértebras Cervicais/diagnóstico por imagem , Vértebras Cervicais/patologia , Glucuronidase/administração & dosagem , Glucuronidase/química , Lâmina de Crescimento/efeitos dos fármacos , Lâmina de Crescimento/patologia , Articulação do Joelho/diagnóstico por imagem , Articulação do Joelho/patologia , Camundongos , Mucopolissacaridose VII/diagnóstico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Tomografia Computadorizada por Raios X
17.
Nat Med ; 1(4): 353-7, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7585066

RESUMO

We have evaluated the feasibility and efficacy of intraperitoneal implants (neo-organs) for protein delivery in large animals. Skin biopsies were taken from four healthy dogs. Primary fibroblast cultures were transduced with a retroviral vector coding for the human beta-glucuronidase. One to six lattices each containing 10(9) skin fibroblasts were implanted into the omentum of the donor animal. Laparotomies performed at regular intervals showed vascularized neo-organs without local inflammation. Human beta-glucuronidase levels equivalent to 0.8 to 3.1% of the endogenous canine activity were detected for up to 340 days on liver biopsy samples. These results indicate that neo-organs can be considered for the long-term delivery of therapeutic proteins or enzymes in humans.


Assuntos
Sistemas de Liberação de Medicamentos , Fibroblastos/transplante , Glucuronidase/metabolismo , Lisossomos/enzimologia , Próteses e Implantes , Animais , Células Cultivadas , Colágeno , DNA/análise , Cães , Eletroforese em Gel de Poliacrilamida , Estudos de Viabilidade , Fibroblastos/enzimologia , Terapia Genética/métodos , Vetores Genéticos , Glucuronidase/administração & dosagem , Glucuronidase/genética , Histocitoquímica , Fígado/enzimologia , Omento , Cavidade Peritoneal , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Retroviridae/genética , Pele/citologia , Baço/enzimologia
18.
Proc Natl Acad Sci U S A ; 105(24): 8375-80, 2008 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-18544647

RESUMO

Glycosaminoglycan storage begins in prenatal life in patients with mucopolysaccharidosis (MPS). In fact, prenatal hydrops is a common manifestation of MPS VII because of beta-glucuronidase (GUS) deficiency. One way to address prenatal storage might be to deliver the missing enzyme across the placenta into the fetal circulation. Maternal IgG is transported across the placenta by the neonatal Fc receptor (FcRn), which recognizes the Fc domain of IgG and mediates transcytosis from maternal to fetal circulation. We hypothesized that we could exploit this process to deliver corrective enzyme to the fetus. To test this hypothesis, the C-terminal fusion protein, GUS-Fc, was compared with native, untagged, recombinant GUS for clearance from the maternal circulation, delivery to the fetus, and reduction of lysosomal storage in offspring of MPS VII mice. We observed that GUS-Fc, infused into pregnant mothers on embryonic days 17 and 18, was transported across the placenta. Similarly infused untagged GUS was not delivered to the fetus. GUS-Fc plasma enzyme activity in newborn MPS VII mice was 1,000 times that seen after administration of untagged GUS and approximately 100 times that of untreated WT newborns. Reduced lysosomal storage in heart valves, liver, and spleen provided evidence that in utero enzyme replacement therapy with GUS-Fc targeted sites of storage in the MPS VII fetus. We hypothesize that this noninvasive approach could deliver the missing lysosomal enzyme to a fetus with any lysosomal storage disease. It might also provide a method for inducing immune tolerance to the missing enzyme or another foreign protein.


Assuntos
Glucuronidase/uso terapêutico , Mucopolissacaridose VII/prevenção & controle , Placenta/metabolismo , Receptores Fc/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Útero , Animais , Feminino , Glucuronidase/administração & dosagem , Glucuronidase/farmacocinética , Glicosaminoglicanos/antagonistas & inibidores , Infusões Parenterais , Lisossomos/metabolismo , Camundongos , Gravidez , Receptores Fc/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacocinética , Distribuição Tecidual
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA