Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.262
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 32(12): 1988-2004, 2023 06 05.
Article in English | MEDLINE | ID: mdl-36795052

ABSTRACT

SURF1 deficiency (OMIM # 220110) causes Leigh syndrome (LS, OMIM # 256000), a mitochondrial disorder typified by stress-induced metabolic strokes, neurodevelopmental regression and progressive multisystem dysfunction. Here, we describe two novel surf1-/- zebrafish knockout models generated by CRISPR/Cas9 technology. While gross larval morphology, fertility, and survival into adulthood appeared unaffected, surf1-/- mutants manifested adult-onset ocular anomalies and decreased swimming activity, as well as classical biochemical hallmarks of human SURF1 disease, including reduced complex IV expression and enzymatic activity and increased tissue lactate. surf1-/- larvae also demonstrated oxidative stress and stressor hypersensitivity to the complex IV inhibitor, azide, which exacerbated their complex IV deficiency, reduced supercomplex formation, and induced acute neurodegeneration typical of LS including brain death, impaired neuromuscular responses, reduced swimming activity, and absent heartrate. Remarkably, prophylactic treatment of surf1-/- larvae with either cysteamine bitartrate or N-acetylcysteine, but not other antioxidants, significantly improved animal resiliency to stressor-induced brain death, swimming and neuromuscular dysfunction, and loss of heartbeat. Mechanistic analyses demonstrated cysteamine bitartrate pretreatment did not improve complex IV deficiency, ATP deficiency, or increased tissue lactate but did reduce oxidative stress and restore glutathione balance in surf1-/- animals. Overall, two novel surf1-/- zebrafish models recapitulate the gross neurodegenerative and biochemical hallmarks of LS, including azide stressor hypersensitivity that was associated with glutathione deficiency and ameliorated by cysteamine bitartrate or N-acetylcysteine therapy.


Subject(s)
Cytochrome-c Oxidase Deficiency , Leigh Disease , Animals , Adult , Humans , Leigh Disease/drug therapy , Leigh Disease/genetics , Leigh Disease/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Acetylcysteine , Cysteamine/pharmacology , Azides/metabolism , Brain Death , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Glutathione/metabolism , Lactates
2.
Biochem Biophys Res Commun ; 677: 168-181, 2023 10 15.
Article in English | MEDLINE | ID: mdl-37597441

ABSTRACT

Transient blockade of glycine decarboxylase (GLDC) can restrict de novo pyrimidine synthesis, which is a well-described strategy for enhancing the host interferon response to viral infection and a target pathway for some licenced anti-inflammatory therapies. The aminothiol, cysteamine, is produced endogenously during the metabolism of coenzyme A, and is currently being investigated in a clinical trial as an intervention in community acquired pneumonia resulting from viral (influenza and SARS-CoV-2) and bacterial respiratory infection. Cysteamine is known to inhibit both bacterial and the eukaryotic host glycine cleavage systems via competitive inhibition of GLDC at concentrations, lower than those required for direct antimicrobial or antiviral activity. Here, we demonstrate for the first time that therapeutically achievable concentrations of cysteamine can inhibit glycine utilisation by epithelial cells and improve cell-mediated responses to infection with respiratory viruses, including human coronavirus 229E and Influenza A. Cysteamine reduces interleukin-6 (IL-6) and increases the interferon-λ (IFN-λ) response to viral challenge and in response to liposomal polyinosinic:polycytidylic acid (poly I:C) simulant of RNA viral infection.


Subject(s)
COVID-19 , Influenza, Human , Virus Diseases , Humans , Cysteamine/pharmacology , Influenza, Human/drug therapy , SARS-CoV-2 , Virus Diseases/drug therapy , Immunity, Innate , Epithelial Cells
3.
Exp Eye Res ; 226: 109338, 2023 01.
Article in English | MEDLINE | ID: mdl-36470430

ABSTRACT

Corneal wound healing is integral for resolution of corneal disease or for post-operative healing. However, corneal scarring that may occur secondary to this process can significantly impair vision. Tissue transglutaminase 2 (TGM2) inhibition has shown promising antifibrotic effects and thus holds promise to prevent or treat corneal scarring. The commercially available ocular solution for treatment of ocular manifestations of Cystinosis, Cystaran®, contains the TGM2 inhibitor cysteamine hydrochloride (CH). The purpose of this study is to assess the safety of CH on corneal epithelial and stromal wounds, its effects on corneal wound healing, and its efficacy against corneal scarring following wounding. Quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC) were first used to quantify and localize TGM2 expression in the cornea. Subsequently, (i) the in vitro effects of CH at 0.163, 1.63, and 16.3 mM on corneal epithelial cell migration was assessed with an epithelial cell migration assay, and (ii) the in vivo effects of application of 1.63 mM CH on epithelial and stromal wounds was assessed in a rabbit model with ophthalmic examinations, inflammation scoring, color and fluorescein imaging, optical coherence tomography (OCT), and confocal biomicroscopy. Post-mortem assessment of corneal tissue post-stromal wounding included biomechanical characterization (atomic force microscopy (AFM)), histology (H&E staining), and determining incidence of myofibroblasts (immunostaining against α-SMA) in wounded corneal tissue. TGM2 expression was highest in corneal epithelial cells. Application of the TGM2 inhibitor CH did not affect in vitro epithelial cell migration at the two lower concentrations tested. At 16.3 mM, decreased cell migration was observed. In vivo application of CH at 57 mM was well tolerated and did not adversely affect wound healing. No difference in corneal scarring was found between CH treated and vehicle control eyes. This study shows that the TGM2 inhibitor CH, at the FDA-approved dose, is well tolerated in a rabbit model of corneal wound healing and does not adversely affect epithelial or stromal wound healing. This supports the safe use of this medication in Cystinosis patients with open corneal wounds. CH did not have an effect on corneal scarring in this study, suggesting that Cystaran® administration to patients with corneal wounds is unlikely to decrease corneal fibrosis.


Subject(s)
Corneal Injuries , Cysteamine , Cystinosis , Epithelium, Corneal , Animals , Rabbits , Cicatrix/metabolism , Cornea/drug effects , Cornea/metabolism , Corneal Diseases/pathology , Corneal Injuries/drug therapy , Corneal Injuries/metabolism , Cysteamine/pharmacology , Cysteamine/therapeutic use , Cysteamine/metabolism , Cystinosis/metabolism , Cystinosis/pathology , Epithelium, Corneal/pathology , Protein Glutamine gamma Glutamyltransferase 2/antagonists & inhibitors , Wound Healing/drug effects
4.
Int J Mol Sci ; 24(6)2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36982309

ABSTRACT

N-propionyl-4-S-cysteaminylphenol (N-Pr-4-S-CAP) is a substrate for tyrosinase, which is a melanin biosynthesis enzyme and has been shown to be selectively incorporated into melanoma cells. It was found to cause selective cytotoxicity against melanocytes and melanoma cells after selective incorporation, resulting in the induction of anti-melanoma immunity. However, the underlying mechanisms for the induction of anti-melanoma immunity remain unclear. This study aimed to elucidate the cellular mechanism for the induction of anti-melanoma immunity and clarify whether N-Pr-4-S-CAP administration could be a new immunotherapeutic approach against melanoma, including local recurrence and distant metastasis. A T cell depletion assay was used for the identification of the effector cells responsible for N-Pr-4-S-CAP-mediated anti-melanoma immunity. A cross-presentation assay was carried out by using N-Pr-4-S-CAP-treated B16-OVA melanoma-loaded bone marrow-derived dendritic cells (BMDCs) and OVA-specific T cells. Administration of N-Pr-4-S-CAP induced CD8+ T cell-dependent anti-melanoma immunity and inhibited the growth of challenged B16F1 melanoma cells, indicating that the administration of N-Pr-4-S-CAP can be a prophylactic therapy against recurrence and metastasis of melanoma. Moreover, intratumoral injection of N-Pr-4-S-CAP in combination with BMDCs augmented the tumor growth inhibition when compared with administration of N-Pr-4-S-CAP alone. BMDCs cross-presented a melanoma-specific antigen to CD8+ T cells through N-Pr-4-S-CAP-mediated melanoma cell death. Combination therapy using N-Pr-4-S-CAP and BMDCs elicited a superior anti-melanoma effect. These results suggest that the administration of N-Pr-4-S-CAP could be a new strategy for the prevention of local recurrence and distant metastasis of melanoma.


Subject(s)
CD8-Positive T-Lymphocytes , Melanoma, Experimental , Animals , Mice , Phenols/pharmacology , Cysteamine/pharmacology , Melanoma, Experimental/drug therapy , Mice, Inbred C57BL , Melanoma, Cutaneous Malignant
5.
Int J Mol Sci ; 24(2)2023 Jan 07.
Article in English | MEDLINE | ID: mdl-36674717

ABSTRACT

Host-directed therapies are emerging as a promising tool in the curing of difficult-to-treat infections, such as those caused by drug-resistant bacteria. In this study, we aim to test the potential activity of the FDA- and EMA-approved drugs cysteamine and cystamine against Mycobacterium abscessus. In human macrophages (differentiated THP-1 cells), these drugs restricted M. abscessus growth similar to that achieved by amikacin. Here, we use the human ex vivo granuloma-like structures (GLS) model of infection with the M. abscessus rough (MAB-R) and smooth (MAB-S) variants to study the activity of new therapies against M. abscessus. We demonstrate that cysteamine and cystamine show a decrease in the number of total GLSs per well in the MAB-S and MAB-R infected human peripheral blood mononuclear cells (PBMCs). Furthermore, combined administration of cysteamine or cystamine with amikacin resulted in enhanced activity against the two M. abscessus morpho variants compared to treatment with amikacin only. Treatment with cysteamine and cystamine was more effective in reducing GLS size and bacterial load during MAB-S infection compared with MAB-R infection. Moreover, treatment with these two drugs drastically quenched the exuberant proinflammatory response triggered by the MAB-R variant. These findings showing the activity of cysteamine and cystamine against the R and S M. abscessus morphotypes support the use of these drugs as novel host-directed therapies against M. abscessus infections.


Subject(s)
Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Humans , Amikacin/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cysteamine/pharmacology , Cysteamine/therapeutic use , Cystamine/pharmacology , Cystamine/therapeutic use , Leukocytes, Mononuclear , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Microbial Sensitivity Tests
6.
Trop Anim Health Prod ; 55(2): 69, 2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36749468

ABSTRACT

This study aimed to determine the effects of coated cysteamine hydrochloride (CSH) and probiotics (PB) supplemented alone or in combination on feed intake, digestibility, ruminal fermentation, and blood metabolites of heifer beef cattle. Sixteen heifers (body weight = 210 ± 41 kg; age = 9 ± 2 months) were assigned according to a randomized complete block design in a 2 × 2 factorial arrangement. All animals were fed the basal diet, which contained an 82:17 concentrate-to-forage ratio, and the forage source was rice straw. The treatments were as follows: (1) 0% PB + 0 g/d CSH, (2) 0.1% PB + 0 g/d CSH, (3) 0% PB + 20 g/d CSH, and (4) 0.1% PB + 20 g/d CSH. The main effect of CSH supplementation has been found to improve feed intake (P < 0.05). There were no treatment interactions with nutrient digestibility or rumen fermentation parameters. Supplementation of CSH did not affect any of the variables evaluated, while probiotics supplementation increased DM digestibility due to the increases in CP and fiber fraction digestibility. Compared to controls and CSH, at 16 h post-feeding, heifers receiving probiotics tended (P = 0.07) to show 17% greater ruminal NH3-N concentration, but this effect was not evident at 2 h post-feeding. However, the main effects of probiotic supplementation showed a tendency to increase the number of total bacteria and fungal zoospores in the rumen at 2 h post-feeding. The blood triglyceride (BTG) concentration of heifers fed a diet supplemented with 20 g/d CSH and 0.1% probiotics was found to be greater than those fed CSH alone (P < 0.1) at 16 h post-feeding, and then, there were greater BTG concentrations than other treatments (P < 0.05) at 2 h post-feeding. In conclusion, the combination of CSH and PB did not potentiate the effects of probiotics on digestibility and rumen fermentation and had minimal effects on blood parameters.


Subject(s)
Cysteamine , Probiotics , Cattle , Animals , Female , Cysteamine/metabolism , Cysteamine/pharmacology , Fermentation , Digestion , Animal Feed/analysis , Dietary Supplements , Diet/veterinary , Eating , Nutrients , Rumen/metabolism
7.
Am J Physiol Renal Physiol ; 323(2): F156-F170, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35695380

ABSTRACT

The lysosomal storage disease cystinosis is caused by mutations in CTNS, encoding the cystine transporter cystinosin, and in its severest form leads to proximal tubule dysfunction followed by kidney failure. Patients receive the drug-based therapy cysteamine from diagnosis. However, despite long-term treatment, cysteamine only slows the progression of end-stage renal disease. Preclinical testing in cystinotic rodents is required to evaluate new therapies; however, the current models are suboptimal. To solve this problem, we generated a new cystinotic rat model using CRISPR/Cas9-mediated gene editing to disrupt exon 3 of Ctns and measured various parameters over a 12-mo time course. Ctns-/- rats display hallmarks of cystinosis by 3-6 mo of age, as demonstrated by a failure to thrive, excessive thirst and urination, cystine accumulation in tissues, corneal cystine crystals, loss of LDL receptor-related protein 2 in proximal tubules, and immune cell infiltration. High levels of glucose, calcium, albumin, and protein were excreted at 6 mo of age, consistent with the onset of Fanconi syndrome, with a progressive diminution of urine urea and creatinine from 9 mo of age, indicative of chronic kidney disease. Kidney histology and immunohistochemistry showed proximal tubule atrophy and glomerular damage as well as classic "swan neck" lesions. Overall, Ctns-/- rats show a disease progression that more faithfully recapitulates nephropathic cystinosis than existing rodent models. The Ctns-/- rat provides an excellent new rodent model of nephropathic cystinosis that is ideally suited for conducting preclinical drug testing and is a powerful tool to advance cystinosis research.NEW & NOTEWORTHY Animal models of disease are essential to perform preclinical testing of new therapies before they can progress to clinical trials. The cystinosis field has been hampered by a lack of suitable animal models that fully recapitulate the disease. Here, we generated a rat model of cystinosis that closely models the human condition in a timeframe that makes them an excellent model for preclinical drug testing as well as being a powerful tool to advance research.


Subject(s)
Amino Acid Transport Systems, Neutral , Cystinosis , Fanconi Syndrome , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Cysteamine/pharmacology , Cysteamine/therapeutic use , Cystine/genetics , Cystine/metabolism , Cystine/therapeutic use , Cystinosis/drug therapy , Cystinosis/genetics , Cystinosis/metabolism , Fanconi Syndrome/genetics , Phenotype , Rats
8.
Am J Physiol Lung Cell Mol Physiol ; 323(3): L372-L389, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35762590

ABSTRACT

The redox status of the cysteine-rich SARS-CoV-2 spike glycoprotein (SARS-2-S) is important for the binding of SARS-2-S to angiotensin-converting enzyme 2 (ACE2), suggesting that drugs with a functional thiol group ("thiol drugs") may cleave cystines to disrupt SARS-CoV-2 cell entry. In addition, neutrophil-induced oxidative stress is a mechanism of COVID-19 lung injury, and the antioxidant and anti-inflammatory properties of thiol drugs, especially cysteamine, may limit this injury. To first explore the antiviral effects of thiol drugs in COVID-19, we used an ACE-2 binding assay and cell entry assays utilizing reporter pseudoviruses and authentic SARS-CoV-2 viruses. We found that multiple thiol drugs inhibit SARS-2-S binding to ACE2 and virus infection. The most potent drugs were effective in the low millimolar range, and IC50 values followed the order of their cystine cleavage rates and lower thiol pKa values. To determine if thiol drugs have antiviral effects in vivo and to explore any anti-inflammatory effects of thiol drugs in COVID-19, we tested the effects of cysteamine delivered intraperitoneally to hamsters infected with SARS-CoV-2. Cysteamine did not decrease lung viral infection, but it significantly decreased lung neutrophilic inflammation and alveolar hemorrhage. We speculate that the concentration of cysteamine achieved in the lungs with intraperitoneal delivery was insufficient for antiviral effects but sufficient for anti-inflammatory effects. We conclude that thiol drugs decrease SARS-CoV-2 lung inflammation and injury, and we provide rationale for future studies to test if direct (aerosol) delivery of thiol drugs to the airways might also result in antiviral effects.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 Drug Treatment , Anti-Inflammatory Agents/pharmacology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cysteamine/pharmacology , Humans , Peptidyl-Dipeptidase A/metabolism , Pharmaceutical Preparations , SARS-CoV-2 , Sulfhydryl Compounds/pharmacology
9.
Mol Genet Metab ; 137(1-2): 192-200, 2022.
Article in English | MEDLINE | ID: mdl-36115282

ABSTRACT

Global COVID-19 pandemic is caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Continuous emergence of new variants and their rapid spread are jeopardizing vaccine countermeasures to a significant extent. While currently available vaccines are effective at preventing illness associated with SARS-CoV-2 infection, these have been shown to be less effective at preventing breakthrough infection and transmission from a vaccinated individual to others. Here we demonstrate broad antiviral activity of cysteamine HCl in vitro against major emergent infectious variants of SARS-CoV-2 in a highly permissible Vero cell line. Cysteamine HCl inhibited infection of wild type, alpha, beta, gamma, delta, lambda, and omicron variants effectively. Cysteamine is a very well-tolerated US FDA-approved drug used chronically as a topical ophthalmic solution to treat ocular cystinosis in patients who receive it hourly or QID lifelong at concentrations 6 times higher than that required to inhibit SARS CoV-2 in tissue culture. Application of cysteamine as a topical nasal treatment can potentially1) mitigate existing infection 2) prevent infection in exposed individuals, and 3) limit the contagion in vulnerable populations.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Humans , Pandemics , Cysteamine/pharmacology , Antiviral Agents/pharmacology , Ophthalmic Solutions
10.
Andrologia ; 54(1): e14243, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34498746

ABSTRACT

Oxidative stress, inflammation and apoptosis are major pathways in pathophysiology of testicular torsion/detorsion (TTDT) reperfusion injury. This study evaluated the antioxidant, anti-inflammatory and anti-apoptotic role of cysteamine in TTDT-induced injury. Male Wistar rats (n = 32) were grouped into four (n = 8): sham, ischaemia-reperfusion injury (IRI), cysteamine (100 mg/kg and 200 mg/kg) for in vivo study. Samples were taken for biomolecular and histological evaluation 48 hr after detorsion. Tissue SOD, GPx, GSH, GST activity, total thiol, H2 O2 and MDA were assessed. Serum levels of NO, MPO, TNF-alpha and IL-6 and sperm motility, count and viability were assessed. Caspase-3 and bax were evaluated by immunohistochemistry. Significant difference was set as p < .05. Significant increase in H2 O2, MDA and nitrite but reduction in SOD, GPx, GSH, GST and total thiol in the testicular tissue of IRI rats was reversed by cysteamine. Serum MPO and TNF-α were significantly elevated in RI, while treated-RI rats showed decrease (p < .05) in tissue level of the inflammation markers. Reduced sperm motility in RI was significantly reversed by cysteamine. Increased tissue expression of bax and caspase-3 was reversed by cysteamine. Cysteamine protected the testis against reperfusion injury through anti-inflammatory, antioxidant effects and inhibition of apoptosis in rats.


Subject(s)
Reperfusion Injury , Spermatic Cord Torsion , Animals , Apoptosis , Cysteamine/metabolism , Cysteamine/pharmacology , Male , Malondialdehyde/metabolism , Oxidative Stress , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Sperm Motility , Spermatic Cord Torsion/complications , Spermatic Cord Torsion/drug therapy , Spermatic Cord Torsion/metabolism , Testis/metabolism
11.
Hum Mol Genet ; 28(11): 1837-1852, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30668749

ABSTRACT

Cysteamine bitartrate is a US Food and Drug Administration-approved therapy for nephropathic cystinosis also postulated to enhance glutathione biosynthesis. We hypothesized this antioxidant effect may reduce oxidative stress in primary mitochondrial respiratory chain (RC) disease, improving cellular viability and organismal health. Here, we systematically evaluated the therapeutic potential of cysteamine bitartrate in RC disease models spanning three evolutionarily distinct species. These pre-clinical studies demonstrated the narrow therapeutic window of cysteamine bitartrate, with toxicity at millimolar levels directly correlating with marked induction of hydrogen peroxide production. Micromolar range cysteamine bitartrate treatment in Caenorhabditis elegans gas-1(fc21) RC complex I (NDUFS2-/-) disease invertebrate worms significantly improved mitochondrial membrane potential and oxidative stress, with corresponding modest improvement in fecundity but not lifespan. At 10 to 100 µm concentrations, cysteamine bitartrate improved multiple RC complex disease FBXL4 human fibroblast survival, and protected both complex I (rotenone) and complex IV (azide) Danio rerio vertebrate zebrafish disease models from brain death. Mechanistic profiling of cysteamine bitartrate effects showed it increases aspartate levels and flux, without increasing total glutathione levels. Transcriptional normalization of broadly dysregulated intermediary metabolic, glutathione, cell defense, DNA, and immune pathways was greater in RC disease human cells than in C. elegans, with similar rescue in both models of downregulated ribosomal and proteasomal pathway expression. Overall, these data suggest cysteamine bitartrate may hold therapeutic potential in RC disease, although not through obvious modulation of total glutathione levels. Careful consideration is required to determine safe and effective cysteamine bitartrate concentrations to further evaluate in clinical trials of human subjects with primary mitochondrial RC disease.


Subject(s)
Antioxidants/pharmacology , Caenorhabditis elegans Proteins/genetics , Cysteamine/pharmacology , Mitochondrial Diseases/drug therapy , NADH Dehydrogenase/genetics , Animals , Brain Death/metabolism , Brain Death/pathology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Dose-Response Relationship, Drug , Electron Transport/drug effects , F-Box Proteins/genetics , Fertility/drug effects , Fibroblasts/drug effects , Glutathione/genetics , Glutathione/metabolism , Humans , Hydrogen Peroxide , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Oxidative Stress/drug effects , Ubiquitin-Protein Ligases/genetics , Zebrafish/genetics
12.
Anim Biotechnol ; 32(4): 401-412, 2021 Aug.
Article in English | MEDLINE | ID: mdl-31900040

ABSTRACT

This study elucidated the molecular markers that decrease oocyte quality during in vitro culture, restricting optimal developmental potential. Here, we evaluated the transcriptomic differences between cysteamine-treated and non-treated bovine cumulus oocyte complexes (COCs) after 22 h of co-culture in the maturation media using RNA sequencing. In total, 39,014 transcripts were sequenced between cysteamine-treated and non-treated mature COCs. We evaluated the relative expression of 21,472 genes, with 59 genes showing differential expression between the two COC groups. The cysteamine-treated group had 36 up-regulated gene transcripts and 23 down-regulated gene transcripts. Moreover, gene ontology (GO) enrichment analysis revealed that multiple biological processes were significantly enriched after cysteamine supplementation. Differentially expressed genes appeared to maintain normal oocyte physiology, regulation of apoptosis, differentiation, ossification or bone formation, cardiac and muscle physiology, hormonal secretion, and membrane construction for further embryonic development. In conclusion, cysteamine affects the mRNA level of COCs during oocyte maturation by upregulating potential molecular markers and downregulating genes that affect further embryonic development.


Subject(s)
Cattle , Cysteamine , Oocytes , Transcriptome , Animals , Cattle/genetics , Cysteamine/pharmacology , Dietary Supplements , Gene Expression Profiling , Republic of Korea
13.
J Am Soc Nephrol ; 31(5): 1092-1106, 2020 05.
Article in English | MEDLINE | ID: mdl-32273301

ABSTRACT

BACKGROUND: Nephropathic cystinosis, a hereditary lysosomal storage disorder caused by dysfunction of the lysosomal cotransporter cystinosin, leads to cystine accumulation and cellular damage in various organs, particularly in the kidney. Close therapeutic monitoring of cysteamine, the only available disease-modifying treatment, is recommended. White blood cell cystine concentration is the current gold standard for therapeutic monitoring, but the assay is technically demanding and is available only on a limited basis. Because macrophage-mediated inflammation plays an important role in the pathogenesis of cystinosis, biomarkers of macrophage activation could have potential for the therapeutic monitoring of cystinosis. METHODS: We conducted a 2-year prospective, longitudinal study in which 61 patients with cystinosis who were receiving cysteamine therapy were recruited from three European reference centers. Each regular care visit included measuring four biomarkers of macrophage activation: IL-1ß, IL-6, IL-18, and chitotriosidase enzyme activity. RESULTS: A multivariate linear regression analysis of the longitudinal data for 57 analyzable patients found chitotriosidase enzyme activity and IL-6 to be significant independent predictors for white blood cell cystine levels in patients of all ages with cystinosis; a receiver operating characteristic analysis ranked chitotriosidase as superior to IL-6 in distinguishing good from poor therapeutic control (on the basis of white blood cell cystine levels of <2 nmol 1/2 cystine/mg protein or ≥2 nmol 1/2 cystine/mg protein, respectively). Moreover, in patients with at least one extrarenal complication, chitotriosidase significantly correlated with the number of extrarenal complications and was superior to white blood cell cystine levels in predicting the presence of multiple extrarenal complications. CONCLUSIONS: Chitotriosidase enzyme activity holds promise as a biomarker for use in therapeutic monitoring of nephropathic cystinosis.


Subject(s)
Cysteamine/therapeutic use , Cystinosis/blood , Drug Monitoring/methods , Hexosaminidases/blood , Macrophage Activation/drug effects , Adolescent , Adult , Biomarkers , Child , Cysteamine/pharmacology , Cystine/blood , Cystinosis/drug therapy , Female , Humans , Inflammation , Interleukin-18/blood , Interleukin-1beta/blood , Interleukin-6/blood , Leukocytes/chemistry , Male , Medication Adherence , Peptide Fragments/blood , Prospective Studies , Young Adult
14.
J Am Soc Nephrol ; 31(5): 962-982, 2020 05.
Article in English | MEDLINE | ID: mdl-32198276

ABSTRACT

BACKGROUND: Mutations in CTNS-a gene encoding the cystine transporter cystinosin-cause the rare, autosomal, recessive, lysosomal-storage disease cystinosis. Research has also implicated cystinosin in modulating the mTORC1 pathway, which serves as a core regulator of cellular metabolism, proliferation, survival, and autophagy. In its severest form, cystinosis is characterized by cystine accumulation, renal proximal tubule dysfunction, and kidney failure. Because treatment with the cystine-depleting drug cysteamine only slows disease progression, there is an urgent need for better treatments. METHODS: To address a lack of good human-based cell culture models for studying cystinosis, we generated the first human induced pluripotent stem cell (iPSC) and kidney organoid models of the disorder. We used a variety of techniques to examine hallmarks of cystinosis-including cystine accumulation, lysosome size, the autophagy pathway, and apoptosis-and performed RNA sequencing on isogenic lines to identify differentially expressed genes in the cystinosis models compared with controls. RESULTS: Compared with controls, these cystinosis models exhibit elevated cystine levels, increased apoptosis, and defective basal autophagy. Cysteamine treatment ameliorates this phenotype, except for abnormalities in apoptosis and basal autophagy. We found that treatment with everolimus, an inhibitor of the mTOR pathway, reduces the number of large lysosomes, decreases apoptosis, and activates autophagy, but it does not rescue the defect in cystine loading. However, dual treatment of cystinotic iPSCs or kidney organoids with cysteamine and everolimus corrects all of the observed phenotypic abnormalities. CONCLUSIONS: These observations suggest that combination therapy with a cystine-depleting drug such as cysteamine and an mTOR pathway inhibitor such as everolimus has potential to improve treatment of cystinosis.


Subject(s)
Cysteamine/therapeutic use , Cystinosis/drug therapy , Disease Models, Animal , Everolimus/therapeutic use , Induced Pluripotent Stem Cells/transplantation , Organoids/transplantation , TOR Serine-Threonine Kinases/antagonists & inhibitors , Amino Acid Transport Systems, Neutral/deficiency , Amino Acid Transport Systems, Neutral/genetics , Animals , Autophagy/drug effects , CRISPR-Cas Systems , Cell Line , Cysteamine/pharmacology , Cystine/blood , Drug Evaluation, Preclinical , Drug Therapy, Combination , Everolimus/pharmacology , Gene Editing , Heterografts , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/ultrastructure , Lysosomes/drug effects , Lysosomes/ultrastructure , Mice , Mice, SCID , Organoids/metabolism , Phenotype
15.
Neurobiol Dis ; 145: 105042, 2020 11.
Article in English | MEDLINE | ID: mdl-32798729

ABSTRACT

A number of publications have reported that cysteamine has significant therapeutic effects on several aspects of Parkinson's disease (PD)-related pathology but none of these studies have evaluated its impact on pathological forms of α-Synuclein (α-Syn), one of the main hallmarks of PD. We therefore tested the efficacy of cysteamine on the Thy1-α-Syn mouse model which over-expresses full-length human wild-type α-Syn. Two-month (early stage disease) and 6-month old (late stage disease) mice and littermate controls were treated daily with cysteamine (20 mg/kg, i.p.) to assess the protective and restorative properties of this compound. After 6 weeks of treatment, animals were tested using a battery of motor tests. Cysteamine-treated transgenic mice displayed significant improvements in motor performance as compared to saline-treated transgenic littermates. Post-mortem readouts revealed a reduction in fibrillation, phosphorylation and total levels of overexpresed human α-Syn. To determine if such outcomes extended to human cells, the benefits of cysteamine were additionally tested using 6-hydroxydopamine (6-OHDA) treated neurons differentiated from induced pluripotent stem cells (iPSCs) derived from a PD patient harbouring a triplication of the SNCA gene. SNCA neurons treated with cysteamine exhibited significantly more intact/healthy neurites than cells treated with 6-OHDA alone. Additionally, SNCA neurons treated with cysteamine in the absence of 6-OHDA showed a trend towards lower total α-Syn levels. Overall, our in vivo and in vitro findings suggest that cysteamine can act as a disease-modifying molecule by enhancing -the survival of dopaminergic neurons and reducing pathological forms of α-Syn.


Subject(s)
Cysteamine/pharmacology , Dopaminergic Neurons/drug effects , Induced Pluripotent Stem Cells/drug effects , Parkinsonian Disorders/pathology , alpha-Synuclein/genetics , Animals , Dopaminergic Neurons/pathology , Humans , Locomotion/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
Bioorg Med Chem Lett ; 30(19): 127458, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32755678

ABSTRACT

Sirtuin proteins are a highly conserved class of nicotinamide adenine dinucleotide (NAD+)-dependent lysine deacylases. The pleiotropic human isoform 2 of Sirtuins (SIRT2) has been engaged in the pathogenesis of cancer in a plethora of reports around the globe. Thus, SIRT2 modulation is deemed as a promising approach for pharmaceutical intervention. Previously, we reported S-Trityl-l-Cysteine (STLC)-ornamented dimethylaminopyridine chemical entity named STC4 with a significant SIRT2 inhibitory capacity; this was separate from the conventional application of STLC scaffold as a kinesin-5 inhibitor. An interactive molecular docking study of SIRT2 and STC4 showed interaction between Asn168 of SIRT2 and the methyl ester of STC4, that appears to hinder STC4 to reach the selective pocket of the protein unlike strong SIRT2 inhibitor SirReal2. To improve its activity, herein, we utilized S-trityl cysteamine pharmacophore lacking the methyl ester. Nine compounds were synthesized and assayed affording three biopertinent SIRT2 inhibitors, and two of them, STCY1 and STCY6 showed higher inhibitory activity than STC4. These compounds have pronounced anti-proliferative activities against different cancer cell lines. A molecular docking study was executed to shed light on the supposed binding mode of the lead compound, STCY1, into the selective pocket of SIRT2 by interaction of the nitrogen of pyridine ring of the compound and Ala135 of the protein. The outcome of the study exposes that the active compounds are effective intermediates to construct more potent biological agents.


Subject(s)
Aminopyridines/pharmacology , Cysteamine/analogs & derivatives , Cysteamine/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Sirtuin 2/antagonists & inhibitors , Trityl Compounds/pharmacology , Aminopyridines/chemical synthesis , Aminopyridines/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cysteamine/metabolism , Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/metabolism , Humans , Molecular Docking Simulation , Molecular Structure , Protein Binding , Sirtuin 2/metabolism , Structure-Activity Relationship , Trityl Compounds/chemical synthesis , Trityl Compounds/metabolism
17.
Reprod Domest Anim ; 55(9): 1229-1239, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32644236

ABSTRACT

The aim of this study was to evaluate the effects of different concentrations of lycopene and cysteamine on characteristics of sperm, liquid peroxidation and enzymatic activities in seminal plasma of canine semen preserved at 5°C for 72 hr. The semen samples were divided into eight aliquots: control, control sham (dimethyl sulfoxide 5%), lycopene groups (250, 500 and 750 µg/ml) and cysteamine groups (2.5, 5 and 10 mM). Motility, viability, membrane integrity, DNA integrity, total antioxidant capacity (TAC) and malondialdehyde (MDA) levels were evaluated. Progressive motility and total motility were higher with the 500 and 750 µg/ml lycopene concentrations, respectively, compared to the control group and the cysteamine groups following 72 hr of storage in the liquid storage. Motility characteristics, viability and hypo-osmotic swelling test (HOST) percentages were significantly improved in 500 µg/ml lycopene compared to other groups. The 500 and 750 µg/ml lycopene concentrations, respectively, showed significantly reduced percentages of spermatozoa with DNA integrity compared to the control group. The 500 and 750 µg/ml lycopene concentrations, respectively, led to the significant decrease of MDA levels. The 500 µg/ml lycopene enhanced TAC levels after 48 and 72 hr that was not observed in other groups. In conclusion, the findings of this study showed that lycopene supplementation in canine semen extenders improved canine semen parameters and TAC levels and decreased MDA levels in the chilling process.


Subject(s)
Cysteamine/pharmacology , Lycopene/pharmacology , Semen Preservation/veterinary , Spermatozoa/drug effects , Animals , Antioxidants/analysis , DNA Damage , Dogs , Male , Malondialdehyde , Oxidative Stress/drug effects , Semen Preservation/methods , Sperm Motility/drug effects
18.
Neurobiol Dis ; 130: 104530, 2019 10.
Article in English | MEDLINE | ID: mdl-31301344

ABSTRACT

To date, medical and surgical interventions offered to patients with Parkinson's disease (PD) serve only to manage clinical symptoms; they have not shown the capacity to halt nor reverse degenerative processes. There is therefore an urgent need to identify and/or develop therapeutic strategies that will demonstrate 'disease modifying' capacities. The molecule cystamine, and its reduced form cysteamine, act via a number of pathways determined to be critical to the pathogenesis of PD. In particular, cystamine is capable of crossing the blood-brain barrier, and both agents (cystamine and cysteamine) can promote the secretion of neurotrophic factors, inhibit oxidative stress, reduce inflammatory responses and importantly, have already been trialed in humans for a number of other clinical indications. In the last decade, our laboratory has accumulated compelling evidence that both cystamine and cysteamine can halt, and even reverse, ongoing neurodegenerative processes in a number of different models of PD, and as such, should now be taken forward to clinical trials in PD.


Subject(s)
Cysteamine/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Clinical Trials as Topic , Cysteamine/pharmacology , Humans , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects
19.
Biomacromolecules ; 20(10): 3864-3872, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31498594

ABSTRACT

The cryopreservation of cells underpins many areas of biotechnology, healthcare, and fundamental science by enabling the banking and distribution of cells. Cryoprotectants are essential to prevent cold-induced damage. Here, we demonstrate that extracellular localization of antifreeze proteins can significantly enhance post-thaw recovery of mammalian cell monolayers cryopreserved using dimethyl sulfoxide, whereas they show less benefit in suspension cryopreservation. A type III antifreeze protein (AFPIII) was used as the macromolecular ice recrystallization inhibitor and its intra/extracellular locations were controlled by using Pep-1, a cell-penetrating peptide. Flow cytometry and confocal microscopy confirmed successful delivery of AFPIII. The presence of extracellular AFPIII dramatically increased post-thaw recovery in a challenging 2-D cell monolayer system using just 0.8 mg·mL-1, from 25% to over 60%, whereas intracellularly delivered AFPIII showed less benefit. Interestingly, the antifreeze protein was less effective when used in suspension cryopreservation of the same cells, suggesting that the cryopreservation format is also crucial. These observations show that, in the discovery of macromolecular cryoprotectants, intracellular delivery of ice recrystallization inhibitors may not be a significant requirement under "slow freezing" conditions, which will help guide the design of new biomaterials, in particular, for cell storage.


Subject(s)
Antifreeze Proteins/pharmacology , Cell Survival/drug effects , Cryoprotective Agents/pharmacology , A549 Cells , Cell Line, Tumor , Cryopreservation/methods , Crystallization/methods , Cysteamine/analogs & derivatives , Cysteamine/pharmacology , Freezing , Humans , Ice/adverse effects , Peptides/pharmacology
20.
BMC Vet Res ; 15(1): 328, 2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31519201

ABSTRACT

BACKGROUND: Cysteamine was coated to cover its odor and maintain the stability. However, coated cysteamine (CC) has not been clearly evaluated for its effects on the gastrointestinal mucosa status. We hypothesize that the appropriate CC supplementation in diet impacts the stomach and intestinal mucosa variously through regulating the morphology, apoptosis, and oxidative stress status in model of pigs. RESULTS: The results showed that villus height increased (P < 0.05), and crypt depth decreased (P < 0.05) in the ileum when pigs were fed the diet with low cysteamine (LCS) compared with the control diet. The ileal lesion score in the LCS group was significantly (P < 0.01) lower than that in the control group, while the gastric lesion score in the CC group was significantly (P < 0.01) higher compared with that of the control group. It also showed that the activities of total superoxide dismutase (T-SOD) and diamine oxidase (DAO) were upregulated (P < 0.05) in the LCS group. In addition, Bax and caspase 3 immunore-activity increased (P < 0.01), and Bcl-2 immunoreactivity decreased (P < 0.01) in the gastric mucosa of pigs fed the diet with high cysteamine (HCS). The Bax and caspase 3 immunoreactivity decreased (P < 0.01), and Bcl-2 immunoreactivity increased (P < 0.01) in ileum mucosa of pigs fed the HCS diet. CONCLUSIONS: Although moderate dietary coated cysteamine showed positive effects on GI mucosal morphology, apoptosis, and oxidative stress status, the excess coated cysteamine may cause apoptosis leading to GI damage in pigs.


Subject(s)
Apoptosis/drug effects , Cysteamine/pharmacology , Dietary Supplements , Intestinal Mucosa/drug effects , Animal Feed/analysis , Animals , Cysteamine/administration & dosage , Diet/veterinary , Ileum , Intestinal Mucosa/metabolism , Oxidative Stress/drug effects , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL