Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Mol Genet Metab ; 142(1): 108454, 2024 May.
Article in English | MEDLINE | ID: mdl-38603816

ABSTRACT

BACKGROUND: Cystine-depleting therapy in nephropathic cystinosis is currently monitored via the white blood cell cystine assay, although its application and usefulness are limited by practical and technical issues. Therefore, alternative biomarkers that are widely available, more economical and less technically demanding, while reliably reflecting long-term adherence to cysteamine treatment, are desirable. Recently, we proposed chitotriosidase enzyme activity as a potential novel biomarker for the therapeutic monitoring of cysteamine treatment in cystinosis. In this study, we aimed to validate our previous findings and to confirm the value of chitotriosidase in the management of cystinosis therapy. MATERIALS & METHODS: A retrospective study was conducted on 12 patients treated at the National Institutes of Health Clinical Center and followed up for at least 2 years. Plasma chitotriosidase enzyme activity was correlated with corresponding clinical and biochemical data. RESULTS: Plasma chitotriosidase enzyme activity significantly correlated with WBC cystine levels, cysteamine total daily dosage and a Composite compliance score. Moreover, plasma chitotriosidase was a significant independent predictor for WBC cystine levels, and cut-off values were established in both non-kidney transplanted and kidney transplanted cystinosis patients to distinguish patients with a good versus poor compliance with cysteamine treatment. Our observations are consistent with those of our previous study and validate our findings. CONCLUSIONS: Chitotriosidase enzyme activity is a valid potential alternative biomarker for monitoring cysteamine treatment in nephropathic cystinosis patients. SYNOPSIS: Chitotriosidase enzyme activity is a valid potential alternative biomarker for monitoring cysteamine treatment in nephropathic cystinosis patients.


Subject(s)
Cysteamine , Cystine , Cystinosis , Hexosaminidases , Humans , Cysteamine/therapeutic use , Male , Female , Cystinosis/drug therapy , Cystinosis/blood , Retrospective Studies , Hexosaminidases/blood , Adolescent , Cystine/blood , Child , Adult , Biomarkers/blood , Young Adult , Drug Monitoring/methods , Cystine Depleting Agents/therapeutic use , Child, Preschool , Kidney Transplantation
2.
Pediatr Nephrol ; 39(8): 2429-2433, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38462547

ABSTRACT

BACKGROUND: The objective of this report is to identify and characterize cases of fibrosing colonopathy, a rare and underrecognized adverse event, associated with cysteamine delayed-release (DR) in patients with nephropathic cystinosis. METHODS: We searched the U.S. Food and Drug Administration Adverse Event Reporting System (FAERS) and the medical literature for postmarketing reports of fibrosing colonopathy associated with cysteamine through August 2, 2023. RESULTS: We identified four cases of fibrosing colonopathy reported with the use of cysteamine DR. The time to onset ranged from 12 to 31 months. In one case, the patient required surgery to have a resection of a section of the strictured colon and a diverting ileostomy. Fibrosing colonopathy was diagnosed by histopathology in two of the cases. CONCLUSIONS: Our case series identified the risk of fibrosing colonopathy in patients taking cysteamine DR and prompted regulatory action by the FDA. As outlined in changes to the U.S. prescribing information for cysteamine DR, healthcare professionals should be aware of the potential risk of fibrosing colonopathy with cysteamine DR, especially as symptoms can be non-specific leading to misdiagnosis or delayed diagnosis. If the diagnosis of fibrosing colonopathy is confirmed, consideration should be given to permanently discontinuing cysteamine DR and switching to cysteamine immediate-release treatment.


Subject(s)
Cysteamine , Cystinosis , Delayed-Action Preparations , Humans , Cysteamine/adverse effects , Cysteamine/administration & dosage , Cystinosis/complications , Cystinosis/diagnosis , Cystinosis/drug therapy , Delayed-Action Preparations/adverse effects , Female , Male , Child , Colonic Diseases/chemically induced , Colonic Diseases/diagnosis , Colonic Diseases/pathology , Colonic Diseases/etiology , Adolescent , Cystine Depleting Agents/administration & dosage , Cystine Depleting Agents/adverse effects , United States , Fibrosis , Colon/pathology , Colon/drug effects , Colon/diagnostic imaging , Capsules , Child, Preschool , Adverse Drug Reaction Reporting Systems
3.
Pediatr Nephrol ; 39(8): 2283-2292, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38127152

ABSTRACT

Cystinosis is a rare autosomal recessive disease with an incidence 1 per 100,000-200,000 live births. It is caused by pathogenic variants of the cystinosin (CTNS) gene that lead to impaired cystine transport from lysosomes to cystosol, resulting in cystine accumulation in lysosomes and subsequent cellular dysfunction. The initial manifestation, cystine accumulation in proximal tubular cells (PTCs), causes renal Fanconi syndrome, which presents with proximal renal tubular acidosis and generalized dysfunction of the proximal tubule, including the presence of polyuria, glycosuria, phosphaturia, aminoaciduria, tubular proteinuria, growth retardation, and rickets. Eventually, glomerular involvement, glomerular proteinuria, focal segmental glomerulosclerosis (FSGS), and progression to kidney failure occur. Although the kidneys are the first organs affected, and play a key role in morbidity and mortality, extrarenal multiorgan involvement can occur in patients with cystinosis, which is seen not only in adults but in early ages in untreated patients, patients with insufficient treatment, and in those that don't comply with treatment. The treatment of cystinosis consists of supportive treatment for Fanconi syndrome, and specific lifelong cystine-depleting therapy using oral cysteamine. There is strong evidence that as early as possible, initiation and ongoing appropriate therapy with cysteamine are essential for delaying the progression to kidney failure, end-organ damage, and extrarenal involvement. The present review aimed to evaluate the extra renal complications of cystinosis.


Subject(s)
Cystinosis , Fanconi Syndrome , Humans , Cystinosis/complications , Cystinosis/genetics , Fanconi Syndrome/etiology , Fanconi Syndrome/complications , Cysteamine/therapeutic use , Cystine Depleting Agents/therapeutic use , Amino Acid Transport Systems, Neutral/genetics
4.
J Inherit Metab Dis ; 44(6): 1393-1408, 2021 11.
Article in English | MEDLINE | ID: mdl-34494673

ABSTRACT

Cystinosis is an inherited metabolic disorder caused by autosomal recessive mutations in the CTNS gene leading to lysosomal cystine accumulation. The disease primarily affects the kidneys followed by extra-renal organ involvement later in life. Azoospermia is one of the unclarified complications which are not improved by cysteamine, which is the only available disease-modifying treatment. We aimed at unraveling the origin of azoospermia in cysteamine-treated cystinosis by confirming or excluding an obstructive factor, and investigating the effect of cysteamine on fertility in the Ctns-/- mouse model compared with wild type. Azoospermia was present in the vast majority of infantile type cystinosis patients. While spermatogenesis was intact, an enlarged caput epididymis and reduced levels of seminal markers for obstruction neutral α-glucosidase (NAG) and extracellular matrix protein 1 (ECM1) pointed towards an epididymal obstruction. Histopathological examination in human and mouse testis revealed a disturbed blood-testis barrier characterized by an altered zonula occludens-1 (ZO-1) protein expression. Animal studies ruled out a negative effect of cysteamine on fertility, but showed that cystine accumulation in the testis is irresponsive to regular cysteamine treatment. We conclude that the azoospermia in infantile cystinosis is due to an obstruction related to epididymal dysfunction, irrespective of the severity of an evolving primary hypogonadism. Regular cysteamine treatment does not affect fertility but has subtherapeutic effects on cystine accumulation in testis.


Subject(s)
Azoospermia/pathology , Blood-Testis Barrier/metabolism , Cysteamine/therapeutic use , Cystinosis/drug therapy , Testis/pathology , Adult , Animals , Azoospermia/complications , Azoospermia/genetics , Cystine Depleting Agents/therapeutic use , Cystinosis/complications , Cystinosis/pathology , Disease Models, Animal , Extracellular Matrix Proteins/metabolism , Humans , Infertility, Male/etiology , Infertility, Male/genetics , Infertility, Male/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neoplasm Proteins/metabolism , Retrospective Studies , Young Adult , Zonula Occludens-1 Protein/metabolism
5.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34428184

ABSTRACT

IgA nephropathy is caused by deposition of circulatory IgA1 in the kidney. Hypogalactosylated IgA1 has the propensity to form poly-IgA aggregates that are prone to deposition. Herein, we purified poly-IgA from the plasma of patients with IgA nephropathy and showed that the complex is susceptible to reducing conditions, suggesting intermolecular disulfide connections between IgA units. We sought to find the cysteine residue(s) that form intermolecular disulfide. Naturally assembled dimeric IgA, also known as secretory IgA, involves a J chain subunit connected with 2 IgA1 molecules via their penultimate cysteine-471 residue on a "tailpiece" segment of IgA heavy chain. It is plausible that, with the absence of J chain, the cysteine residue of mono-IgA1 might aberrantly form a disulfide bond in poly-IgA formation. Mutagenesis confirmed that cysteine-471 is capable of promoting IgA aggregation. These discoveries prompted us to test thiol-based drugs for stabilizing cysteine. Specifically, the cystine-reducing drug cysteamine used for treatment of cystinosis showed a remarkable potency in preventing self-aggregation of IgA. When administrated to rat and mouse models of IgA nephropathy, cysteamine significantly reduced glomerular IgA deposition. Collectively, our results reveal a potentially novel molecular mechanism for aberrant formation of IgA aggregates, to which the repurposed cystinosis drug cysteamine was efficacious in preventing renal IgA deposition.


Subject(s)
Cysteine/metabolism , Glomerulonephritis, IGA/metabolism , Immunoglobulin A/metabolism , Kidney Glomerulus/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Cysteamine/pharmacology , Cysteine/drug effects , Cystine Depleting Agents/pharmacology , Disease Models, Animal , Humans , Immunoglobulin A/drug effects , Immunoglobulin J-Chains/metabolism , Kidney Glomerulus/drug effects , Mice , Rats
6.
Kidney Int ; 100(5): 1112-1123, 2021 11.
Article in English | MEDLINE | ID: mdl-34237326

ABSTRACT

Nephropathic cystinosis is a rare disease secondary to recessive mutations of the CTNS gene encoding the lysosomal cystine transporter cystinosin, causing accumulation of cystine in multiple organs. Over the years, the disease has evolved from being a fatal condition during early childhood into a treatable condition, with patients surviving into adulthood. Data on cystinosis are limited by the rarity of the disease. Here, we have investigated factors associated with kidney and growth outcome in a very large cohort of 453 patients born between 1964 and 2016 and followed in Belgium, Germany, Austria, France, Italy, Spain, The Netherlands, Turkey and United Kingdom. From the 1970s to the 1990s, the median increase in kidney survival was 9.1 years. During these years, cysteamine, a cystine-depleting agent, was introduced for the treatment of cystinosis. Significant risk factors associated with early progression to end-stage kidney disease assessed by Cox proportional multivariable analysis included delayed initiation of cysteamine therapy and higher mean leucocyte cystine levels. No significant effect on kidney function was observed for gender, pathogenic variant of the CTNS gene, and the prescription of indomethacin or renin angiotensin system blockers. Significantly improved linear growth was associated with early use of cysteamine and lower leukocyte cystine levels. Thus, our study provides strong evidence in favor of early diagnosis and optimization of cystine depletion therapy in nephropathic cystinosis.


Subject(s)
Cystinosis , Fanconi Syndrome , Adult , Child, Preschool , Cohort Studies , Cysteamine/therapeutic use , Cystine , Cystine Depleting Agents , Cystinosis/genetics , Humans
7.
Pharmacol Res Perspect ; 9(2): e00739, 2021 04.
Article in English | MEDLINE | ID: mdl-33764642

ABSTRACT

The strict intake regimen of cysteamine bitartrate formulations, associated with side effects, is a concern for the treatment compliance in cystinosis therapy. Therefore, there is a need for a cysteamine formulation with an improved pharmacokinetic profile. This study investigated the pharmacokinetics, safety and tolerability of a new sustained-release cysteamine dosage form, PO-001, in healthy volunteers. This was a randomized, investigator-blinded, three-way cross-over study to compare single doses (600 mg) of PO-001 with Cystagon® (immediate-release) and Procysbi® (delayed-release). Collected blood samples were analyzed for plasma cysteamine concentrations and pharmacokinetic parameters were estimated by noncompartmental analysis. In addition, plasma cysteamine concentrations were analyzed using a population pharmacokinetic approach using NONMEM® . Pharmacokinetics showed clear sustained-release characteristics of PO-001 over time with a lower Cmax and longer Tmax compared to Cystagon® and Procysbi® . All treatment-emergent adverse events were of mild severity, with the exception of two subjects who reported moderate severity gastrointestinal problems including vomiting and diarrhea, which were related to Cystagon® intake. Population PK simulations showed a favourable PK profile based on Cmax and Ctrough concentrations at steady state. In conclusion, a single dose of 600 mg PO-001 was well tolerated with no findings of clinical concern. This new cysteamine bitartrate formulation showed pharmacokinetics of a sustained-release formulation, which may be beneficial for the treatment of cystinosis patients. This study supports advancing this type of sustained-release formulation into a subsequent study to confirm reduced dosing frequency with efficient control of white blood cells (WBCs) cystine levels. Netherlands Trial Registry (NTR) (NL67638.056.18).


Subject(s)
Cysteamine/pharmacokinetics , Cystine Depleting Agents/pharmacokinetics , Cystinosis/drug therapy , Adult , Area Under Curve , Cross-Over Studies , Cysteamine/administration & dosage , Cysteamine/adverse effects , Cystine Depleting Agents/administration & dosage , Cystine Depleting Agents/adverse effects , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/adverse effects , Delayed-Action Preparations/pharmacokinetics , Healthy Volunteers , Humans , Male , Netherlands , Young Adult
8.
Am J Ophthalmol ; 223: 275-285, 2021 03.
Article in English | MEDLINE | ID: mdl-32888903

ABSTRACT

PURPOSE: To evaluate safety and efficacy of topical cysteamine ophthalmic solution for corneal cystinosis. METHODS: Seven databases were searched (PubMed, OVID, EMBASE, Web of Science, Cochrane Central, Google Scholar, and ClinicalTrials.gov) for relevant studies, using appropriate keywords. Comparative observational studies and randomized controlled trials comparing cysteamine with control or other formulations for treatment of corneal or ophthalmic cystinosis were included. Outcome measurements were improvement or response to therapy, change in corneal cystine crystal score (CCCS), in vivo confocal microscopy score (IVCM), cystine crystal depth, contrast sensitivity (CS), photophobia score, and safety. DESIGN: Systematic review and meta-analysis. RESULTS: Seven studies were included. Compared to placebo and control, the cysteamine arm was better in terms of improvements and responses to therapy (2 studies showed a risk ratio [RR] of 16; 95% confidence interval [CI]: 2.30-111.37) and crystal density score (1 study showed a mean difference [MD] of -0.80; 95% CI: -1.56 to -0.04). No significant differences were observed in terms of improvement in CS (1 study showed an RR of 7.00; 95% CI: 0.47-103.27). Compared to cystamine, cysteamine showed benefits in terms of crystal density score (MD -0.94; 95% CI: -1.64 to -0.24). Compared to a newer formulation, the standard formulation (cysteamine [Cystaran]; 0.55% cysteamine hydrochloride + benzalkonium chloride 0.01%) performed better in terms of decreasing CCCS. Another newer, viscous formulation, Cystadrops, performed better than the standard formulation in terms of change in CCCS, IVCM score, corneal crystal depth, and photophobia score; however, local adverse effects and blurring were higher in the group receiving Cystadrops. CONCLUSIONS: Conventional cysteamine (0.1% to 0.3%) performed better than placebo (control) in terms of response to therapy. In terms of decreasing corneal cystine density, cysteamine (0.55%) was better than cystamine (0.55%), and the viscous Cystadrops (0.55%) was better than the standard formulation (0.1%).


Subject(s)
Cysteamine/administration & dosage , Cystinosis/drug therapy , Visual Acuity , Corneal Diseases/drug therapy , Cystine Depleting Agents/administration & dosage , Humans , Ophthalmic Solutions/administration & dosage
9.
Cells ; 11(1)2021 12 24.
Article in English | MEDLINE | ID: mdl-35011614

ABSTRACT

The ongoing pandemic of coronavirus disease-2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), needs better treatment options both at antiviral and anti-inflammatory levels. It has been demonstrated that the aminothiol cysteamine, an already human applied drug, and its disulfide product of oxidation, cystamine, have anti-infective properties targeting viruses, bacteria, and parasites. To determine whether these compounds exert antiviral effects against SARS-CoV-2, we used different in vitro viral infected cell-based assays. Moreover, since cysteamine has also immune-modulatory activity, we investigated its ability to modulate SARS-CoV-2-specific immune response in vitro in blood samples from COVID-19 patients. We found that cysteamine and cystamine decreased SARS-CoV-2-induced cytopathic effects (CPE) in Vero E6 cells. Interestingly, the antiviral action was independent of the treatment time respect to SARS-CoV-2 infection. Moreover, cysteamine and cystamine significantly decreased viral production in Vero E6 and Calu-3 cells. Finally, cysteamine and cystamine have an anti-inflammatory effect, as they significantly decrease the SARS-CoV-2 specific IFN-γ production in vitro in blood samples from COVID-19 patients. Overall, our findings suggest that cysteamine and cystamine exert direct antiviral actions against SARS-CoV-2 and have in vitro immunomodulatory effects, thus providing a rational to test these compounds as a novel therapy for COVID-19.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Cysteamine/pharmacology , Drug Repositioning/methods , Immunomodulating Agents/pharmacology , SARS-CoV-2/drug effects , Aged , Animals , COVID-19/virology , Cell Line, Tumor , Chlorocebus aethiops , Cystamine/pharmacology , Cystine Depleting Agents/pharmacology , Female , Humans , Male , Middle Aged , RNA, Viral/genetics , RNA, Viral/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Vero Cells , Virus Replication/drug effects , Virus Replication/genetics
10.
Br J Ophthalmol ; 105(5): 608-613, 2021 05.
Article in English | MEDLINE | ID: mdl-32593979

ABSTRACT

BACKGROUND/AIMS: Cystinosis is a rare, autosomal recessive disorder causing defective transport of cystine out of lysosomes. Cystadrops (0.55% cysteamine hydrochloride in viscous solution) has been used on a named-patient basis to treat the accumulation of cystine crystals in the cornea in patients with cystinosis. METHODS: Retrospective analysis of the Temporary Authorisation for Use cohort of 130 patients who received Cystadrops between 2013 and 2017 in France. RESULTS: Patients received an average dosage of 3.3 (±0.94) instillations per eye per day. Over the duration of follow-up, of up to 45 months, patients maintained visual acuity scores of 0.0, which approximated normal. Corneal cystine crystal scores tended to decrease over time, stabilising after around 27 months between 1.22 and 1.87. Photophobia decreased within 3 months, stabilising on scores of around 1.5 and 1.7. 47 non-serious adverse reactions were reported, which were generally transient irritation, stinging or blurred vision. Four serious adverse events were reported, including keratitis and corneal ulcer, but these may have been caused by the underlying disease. CONCLUSION: This large safety cohort confirms the efficacy, safety and tolerability of Cystadrops in real-world clinical practice.


Subject(s)
Cornea/metabolism , Cysteamine/administration & dosage , Cystinosis/drug therapy , Visual Acuity , Adolescent , Adult , Aged , Child , Child, Preschool , Cornea/diagnostic imaging , Cystine Depleting Agents/pharmacology , Cystinosis/diagnosis , Female , Follow-Up Studies , Humans , Male , Middle Aged , Ophthalmic Solutions , Retrospective Studies , Time Factors , Young Adult
11.
Curr Gastroenterol Rep ; 22(10): 52, 2020 Aug 19.
Article in English | MEDLINE | ID: mdl-32814993

ABSTRACT

PURPOSE OF REVIEW: Nonalcoholic fatty liver disease (NAFLD) has emerged as the leading cause of chronic liver disease in both adults and children. In this article, we review recent developments in the screening, diagnosis, and treatment of pediatric NAFLD. RECENT FINDINGS: Although alanine aminotransferase (ALT) remains the best screening test for NAFLD in children, and liver biopsy is still required for the diagnosis of nonalcoholic steatohepatitis (NASH), other noninvasive biomarker/imaging studies (MRI-PDFF and VCTE) have emerged as diagnostic methods for pediatric NAFLD. Two large clinical therapeutic trials testing vitamin E, metformin, and cysteamine in pediatric NAFLD yielded mostly inconclusive results. Bariatric surgery has begun to be used in adolescents with severe obesity. An adult phase 2 study using obeticholic acid (OCA) to treat NASH patients with fibrosis showed some positive results. As we continue to await the first FDA-approved therapeutic agent for NASH, lifestyle change remains the main modality of treatment. Newer diagnostic and treatment modalities for pediatric NAFLD continue to be in development under FDA guidance.


Subject(s)
Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/therapy , Pediatric Obesity/therapy , Adolescent , Alanine Transaminase/blood , Antioxidants/therapeutic use , Bariatric Surgery , Child , Child, Preschool , Cysteamine/therapeutic use , Cystine Depleting Agents/therapeutic use , Delayed-Action Preparations/therapeutic use , Diet , Humans , Hypoglycemic Agents/therapeutic use , Infant , Infant, Newborn , Life Style , Liver Cirrhosis/diagnostic imaging , Metformin/therapeutic use , Non-alcoholic Fatty Liver Disease/etiology , Pediatric Obesity/complications , Vitamin E/therapeutic use
12.
Eur J Ophthalmol ; 30(6): NP7-NP10, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31074291

ABSTRACT

PURPOSE: To report a case of intermediate cystinosis with an atypical presentation in which a delayed non-treatment decision has been proved right over a large period of time. METHODS: This is a unique case report of a patient who underwent full ophthalmologic evaluations including anterior chamber optical coherence tomography on a regular basis during a 7-year follow-up period. RESULTS: A 30-year-old woman with photophobia was being studied by the Department of Nephrology with a suspicion of Alport syndrome. Slit-lamp examination showed iridescent deposits throughout the corneal anterior stroma and the inferior tarsal conjunctiva bilaterally. Anterior chamber optical coherence tomography showed stromal hyperreflectivity. CTNS gene was found to be positive for c.416C>T (Ser139Phe) mutation. The patient was offered oral and topical cysteamine which was refused. After a period of 5 years of follow-up, general health status remained stable, corneal disease showed no progression and photophobia complaints diminished. However, the patient was advised to start systemic and topical cysteamine because of the unknown development of the disease. CONCLUSION: In this reported case, a delayed non-treatment decision has been proved right contrary to published evidence of active treatment of photophobia. The decision whether to treat or not to treat corneal involvement of the disease is not straightforward. Besides biomicroscopic evaluations, patients' complaints and expectations should be taken into account.


Subject(s)
Cornea/pathology , Corneal Diseases/etiology , Cysteamine/therapeutic use , Cystinosis/complications , Tomography, Optical Coherence/methods , Visual Acuity , Adult , Corneal Diseases/diagnosis , Corneal Diseases/drug therapy , Cystine Depleting Agents/therapeutic use , Cystinosis/diagnosis , Cystinosis/drug therapy , Female , Follow-Up Studies , Humans , Microscopy, Confocal/methods , Slit Lamp Microscopy , Time Factors
13.
Theriogenology ; 146: 152-161, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-31787466

ABSTRACT

The eicosapentaenoic acid (EPA) is an n-3 polyunsaturated fatty acid (PUFA) present in the lipid composition of bovine oocytes. Little is known about the importance of EPA in bovine oocyte maturation and embryo development in vitro. Although previous work suggest that n-3 PUFAs may inhibit oocyte maturation, the available data are inconsistent. In this study, we evaluated the effect of EPA (1, 10, 100 nM) during in vitro maturation (IVM) of bovine oocytes, alone and in combination with vitamin E (VE) or cysteamine (CYS). EPA treatment in IVM decreased oocyte lipid content and affected lipid droplets pattern (P < 0.05). EPA 100 nM reduced oocytes maturation rate (P < 0.05), without affecting cumulus expansion. At the concentrations tested, EPA did not modify embryo development. However, the addition of antioxidants during IVM reduced the levels of reactive oxygen species in the culture system by increasing intracellular glutathione content (P < 0.05). Besides, the combination of EPA with VE or CYS reduced the percentages of MI oocytes after 24 h of IVM (P < 0.05). EPA reduced oocyte lipid content without any detrimental for embryo development.


Subject(s)
Eicosapentaenoic Acid/pharmacology , Embryonic Development/drug effects , In Vitro Oocyte Maturation Techniques/veterinary , Lipids/chemistry , Oocytes/drug effects , Animals , Antioxidants , Cattle , Cysteamine/administration & dosage , Cysteamine/pharmacology , Cystine Depleting Agents/administration & dosage , Cystine Depleting Agents/pharmacology , Eicosapentaenoic Acid/administration & dosage , Embryo Culture Techniques/veterinary , Oocytes/chemistry , Vitamin E/administration & dosage , Vitamin E/pharmacology
14.
BMC Nephrol ; 20(1): 400, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31672123

ABSTRACT

BACKGROUND: Cystinosis is an autosomal recessive lysosomal storage disorder characterized by accumulation of cystine in lysosomes throughout the body. Cystinosis is caused by mutations in the CTNS gene that encodes the lysosomal cystine carrier protein cystinosin. CTNS mutations result in either complete absence or reduced cystine transporting function of the protein. The diagnosis of nephropathic cystinosis is generally based on measuring leukocyte cystine level, demonstration of corneal cystine crystals by the slit lamp examination and confirmed by genetic analysis of the CTNS gene. CASE PRESENTATION: A boy born to consanguineous Caucasian parents had the characteristic clinical features of the infantile nephropathic cystinosis including renal Fanconi syndrome (polydipsia/polyuria, metabolic acidosis, hypokalemia, hypophosphatemia, low molecular weight proteinuria, glycosuria, cystine crystals in the cornea) and elevated WBC cystine levels. Initially we performed RFLP analysis of the common in the Northern European population 57-kb deletion of proband's DNA, then a direct Sanger sequencing which revealed no mutations in the coding part of the CTNS gene. To confirm the diagnosis we performed RT-PCR analysis of total RNA obtained from patient-derived fibroblasts in combination with cDNA sequencing. This revealed the skipping of exon 4 and exon 5 in the CTNS in our patient. Therefore, we detected a novel 9-kb homozygous deletion in the CTNS gene at genomic DNA level, spanning region from intron 3 to intron 5. In order to identify the inheritance pattern of the deletion we analyzed DNA of proband's mother and father. Both parents were found to be heterozygous carriers of the CTNS mutation. CONCLUSIONS: Analysis of CTNS gene transcript allowed to identify a large homozygous deletion in the patient with infantile nephropathic cystinosis. Mutational detection at RNA level may be an efficient tool to establish the genetic defect in some cystinosis patients.


Subject(s)
Amino Acid Transport Systems, Neutral/genetics , Cystinosis/genetics , Mutation , Child, Preschool , Consanguinity , Cysteamine/therapeutic use , Cystine Depleting Agents/therapeutic use , Cystinosis/drug therapy , Cystinosis/metabolism , DNA Mutational Analysis , Exons/genetics , Fibroblasts/chemistry , Humans , Infant , Introns/genetics , Male , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
15.
J Pediatr Endocrinol Metab ; 32(10): 1187-1191, 2019 Oct 25.
Article in English | MEDLINE | ID: mdl-31600138

ABSTRACT

Background Short stature is a common presentation in paediatric practice. Rickets can lead to poor growth and finding the underlying cause of rickets can, at times, be challenging. Case presentation The child was initially referred due to parental concerns of delayed walking, bowed legs, waddling gait and faltering growth. She was noted to have features of rickets. Bone profile and renal functions were reported to be within the normal range, however, on later review it was noted that adult values for inorganic phosphate had been given for reference ranges. Following a series of investigations, the underlying diagnosis for all her problems was made. Discussion This case demonstrates the complex diagnostic journey of a child whose presentation was not typical of the rare disorder. Unusually, the patient had no symptoms of polyuria or polydipsia and urine osmolality was normal.


Subject(s)
Cysteamine/administration & dosage , Cystinosis/diagnosis , Dwarfism/diagnosis , Rickets/diagnosis , Child, Preschool , Cystine Depleting Agents/administration & dosage , Cystinosis/complications , Cystinosis/drug therapy , Diagnosis, Differential , Dwarfism/complications , Female , Humans , Prognosis , Rickets/complications
16.
Transplant Proc ; 51(2): 545-547, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30879586

ABSTRACT

BACKGROUND: Cystinosis is a rare genetic disorder characterized by the abnormal accumulation of cystine in the lysosomes of various tissues and organs leading to their dysfunction. The most common type is the infantile nephropathic cystinosis which without treatment leads to renal failure and before the introduction of cysteamine was the cause of death before puberty. CASE PRESENTATION: A 27-year-old female patient with infantile cystinosis developed end-stage renal disease at the age of 10. The first kidney transplantation from patient's father was carried out at the age of 12. The recurrent urinary tract infections led to the graft failure after 6 years. Following the removal of right appendages due to the ovarian tumor, the patient underwent the second kidney transplantation from her mother at the age of 19. After the transplantation, the cysteamine treatment was irregular due to limited availability of the medicine. When it became regular in 2017 the patient did not tolerate full doses. Despite elevated blood levels of cystine and the removal of right appendages, the patient naturally became pregnant in August 2017. Except for recurrent urinary tract infections, the renal parameters remained normal throughout the entire pregnancy. However, in the 32nd week of gestation, due to preeclampsia a caesarean section was performed. A healthy daughter was born, 1400/41 and with a 9 point Apgar score. CONCLUSIONS: Due to the possibility of treatment with cysteamine and kidney transplantations, patients with cystinosis live longer and their quality of life improves. These female patients can even naturally become pregnant and give birth to healthy children.


Subject(s)
Cystinosis , Pregnancy Complications , Adult , Cesarean Section , Cysteamine/therapeutic use , Cystine Depleting Agents/therapeutic use , Cystinosis/therapy , Female , Humans , Kidney Transplantation , Pregnancy , Pregnancy Outcome
17.
Curr Eye Res ; 44(5): 497-504, 2019 05.
Article in English | MEDLINE | ID: mdl-30624086

ABSTRACT

PURPOSE: To examine if current development on using contact lenses for drug delivery of cysteamine to treat ocular symptoms of cystinosis can be tinted to mitigate photophobia common in patients by reducing transmittance Methods: Commercial contact lenses were placed in a carbon black solution to examine loading after lens synthesis. Silicone hydrogel contact lenses were also synthesized with carbon black added prior to UV curing. Transmittance was measured using UV-vis spectrophotometry over the range of 190-1190 nm and compared to unmodified contact lenses. Lens parameters of refractive index, ion permeability, and Young's modulus were measured using a refractometer, release of sodium chloride, and the cantilever method. Cysteamine release was measured by loading lenses into 5% cysteamine solution and then monitoring the release of the drug using UV-vis spectrophotometry. Vitamin E diffusion barriers were also added to lenses via ethanol solution, and the release of cysteamine from these modified lenses was also examined. RESULTS: No leeching of carbon black was detected during experiments. Loading of pre-made contact lenses led to uneven distribution of carbon black throughout lens. Adding 0.3% carbon black to lens monomer solution prior to UV-curing led to even distribution and a transmittance reduction of approximately 50%. Ion permeability was reduced from 6.19 ± 0.90 x 10-3 to 1.28 ± 0.06 x 10-3 mm2 min-1, and Young's modulus was decreased from 1.58 ± 0.08 to 1.29 ± 0.06 MPa. Cysteamine releases from carbon black lenses with and without vitamin E were comparable to controls, although the loading solution of vitamin E/ethanol had to be tripled to achieve a similar mass loading to control. CONCLUSIONS: Carbon black increases the softness of contact lenses, but a loading of 0.3% maintains lens parameters required for wear. The release of cysteamine is also possible with carbon black lenses, albeit requiring a higher loading concentration of vitamin E to achieve similar release times.


Subject(s)
Contact Lenses, Hydrophilic , Corneal Diseases/metabolism , Cystine Depleting Agents/pharmacokinetics , Cystinosis/metabolism , Drug Delivery Systems , Photophobia/prevention & control , Soot/chemistry , Animals , Antihypertensive Agents/pharmacokinetics , Corneal Diseases/drug therapy , Cysteamine/pharmacokinetics , Cysteamine/therapeutic use , Cystine Depleting Agents/therapeutic use , Cystinosis/drug therapy , Elastic Modulus , Microscopy, Electron, Scanning , Rabbits , Refractometry , Spectrophotometry, Ultraviolet , Timolol/pharmacokinetics , Vitamin E/administration & dosage , Vitamins/administration & dosage
18.
Pediatr Nephrol ; 34(4): 571-578, 2019 04.
Article in English | MEDLINE | ID: mdl-29260317

ABSTRACT

Cystinosis is a rare autosomal-recessive lysosomal storage disease with high morbidity and mortality. It is caused by mutations in the CTNS gene that encodes the cystine transporter, cystinosin, which leads to lysosomal cystine accumulation. Patients with infantile nephropathic cystinosis, the most common and most severe clinical form of cystinosis, commonly present with renal Fanconi syndrome by 6-12 months of age, and without specific treatment, almost all will develop end-stage renal disease (ESRD) by 10-12 years of age. Early corneal cystine crystal deposition is a hallmark of the disease. Cystinosis also presents with gastrointestinal symptoms (e.g., vomiting, decreased appetite, and feeding difficulties) and severe growth retardation and may affect several other organs over time, including the eye, thyroid gland, gonads, pancreas, muscles, bone marrow, liver, nervous system, lungs, and bones. Cystine-depleting therapy with cysteamine orally is the only specific targeted therapy available for managing cystinosis and needs to be combined with cysteamine eye drops for corneal disease involvement. In patients with early treatment initiation and good compliance to therapy, long-term cysteamine treatment delays progression to ESRD, significantly improves growth, decreases the frequency and severity of extrarenal complications, and is associated with extended life expectancy. Therefore, early diagnosis of cystinosis and adequate life-long treatment with cysteamine are essential for preventing end-organ damage and improving the overall prognosis in these patients.


Subject(s)
Cysteamine/administration & dosage , Cystine Depleting Agents/administration & dosage , Cystinosis/drug therapy , Amino Acid Transport Systems, Neutral/genetics , Cysteamine/adverse effects , Cystine Depleting Agents/adverse effects , Cystinosis/complications , Cystinosis/diagnosis , Cystinosis/genetics , Disease Progression , Drug Administration Schedule , Genetic Predisposition to Disease , Humans , Mutation , Time Factors , Treatment Outcome
19.
Indian J Ophthalmol ; 67(1): 153-155, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30574930

ABSTRACT

A 36-year-old female presented initially with photophobia and visual deterioration. After examination and laboratory tests, patient was diagnosed with cystinosis. Cysteamine drops 4 × 1 drops/day was given as treatment for 1 year. During follow-up, in vivo confocal microscopy (IVCM) and anterior segment optical coherence tomography (AS-OCT) was performed. Photophobia was relieved and IVCM obtained the decrease in size and density of corneal crystals 1 year after. Depth of corneal crystals did not change but crystal density score reduced with cysteamine treatment.


Subject(s)
Anterior Eye Segment/diagnostic imaging , Cornea/pathology , Corneal Diseases/diagnosis , Cysteamine/administration & dosage , Cystinosis/drug therapy , Microscopy, Confocal/methods , Tomography, Optical Coherence/methods , Adult , Corneal Diseases/drug therapy , Crystallization , Cystine Depleting Agents/administration & dosage , Cystinosis/diagnosis , Female , Follow-Up Studies , Humans , Ophthalmic Solutions , Visual Acuity
20.
Int J Pharm ; 549(1-2): 466-475, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30099213

ABSTRACT

Cystinosis is a genetic disorder that leads to the formation of cystine crystals in many organs in the body including cornea. Ocular manifestation of this disease is treated by eye drops of cysteamine which can easily oxidize into its disulfide cystamine. The rapid oxidation limits the shelf life as well the duration during which the drug can be used after opening the eye drop bottle. We evaluate two approaches of preventing the oxidation of cysteamine with the goal of increasing the time of use after opening the bottle to one month. The first approach integrates antioxidants such as catalase enzyme and vitamins C and E into the aqueous solution. Results show that catalase is the most effective additive as it decreases the oxidation rate by 58%, which on its own is not sufficient to reach targeted one month stability. The second approach focuses on incorporating diffusion barriers to prevent oxygen from reaching the cysteamine solution. This was accomplished by two methods: formulation of a hydrophobic layer which floats on the surface of the aqueous solution and integration of OMAC® oxygen-resistant material into the eye drop bottle. Both methods delay the onset of cysteamine degradation and decrease the rate of degradation. In particular, an eye drop bottle with three layers of OMAC® has less than 10% degradation after one month of opening the bottle and withdrawing a drop each day. By integrating all three methods, we designed a system where >90% of cysteamine remains in the active form for 70 days after opening the bottle. In addition, we examine the use of OMAC® as heat-sealed pouches for storage of cysteamine eye drop bottles during packaging to eliminate the need for the current approach of freezing the formulation during shipping. The results show that such heat-sealed pouches would keep cysteamine stable for over one year at ambient conditions.


Subject(s)
Antioxidants/chemistry , Catalase/chemistry , Cysteamine/chemistry , Cystine Depleting Agents/chemistry , Drug Packaging , Oxygen/chemistry , Soybean Oil/chemistry , Administration, Ophthalmic , Ascorbic Acid/chemistry , Cysteamine/administration & dosage , Cystine Depleting Agents/administration & dosage , Drug Compounding , Drug Stability , Hydrophobic and Hydrophilic Interactions , Ophthalmic Solutions , Oxidation-Reduction , Technology, Pharmaceutical/methods , Time Factors , Vitamin E/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...