Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Orphanet J Rare Dis ; 19(1): 189, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715031

ABSTRACT

BACKGROUND: Mucopolysaccharidosis VII (MPS VII) is an ultra-rare, autosomal recessive, debilitating, progressive lysosomal storage disease caused by reduced activity of ß-glucuronidase (GUS) enzyme. Vestronidase alfa (recombinant human GUS) intravenous enzyme replacement therapy is an approved treatment for patients with MPS VII. METHODS: This disease monitoring program (DMP) is an ongoing, multicenter observational study collecting standardized real-world data from patients with MPS VII (N ≈ 50 planned) treated with vestronidase alfa or any other management approach. Data are monitored and recorded in compliance with Good Clinical Practice guidelines and planned interim analyses of captured data are performed annually. Here we summarize the safety and efficacy outcomes as of 17 November 2022. RESULTS: As of the data cutoff date, 35 patients were enrolled: 28 in the Treated Group and seven in the Untreated Group. Mean (SD) age at MPS VII diagnosis was 4.5 (4.0) years (range, 0.0 to 12.4 years), and mean (SD) age at DMP enrollment was 13.9 (11.1) years (range, 1.5 to 50.2 years). Ten patients (29%) had a history of nonimmune hydrops fetalis. In the 23 patients who initiated treatment prior to DMP enrollment, substantial changes in mean excretion from initial baseline to DMP enrollment were observed for the three urinary glycosaminoglycans (uGAGs): dermatan sulfate (DS), -84%; chondroitin sulfate (CS), -55%; heparan sulfate (HS), -42%. Also in this group, mean reduction from initial baseline to months 6, 12, and 24 were maintained for uGAG DS (-84%, -87%, -89%, respectively), CS (-70%, -71%, -76%, respectively), and HS (+ 3%, -32%, and - 41%, respectively). All adverse events (AEs) were consistent with the known vestronidase alfa safety profile. No patients discontinued vestronidase alfa. One patient died. CONCLUSIONS: To date, the DMP has collected invaluable MPS VII disease characteristic data. The benefit-risk profile of vestronidase alfa remains unchanged and favorable for its use in the treatment of pediatric and adult patients with MPS VII. Reductions in DS and CS uGAG demonstrate effectiveness of vestronidase alfa to Month 24. Enrollment is ongoing.


Subject(s)
Enzyme Replacement Therapy , Glucuronidase , Mucopolysaccharidosis VII , Recombinant Proteins , Humans , Mucopolysaccharidosis VII/drug therapy , Glucuronidase/therapeutic use , Glucuronidase/metabolism , Male , Child, Preschool , Female , Child , Enzyme Replacement Therapy/methods , Recombinant Proteins/therapeutic use , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Infant , Longitudinal Studies , Adolescent
2.
Prenat Diagn ; 43(12): 1567-1569, 2023 11.
Article in English | MEDLINE | ID: mdl-37964423

ABSTRACT

Duo exome testing was performed on a fetus conceived via in vitro fertilization with an egg donor. The fetus presented with non-immune hydrops fetalis (NIHF) at 20 + 0 weeks gestation. Two variants were detected in the GUSB gene. Biallelic pathogenic variants cause mucopolysaccharidosis type VII (MPS-VII), which can present with NIHF prenatally. At the time of analysis and initial report, one variant was classified as likely pathogenic and the other as of uncertain clinical significance. Biochemical testing of the amniotic fluid supernatant showed elevated glycosaminoglycans and low ß-glucuronidase activity consistent with the diagnosis of MPS-VII. This evidence allowed the upgrade of the pathogenicity for both variants, confirming the diagnosis of MPS-VII. The infant was born at 36 + 5 weeks and enzyme replacement therapy (ERT) using vestronidase was initiated at 20 days with planning for hematopoietic stem cell transplant ongoing. The ERT therapy has been well tolerated, with decreasing quantitative urine glycosaminoglycans. Long-term follow up is required to determine whether treatment has been successful. This case demonstrates the utility of alternative testing methods to clarify the pathogenicity of variants and the clinical utility of obtaining a diagnosis antenatally in facilitating treatment in the neonatal period, and specifically highlights MPS-VII as a treatable cause of NIHF.


Subject(s)
Mucopolysaccharidosis VII , Infant, Newborn , Pregnancy , Female , Humans , Mucopolysaccharidosis VII/diagnosis , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/therapy , Glucuronidase/genetics , Glucuronidase/therapeutic use , Hydrops Fetalis/diagnosis , Hydrops Fetalis/genetics , Hydrops Fetalis/therapy , Prenatal Diagnosis , Amniotic Fluid , Glycosaminoglycans
3.
Bioorg Med Chem ; 90: 117335, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37257254

ABSTRACT

Heparanase-1 (HPSE) is a promising yet challenging therapeutic target. It is the only known enzyme that is responsible for cleavage of heparan sulfate (HS) side chains from heparan sulfate proteoglycans (HSPGs), and is the key enzyme involved in the remodeling and degradation of the extracellular matrix (ECM). Overexpression of HPSE is found in various types of diseases, including cancers, inflammations, diabetes, and viral infections. Inhibiting HPSE can restore ECM functions and integrity, making the development of HPSE inhibitors a highly sought-after topic. So far, all HPSE inhibitors that have entered clinical trials belong to the category of HS mimetics, and no small-molecule or drug-like HPSE inhibitors have made similar progress. None of the HS mimetics have been approved as drugs, with some clinical trials discontinued due to poor bioavailability, side effects, and unfavorable pharmacokinetics characteristics. Small-molecule HPSE inhibitors are, therefore, particularly appealing due to their drug-like characteristics. Advances in the chemical spaces and drug design technologies, including the increasing use of in vitro and in silico screening methods, have provided new opportunities in drug discovery. This article aims to review the discovery and development of small-molecule HPSE inhibitors via screening strategies to shed light on the future endeavors in the development of novel HPSE inhibitors.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Heparan Sulfate Proteoglycans/metabolism , Heparan Sulfate Proteoglycans/therapeutic use , Heparitin Sulfate/metabolism , Heparitin Sulfate/therapeutic use , Glucuronidase/metabolism , Glucuronidase/therapeutic use
4.
Drug Discov Today ; 28(2): 103466, 2023 02.
Article in English | MEDLINE | ID: mdl-36509391

ABSTRACT

Worldwide, around 850 million people are diagnosed with kidney disease but the available treatment options are still limited. Preclinical studies propose a plethora of druggable targets that can attenuate kidney disease and could qualify as novel therapeutic strategies, although most of these targets still await clinical testing. Here, we review some promising candidate targets for chronic kidney disease: intermedin, periostin, sirtuin, the cannabinoid receptor, Klotho, and uromodulin. For acute kidney injury, we discuss Apelin, Elabela, growth differentiation factor-15, Fyn kinase, and Klotho. Target selection for further clinical development should consider redundancies with the standard of care, potential synergistic effects with existing treatments, as well as the potential of additional effects on the cardiovascular system as a common comorbidity in patients with kidney disease.


Subject(s)
Acute Kidney Injury , Renal Insufficiency, Chronic , Humans , Glucuronidase/metabolism , Glucuronidase/therapeutic use , Kidney/metabolism , Renal Insufficiency, Chronic/drug therapy
6.
Int J Pharm ; 606: 120931, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34310961

ABSTRACT

Cancer is the second leading cause of death in the world. Some of the usual cancer treatments include surgery, chemotherapy, and radiotherapy. However, due to low efficacy and side effects of these treatments, novel targeted therapeutic methods are needed. One of the common drawbacks of cancer chemotherapy is off-target toxicity. In order to overcome this problem, many investigations have been conducted. One of the new targeted therapy methods known as bacterial directed enzyme-prodrug therapy (BDEPT) employs bacteria as enzyme carriers to convert a pro-drug to a drug specifically within the tumor site. In the present study, we used Escherichia coli DH5α carrying luxCDABE gene cluster and overexpressing ß-glucuronidase for luminescent emission and enzyme expression, respectively. Enzyme expression can lead to the conversion of glycyrrhizic acid as a prodrug to glycyrrhetinic acid, a potent anti-cancer agent. DH5α-lux/ßG was characterized and its stability was also evaluated. Bacteria colonization in the tumor site was measured by tissue homogenate preparation and colony counting method. Histopathological studies on the liver, spleen, and tumor were also conducted. According to the results, co-treatment of 4T1, a highly metastatic mouse breast cancer cell line, with GL and DH5α-lux/ßG could significantly decrease the IC50 values. Moreover, increased number of bacteria could lead to a dramatic drop in IC50 value. Specific colonization of DH5α-lux/ßG was observed in the tumor site compared with other tissues (p< 0.0001). Moreover, the biocompatibility evaluation proved that DH5α-lux/ßG had no adverse effects on normal tissues. Furthermore, concurrent usage of GL and bacteria in the treatment of induced 4T1 tumors in BALB/c mice significantly delayed tumor growth (p<0.001) during 16 days of investigation. Based on these findings, BDEPT might be useful for targeted breast cancer therapy, although further investigations are required to confirm this.


Subject(s)
Breast Neoplasms/drug therapy , Glucuronidase/therapeutic use , Prodrugs , Animals , Bacteria , Cell Line, Tumor , Mice , Mice, Inbred BALB C
7.
Int J Mol Sci ; 22(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478014

ABSTRACT

Klotho was initially introduced as an antiaging molecule. Klotho deficiency significantly reduces lifespan, and its overexpression extends it and protects against various pathological phenotypes, especially renal disease. It was shown to regulate phosphate and calcium metabolism, protect against oxidative stress, downregulate apoptosis, and have antiinflammatory and antifibrotic properties. The course of diabetes mellitus and diabetic nephropathy resembles premature cellular senescence and causes the activation of various proinflammatory and profibrotic processes. Klotho was shown to exert many beneficial effects in these disorders. The expression of Klotho protein is downregulated in early stages of inflammation and diabetic nephropathy by proinflammatory factors. Therefore, its therapeutic effects are diminished in this disorder. Significantly lower urine levels of Klotho may serve as an early biomarker of renal involvement in diabetes mellitus. Recombinant Klotho administration and Klotho overexpression may have immunotherapeutic potential for the treatment of both diabetes and diabetic nephropathy. Therefore, the current manuscript aims to characterize immunopathologies occurring in diabetes and diabetic nephropathy, and tries to match them with antiinflammatory actions of Klotho. It also gives reasons for Klotho to be used in diagnostics and immunotherapy of these disorders.


Subject(s)
Diabetic Nephropathies/therapy , Glucuronidase/physiology , Inflammation/prevention & control , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cellular Senescence/genetics , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/genetics , Glucuronidase/pharmacology , Glucuronidase/therapeutic use , Humans , Inflammation/diagnosis , Inflammation/etiology , Inflammation/therapy , Klotho Proteins , Protective Agents/pharmacology , Protective Agents/therapeutic use
8.
Biochem Biophys Res Commun ; 534: 1040-1046, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33121684

ABSTRACT

Diabetic nephropathy (DN) is associated with renal mitochondrial injury and decreased renal klotho expression. Klotho is known as an aging suppressor, and mitochondrial dysfunction is the hallmark of aging. Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) is a master regulator of mitochondrial biogenesis, and adenosine monophosphate-activated protein kinase (AMPK) is known as a guardian of mitochondria. Here, we report that recombinant soluble klotho protein (rKL) protects against DN in db/db mice via PGC1α-AMPK-mediated mitochondrial recovery in the kidney. We injected rKL into db/db and db/m mice for 8 weeks and collected the serum and kidney tissue. We treated murine renal tubular cells with rKL in vitro, with and without exposure to 30 mM high glucose (HG). rKL treatment ameliorated major disorders from diabetes, such as obesity, hyperglycemia, and intrarenal reactive oxygen species (ROS) generation, in db/db mice. rKL also diminished albuminuria, recovered renal proximal tubular mitochondria, increased renal p-AMPK and PGC1α, and down-regulated mTOR/TGF-ß in db/db mice. In S1 mouse proximal tubular cells, rKL treatment ameliorated HG-mediated cellular and mitochondrial damage and enhanced oxidative phosphorylation, with an increase in PGC1α-AMPK-induced mitochondrial recovery. Our data suggest that klotho exerts a mitochondrial protective effect in diabetic kidney disease by inducing AMPK-PGC1α expression.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetic Nephropathies/drug therapy , Glucuronidase/therapeutic use , Mitochondria/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Klotho Proteins , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress/drug effects , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects
9.
J Am Heart Assoc ; 9(7): e016041, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32212912

ABSTRACT

Chronic kidney disease is highly prevalent, affecting 10% to 15% of the adult population worldwide and is associated with increased cardiovascular morbidity and mortality. As chronic kidney disease worsens, a unique cardiovascular phenotype develops characterized by heart muscle disease, increased arterial stiffness, atherosclerosis, and hypertension. Cardiovascular risk is multifaceted, but most cardiovascular deaths in patients with advanced chronic kidney disease are caused by heart failure and sudden cardiac death. While the exact drivers of these deaths are unknown, they are believed to be caused by uremic cardiomyopathy: a specific pattern of myocardial hypertrophy, fibrosis, with both diastolic and systolic dysfunction. Although the pathogenesis of uremic cardiomyopathy is likely to be multifactorial, accumulating evidence suggests increased production of fibroblast growth factor-23 and αKlotho deficiency as potential major drivers of cardiac remodeling in patients with uremic cardiomyopathy. In this article we review the increasing understanding of the physiology and clinical aspects of uremic cardiomyopathy and the rapidly increasing knowledge of the biology of both fibroblast growth factor-23 and αKlotho. Finally, we discuss how dissection of these pathological processes is aiding the development of therapeutic options, including small molecules and antibodies, directly aimed at improving the cardiovascular outcomes of patients with chronic kidney disease and end-stage renal disease.


Subject(s)
Cardio-Renal Syndrome/drug therapy , Cardiomyopathies/drug therapy , Fibroblast Growth Factors/antagonists & inhibitors , Glucuronidase/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Uremia/drug therapy , Animals , Cardio-Renal Syndrome/blood , Cardio-Renal Syndrome/mortality , Cardio-Renal Syndrome/physiopathology , Cardiomyopathies/blood , Cardiomyopathies/mortality , Cardiomyopathies/physiopathology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Glucuronidase/blood , Humans , Klotho Proteins , Molecular Targeted Therapy , Prognosis , Recombinant Proteins/therapeutic use , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/physiopathology , Risk Assessment , Risk Factors , Uremia/blood , Uremia/mortality , Uremia/physiopathology
10.
Biochem Biophys Res Commun ; 525(3): 693-698, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32139120

ABSTRACT

The levels of the anti-aging protein α-Klotho, in its soluble form (s-Klotho), are depressed in the circulation of patients with type 1 diabetes (T1D) or type 2 diabetes (T2D). Gene transfer experiments have suggested a protective role for ß-cell specific expression of α-Klotho in murine models of T1D and T1D, but these approaches are not easily translatable to clinical therapy. It is unknown whether systemic s-Klotho protein treatment ameliorates disease in T1D, which is characterized by autoimmune destruction of ß cells. We previously reported from in vitro experiments with ß cells that s-Klotho increases insulin secretion, reduces cells death and promotes ß-cell replication. Here, we investigated s-Klotho protein therapy in NOD mice, which have autoimmune T1D. We observed that diabetic NOD mice have significantly lower plasma levels of s-Klotho, compared to their non-diabetic counterparts. To examine in vivo effects of Klotho, we treated NOD mice with s-Klotho protein, or with a Klotho blocking antibody. Systemic treatment with s-Klotho ameliorated diabetes; notably increasing ß-cell replication and total ß-cell mass. Klotho expression was increased locally in the islets. s-Klotho also markedly reduced immune-cell infiltration of islets (insulitis). In contrast, administration of the Klotho antibody was detrimental, and aggravated the loss of ß-cell mass. Thus, s-Klotho has protective effects in this model of T1D, and this appears to depend on a combination of increased ß-cell replication and reduced insulitis. These findings suggest that s-Klotho might be effective as a new therapeutic agent for T1D.


Subject(s)
Glucuronidase/therapeutic use , Insulin-Secreting Cells/pathology , Animals , Cell Proliferation , Female , Glucuronidase/blood , Humans , Klotho Proteins , Mice, Inbred NOD , Organ Size
11.
Mol Genet Metab ; 129(3): 219-227, 2020 03.
Article in English | MEDLINE | ID: mdl-32063397

ABSTRACT

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.


Subject(s)
Enzyme Replacement Therapy , Glucuronidase/therapeutic use , Glycosaminoglycans/urine , Mucopolysaccharidosis VII/drug therapy , Adolescent , Adult , Antibodies, Neutralizing , Blood-Brain Barrier/drug effects , Child , Cross-Over Studies , Female , Glucuronidase/administration & dosage , Glucuronidase/adverse effects , Glucuronidase/immunology , Humans , Male , Mucopolysaccharidosis VII/immunology , Mucopolysaccharidosis VII/physiopathology , Rare Diseases/therapy , Treatment Outcome
12.
Int J Pharm ; 579: 119159, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32081798

ABSTRACT

Bacteria-directed enzyme prodrug therapy (BDEPT), is an emerging alternative directed and tumor-specific approach. The basis of this method is the conversion of a non-toxic prodrug by a bacterial enzyme to a toxic drug within the tumor-microenvironment (TME). In the present study, the therapeutic efficacy of BDEPT was investigated based on the ability of Escherichia coli DH5α-lux/ßG in activation of glycyrrhizic acid (GL), a natural and non-toxic compound purified from licorice, to glycyrrhetinic acid (GA) only in TME. To do so, the anti-bacterial effects of GL on bacteria and the cytotoxic effects of the produced GA on survival rate of CT26 mouse colon carcinoma cells were evaluated. The IC50 values of the produced GA and cisplatin were determined as 210 µM and 100 µM, respectively. Comparing these values to GL treatment (1305 µM) indicates that bacteria could have efficiently activated GL to GA to inhibit the growth of tumor cells. Afterward, the anti-cancer effects of bacteria used in combination with GL was investigated in a mouse model of colon carcinoma. Results were indicative of targeted homing and even proliferation of luminescent bacteria in TME. Moreover, combined treatment greatly inhibited tumor growth. Histopathological analysis of dissected tissues also demonstrated increased apoptosis rate in tumor cells after combined treatment and interestingly, showed no obvious damage to the spleen and liver of treated mice. Accordingly, this BDEPT approach could be considered as an effective alternative tumor-specific therapy utilizing prodrug-activating enzymes expressing from tumor-targeting bacteria to allow the development of new tumor-specific pharmacotherapy protocols.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colonic Neoplasms/drug therapy , Escherichia coli/enzymology , Genetic Engineering/methods , Glucuronidase/biosynthesis , Glucuronidase/therapeutic use , Glycyrrhizic Acid/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Drug Delivery Systems/methods , Escherichia coli/genetics , Male , Mice , Prodrugs/therapeutic use , Tumor Microenvironment/drug effects
14.
Am J Physiol Renal Physiol ; 318(3): F557-F564, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31928223

ABSTRACT

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.


Subject(s)
Blood Pressure/drug effects , Glucuronidase/therapeutic use , Polycystic Kidney Diseases/drug therapy , Polycystic Kidney Diseases/genetics , Animals , Cells, Cultured , Female , Glucuronidase/administration & dosage , Injections, Subcutaneous , Kidney/physiology , Klotho Proteins , Mice , Myofibroblasts/drug effects , Polycystic Kidney Diseases/physiopathology , Recombinant Proteins
16.
Int J Mol Sci ; 20(21)2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31661765

ABSTRACT

We report the case of a boy who was diagnosed with mucopolysaccharidosis (MPS) VII at two weeks of age. He harbored three missense ß-glucuronidase (GUSB) variations in exon 3: two novel, c.422A>C and c.424C>T, inherited from his mother, and the rather common c.526C>T, inherited from his father. Expression of these variations in transfected HEK293T cells demonstrated that the double mutation c.422A>C;424C>T reduces ß-glucuronidase enzyme activity. Enzyme replacement therapy (ERT), using UX003 (vestronidase alfa), was started at four months of age, followed by a hematopoietic stem cell allograft transplantation (HSCT) at 13 months of age. ERT was well tolerated and attenuated visceromegaly and skin infiltration. After a severe skin and gut graft-versus-host disease, ERT was stopped six months after HSCT. The last follow-up examination (at the age of four years) revealed a normal psychomotor development, stabilized growth curve, no hepatosplenomegaly, and no other organ involvement. Intriguingly, enzyme activity had normalized in leukocytes but remained low in plasma. This case report illustrates: (i) The need for an early diagnosis of MPS, and (ii) the possible benefit of a very early enzymatic and/or cellular therapy in this rare form of lysosomal storage disease.


Subject(s)
Enzyme Replacement Therapy , Glucuronidase/genetics , Hematopoietic Stem Cell Transplantation , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/therapy , Combined Modality Therapy , Glucuronidase/blood , Glucuronidase/therapeutic use , Glucuronidase/urine , HEK293 Cells , Hematopoietic Stem Cell Transplantation/adverse effects , Hepatomegaly/drug therapy , Humans , Infant, Newborn , Leukocytes/enzymology , Leukocytes/metabolism , Male , Mucopolysaccharidosis VII/blood , Mucopolysaccharidosis VII/diagnosis , Mutation , Splenomegaly/drug therapy
17.
Life Sci ; 234: 116773, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31422095

ABSTRACT

AIMS: NLRP3 inflammasome activation is essential for the development and prognosis of diabetic cardiomyopathy (DCM). The anti-aging protein Klotho is suggested to modulate tissue inflammatory responses. The aim of the present study was to examine the protective effects of Klotho on DCM. MAIN METHODS: A streptozotocin-induced diabetes mouse model was established to assess the effects of Klotho in vivo, which was administered for 12 weeks. The characteristics of type 1 DCM were evaluated by general status, echocardiography, and histopathology. The expression of associated factors was determined by RT-qPCR and western blotting. Parallel experiments to determine the molecular mechanism through which Klotho prevents DCM were performed using H9C2 cells exposed to high glucose (35 mM). KEY FINDINGS: Diabetes-induced increases in serum creatine kinase-muscle/brain and lactate dehydrogenase levels, cardiac fibrosis, cardiomyocyte apoptosis, and cardiac dysfunction were ameliorated by Klotho. Additionally, Klotho suppressed TXNIP expression, NLRP3 inflammasome activation, and expression of the inflammatory cytokines tumor necrosis factor ɑ, interleukin-1ß, and interleukin-18 in vivo. In high glucose-cultured cardiomyocytes, Klotho and N-acetylcysteine significantly downregulated intracellular reactive oxygen species generation and TXNIP/NLRP3 inflammasome activation. Pretreatment of H9C2 cells with NLRP3 siRNA or Klotho prevented high glucose-induced inflammation and apoptosis in H9C2 cells. SIGNIFICANCE: Our results demonstrate that the protective effect of Klotho on diabetes-induced cardiac injury is associated with inhibition of the NLRP3 inflammasome pathway, suggesting its therapeutic potential for DCM.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Diabetic Cardiomyopathies/immunology , Glucuronidase/immunology , Inflammasomes/immunology , Inflammation/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/therapeutic use , Cardiotonic Agents/immunology , Cardiotonic Agents/therapeutic use , Cell Line , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Glucuronidase/therapeutic use , Inflammation/drug therapy , Inflammation/etiology , Inflammation/pathology , Klotho Proteins , Male , Mice, Inbred C57BL , Rats , Reactive Oxygen Species/immunology
18.
Anal Cell Pathol (Amst) ; 2019: 5853426, 2019.
Article in English | MEDLINE | ID: mdl-31309036

ABSTRACT

BACKGROUND AND AIMS: Klotho is an aging-suppressor gene mainly expressed in the renal tubules. The klotho gene encodes the α-klotho protein, which has many functions. Previous studies have found that α-klotho protein has a cardiorenal protective function. α-Klotho deficiency renders the kidney more susceptible to injury and results in cardiovascular calcification and left ventricular hypertrophy in chronic kidney disease. However, the role of α-klotho in acute heart injury and acute kidney injury with sepsis remains unknown. This study aimed to investigate the effects and mechanisms of α-klotho in septic cardiorenal injury. METHODS: Male 8-week-old C57BL/6 mice were randomly assigned to the control group, lipopolysaccharide (LPS; 10 mg/kg) group, LPS (10 mg/kg)+α-klotho (0.01 mg/kg) group, and LPS (10 mg/kg)+α-klotho (0.02 mg/kg) group. Recombinant α-klotho was intraperitoneally injected an hour before LPS injection. Mice were euthanized at 24 h after LPS injection. The serum troponin, brain natriuretic peptide (BNP), neutrophil gelatinase-associated lipocalin (NGAL), and creatinine levels were measured in all groups at 24 h. Biomarkers of mice heart apoptosis, inflammation, oxidative stress, and endoplasmic reticulum stress, such as caspase-3, interleukin 1 (IL-1), reactive oxygen species (ROS), and glucose-regulated protein 78 (GRP78), were also measured. RESULTS: α-Klotho was mainly expressed in mice kidneys and was undetectable in the control mice hearts. α-Klotho substantially decreased after LPS injection. In the LPS group, the serum troponin levels significantly increased as early as 6 h (p < 0.05) after LPS injection, while the BNP, NGAL, and creatinine levels significantly increased at 24 h (p < 0.05). Pretreatment with α-klotho significantly ameliorated acute cardiorenal injury. In the LPS+α-klotho (0.01 mg/kg) group, the levels of apoptosis, inflammation, and oxidative stress were decreased, while the level of endoplasmic reticulum stress was elevated. CONCLUSIONS: α-Klotho significantly alleviates acute cardiorenal injury in LPS-induced septic cardiorenal injury due to the inhibition of apoptosis, inflammation, and oxidation, as well as the regulation of endoplasmic reticulum stress levels.


Subject(s)
Cardio-Renal Syndrome/drug therapy , Glucuronidase/therapeutic use , Kidney/drug effects , Recombinant Proteins/therapeutic use , Animals , Cardio-Renal Syndrome/metabolism , Cardio-Renal Syndrome/pathology , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Glucuronidase/pharmacology , Kidney/metabolism , Kidney/pathology , Klotho Proteins , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology
19.
Biochem Biophys Res Commun ; 513(2): 426-433, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30967260

ABSTRACT

OBJECTIVE: Inflammatory bowel diseases (IBD) including ulcerative colitis and Crohn's disease are devastating diseases of the gut. At present, all the treatments are mainly targeting symptoms like inflammation. The disease remains regarded as incurable, largely due to lacking of knowledge on its etiology. Our previous studies suggested that impaired inactivation of digestive proteases by deconjugated bilirubin in experimental colitis, thus bacterial ß-glucuronidase for catalyzing the reaction, may have played critical role in the pathogenesis of IBD. METHODS: We first analyzed ß-glucuronidase activity in gut tissue and feces of mice by a colitis model. Then the effect of ß-glucuronidase on experimental colitis was investigated in detail by administration of ß-glucuronidase (from E. coli) and fecal material transplantation to mice with 3% DSS in drinking water for 7 days. RESULTS: Mice with colitis showed unchanged activity of ß-glucuronidase in colon tissue but decreased activity in feces. Treatment with bacterial ß-glucuronidase at 100 U or above alleviated DSS-induced colitis as demonstrated by the less body weight loss, less disease activity score, increased expression of tight junction proteins and decreased gut permeability, decreases in MPO, TNF-α, IL-1ß, TLR-4 and MyD88, and increase in IL-10 and IκBα in gut, restored fecal ß-glucuronidase and gut microbiota along with decreases in fecal digestive proteases. Transplantation of fecal material from control to colitis mice showed similar effects as treatment with ß-glucuronidase. CONCLUSIONS: Bacterial ß-glucuronidase showed strong inhibition on colitis along with the reduction in fecal digestive proteases, which may be a crucial diagnostic and therapeutic target for IBD.


Subject(s)
Colitis/drug therapy , Escherichia coli/enzymology , Glucuronidase/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Animals , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate , Disease Models, Animal , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/pathology , Male , Mice , Mice, Inbred C57BL
20.
Drugs Today (Barc) ; 55(1): 17-23, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30740609

ABSTRACT

Lumbar disc herniation (LDH) is generally treated with a conservative therapy, and surgery is the only therapeutic option currently available for patients unresponsive to the conservative therapy. In the 1980s, chemonucleolysis with chymopapain, a protease, was widely used as the intermediate treatment between conservative therapy and surgical therapy in the Western countries. However, since chymopapain was withdrawn from the market in 2002 for non-scientific commercial reasons, chemonucleolysis has not been a therapeutic option for LDH. Condoliase (chondroitin sulfate ABC endolyase), a glycosaminoglycan-degrading enzyme, was approved by the drug regulatory authority in Japan as a newer intradiscal therapy for LDH after clinical studies conducted in Japan demonstrated efficacy and safety for patients with LDH. This review will focus on the preclinical pharmacology, pharmacokinetics, efficacy and safety of condoliase as a new option for treatment of LDH.


Subject(s)
Glucuronidase/therapeutic use , Intervertebral Disc Displacement/therapy , Lyases/therapeutic use , Chymopapain , Glucuronidase/pharmacokinetics , Humans , Intervertebral Disc Chemolysis , Japan , Lyases/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...