Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Cell Death Dis ; 15(2): 145, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360744

ABSTRACT

Cancer cells develop multiple strategies to evade T cell-mediated killing. On one hand, cancer cells may preferentially rely on certain amino acids for rapid growth and metastasis. On the other hand, sufficient nutrient availability and uptake are necessary for mounting an effective T cell anti-tumor response in the tumor microenvironment (TME). Here we demonstrate that tumor cells outcompete T cells for cystine uptake due to high Slc7a11 expression. This competition induces T-cell exhaustion and ferroptosis, characterized by diminished memory formation and cytokine secretion, increased PD-1 and TIM-3 expression, as well as intracellular oxidative stress and lipid-peroxide accumulation. Importantly, either Slc7a11 deletion in tumor cells or intratumoral cystine supplementation improves T cell anti-tumor immunity. Mechanistically, cystine deprivation in T cells disrupts glutathione synthesis, but promotes CD36 mediated lipid uptake due to dysregulated cystine/glutamate exchange. Moreover, enforced expression of glutamate-cysteine ligase catalytic subunit (Gclc) promotes glutathione synthesis and prevents CD36 upregulation, thus boosting T cell anti-tumor immunity. Our findings reveal cystine as an intracellular metabolic checkpoint that orchestrates T-cell survival and differentiation, and highlight Gclc as a potential therapeutic target for enhancing T cell anti-tumor function.


Subject(s)
Cystine , Ferroptosis , Cystine/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Glutathione/metabolism , Lipids
2.
Sci Rep ; 13(1): 20961, 2023 11 28.
Article in English | MEDLINE | ID: mdl-38016974

ABSTRACT

Messenger RNA (mRNA) therapies are emerging in different disease areas, but have not yet reached the kidney field. Our aim was to study the feasibility to treat the genetic defect in cystinosis using synthetic mRNA in cell models and ctns-/- zebrafish embryos. Cystinosis is a prototype lysosomal storage disorder caused by mutations in the CTNS gene, encoding the lysosomal cystine-H+ symporter cystinosin, and leading to cystine accumulation in all cells of the body. The kidneys are the first and the most severely affected organs, presenting glomerular and proximal tubular dysfunction, progressing to end-stage kidney failure. The current therapeutic standard cysteamine, reduces cystine levels, but has many side effects and does not restore kidney function. Here, we show that synthetic mRNA can restore lysosomal cystinosin expression following lipofection into CTNS-/- kidney cells and injection into ctns-/- zebrafish. A single CTNS mRNA administration decreases cellular cystine accumulation for up to 14 days in vitro. In the ctns-/- zebrafish, CTNS mRNA therapy improves proximal tubular reabsorption, reduces proteinuria, and restores brush border expression of the multi-ligand receptor megalin. Therefore, this proof-of-principle study takes the first steps in establishing an mRNA-based therapy to restore cystinosin expression, resulting in cystine reduction in vitro and in the ctns-/- larvae, and restoration of the zebrafish pronephros function.


Subject(s)
Amino Acid Transport Systems, Neutral , Cystinosis , Animals , Cystinosis/genetics , Cystinosis/therapy , Cystine/metabolism , Zebrafish/genetics , Zebrafish/metabolism , RNA, Messenger/genetics , RNA, Messenger/therapeutic use , Models, Theoretical , Dietary Supplements , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism
3.
Sci Rep ; 13(1): 17276, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37828038

ABSTRACT

Megalin/LRP2 is a major receptor supporting apical endocytosis in kidney proximal tubular cells. We have previously reported that kidney-specific perinatal ablation of the megalin gene in cystinotic mice, a model of nephropathic cystinosis, essentially blocks renal cystine accumulation and partially preserves kidney tissue integrity. Here, we examined whether inhibition of the megalin pathway in adult cystinotic mice by dietary supplementation (5x-fold vs control regular diet) with the dibasic amino-acids (dAAs), lysine or arginine, both of which are used to treat patients with other rare metabolic disorders, could also decrease renal cystine accumulation and protect cystinotic kidneys. Using surface plasmon resonance, we first showed that both dAAs compete for protein ligand binding to immobilized megalin in a concentration-dependent manner, with identical inhibition curves by L- and D-stereoisomers. In cystinotic mice, 2-month diets with 5x-L-lysine and 5x-L-arginine were overall well tolerated, while 5x-D-lysine induced strong polyuria but no weight loss. All diets induced a marked increase of dAA urinary excretion, most prominent under 5x-D-lysine, without sign of kidney insufficiency. Renal cystine accumulation was slowed down approx. twofold by L-dAAs, and totally suppressed by D-lysine. We conclude that prolonged dietary manipulation of the megalin pathway in kidneys is feasible, tolerable and can be effective in vivo.


Subject(s)
Cystine , Cystinosis , Adult , Humans , Animals , Mice , Cystine/metabolism , Cystinosis/metabolism , Lysine , Low Density Lipoprotein Receptor-Related Protein-2 , Kidney/metabolism , Dietary Supplements
4.
Cell Death Dis ; 14(6): 347, 2023 06 03.
Article in English | MEDLINE | ID: mdl-37268653

ABSTRACT

Studies have indicated dietary restriction of methionine/cystine provided a therapeutic benefit in diseases such as cancer. However, the molecular and cellular mechanisms that underlie the interaction between methionine/cystine restriction (MCR) and effects on esophageal squamous cell carcinoma (ESCC) have remained elusive. Here, we discovered the dietary restriction of methionine/cystine has a large effect on cellular methionine metabolism as assayed in a ECA109 derived xenograft model. RNA-seq and enrichment analysis suggested the blocked tumor progression was affected by ferroptosis, together with the NFκB signaling pathway activation in ESCC. Consistently, GSH content and GPX4 expression were downregulated by MCR both in vivo and in vitro. The contents of Fe2+ and MDA were negatively correlated with supplementary methionine in a dose-dependent way. Mechanistically, MCR and silent of SLC43A2, a methionine transporter, diminished phosphorylation of IKKα/ß and p65. Blocked NFκB signaling pathway further decreased the expression of SLC43A2 and GPX4 in both mRNA and protein level, which in turn downregulated the methionine intake and stimulated ferroptosis, respectively. ESCC progression was inhibited by enhanced ferroptosis and apoptosis and impaired cell proliferation. In this study, we proposed a novel feedback regulation mechanism underlie the correlation between dietary restriction of methionine/cystine and ESCC progression. MCR blocked cancer progression via stimulating ferroptosis through the positive feedback loop between SLC43A2 and NFκB signaling pathways. Our results provided the theoretical basis and new targets for ferroptosis-based clinical antitumor treatments for ESCC patients.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Ferroptosis , Humans , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Cystine/metabolism , Esophageal Neoplasms/pathology , Carcinoma, Squamous Cell/pathology , Methionine/metabolism , Feedback , NF-kappa B/metabolism , Signal Transduction , Cell Proliferation , Racemethionine/metabolism , Racemethionine/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
5.
Mar Biotechnol (NY) ; 25(4): 537-547, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37369882

ABSTRACT

Melanogenesis is a multistep process to produce melanin for dark pigmentation in skin coloration. Previous studies in vertebrates demonstrated that cystine and tyrosine amino acids are involved in the melanin synthesis. However, very little is known about the melanogenesis in bivalve. In this study, cystine supplementation for 30 days significantly upregulated the expression of CgB-aat1, CgCbs and CgTyr and pheomelanin content in the Pacific oyster Crassostrea gigas. Transmission electron microscope (TEM) results revealed more melanosomes in the connective tissue and melanin granules were secreted in epithelium of mantle. In contrast, tyrosine supplementation had no clear effect on melanogenesis except the gene expression changes of CgB-aat1 and CgCbs. In addition, prolonged supplementation of cystine or tyrosine for 60 days had a negative impact on melanogenesis. Indeed, after 60 days, expression of most of the melanin synthesis-related genes under study was decreased, and melanin content was significantly reduced, indicating that cystine and tyrosine might inhibit production of eumelanin and pheomelanin, respectively. In addition, in vitro analysis using primary cell culture from mantle tissue indicated that incubation with cystine, tyrosine, or B-AAT1 polypeptide, CBS/TYR recombinant proteins induced the increase of CgB-aat1 and CgCbs expression in a dose-dependent manner, suggesting the presence of a regulatory network in response to cystine and tyrosine amino acids intakes in pheomelanin synthesis-related gene expression. Taken together, these data indicate that cystine-CgB-aat1-CgCbs-CgTyr axis is a potential regulator of the pheomelanin biosynthesis pathway, and thus plays an important role in the mantle pigmentation in C. gigas. This work provides a new clue for selective cultivation of oyster strains with specific shell colors in bivalve breeding.


Subject(s)
Crassostrea , Tyrosine , Animals , Tyrosine/metabolism , Tyrosine/pharmacology , Melanins/metabolism , Cystine/metabolism , Crassostrea/metabolism , Dietary Supplements
6.
Pediatr Nephrol ; 38(11): 3671-3679, 2023 11.
Article in English | MEDLINE | ID: mdl-37219641

ABSTRACT

BACKGROUND: Nephropathic cystinosis is a rare lysosomal storage disorder in which accumulation of cystine and formation of crystals particularly impair kidney function and gradually lead to multi-organ dysfunction. Lifelong therapy with the aminothiol cysteamine can delay the development of kidney failure and the need for transplant. The purpose of our long-term study was to explore the effects of transitioning from immediate release (IR) to extended release (ER) formulation in Norwegian patients in routine clinical care. METHODS: We retrospectively analysed data on efficacy and safety in 10 paediatric and adult patients. Data were obtained from up to 6 years before and 6 years after transitioning from IR- to ER-cysteamine. RESULTS: Mean white blood cell (WBC) cystine levels remained comparable between the different treatment periods (1.19 versus 1.38 nmol hemicystine/mg protein) although most patients under ER-cysteamine underwent dose reductions. For the non-transplanted patients, the mean estimated glomerular filtration rate (eGFR) change/year was more pronounced during ER-treatment (- 3.39 versus - 6.80 ml/min/1.73 m2/year) possibly influenced by individual events, such as tubulointerstitial nephritis and colitis. Growth measured by Z-height score tended to develop positively. Four of seven patients reported improvement of halitosis, one reported unchanged and two reported worsened symptoms. Most adverse drug reactions (ADRs) were of mild severity. One patient developed two serious ADRs and switched back to IR-formulation. CONCLUSIONS: The results from this long-term retrospective study indicate that switching from IR- to ER-cysteamine was feasible and well tolerated under routine clinical practice. ER-cysteamine allowed satisfactory disease control over the long period considered. A higher resolution version of the Graphical abstract is available as Supplementary information.


Subject(s)
Cystinosis , Fanconi Syndrome , Adult , Humans , Child , Cystinosis/drug therapy , Cysteamine/adverse effects , Retrospective Studies , Cystine/metabolism
7.
Biochem Pharmacol ; 205: 115241, 2022 11.
Article in English | MEDLINE | ID: mdl-36084707

ABSTRACT

The solute carrier family 7 member 11 (SLC7A11), an amino acid transporter protein is frequently overexpressed in human malignancies. The expression and activity of SLC7A11 is finely regulated by oncogenes and tumor suppressors in tumor cells through various mechanisms and is highly specific for cystine and glutamate. Cystine is mainly transported intracellularly by SLC7A11 in the tumor microenvironment (TME) and is involved in GSH synthesis, which leads to ferroptosis resistance in tumor cells and promotes tumorigenesis and progression. The downregulation of SLC7A11 presents a unique drug discovery opportunity for ferroptosis-related diseases. Experimental work has shown that the combination of targeting SLC7A11 and tumor immunotherapy triggers ferroptosis more potently. Moreover, immunotargeting of SLC7A11 increases the chemosensitivity of cancer stem cells to doxorubicin, suggesting that it may act as an adjuvant to chemotherapy. Thus, SLC7A11 could be a promising target to overcome resistance mechanisms in conventional cancer treatments. This review provides an overview of the regulatory network of SLC7A11 in the TME and progress in the development of SLC7A11 inhibitors. In addition, we summarize the cytotoxic effects of blocking SLC7A11 in cancer cells, cancer stem cells and immune cells.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Cystine/metabolism , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Glutamic Acid/metabolism , Neoplasms/drug therapy , Neoplasms/genetics
8.
Nat Commun ; 13(1): 5696, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36171419

ABSTRACT

Fatty liver is a highly heterogenous condition driven by various pathogenic factors in addition to the severity of steatosis. Protein insufficiency has been causally linked to fatty liver with incompletely defined mechanisms. Here we report that fatty liver is a sulfur amino acid insufficient state that promotes metabolic inflexibility via limiting coenzyme A availability. We demonstrate that the nutrient-sensing transcriptional factor EB synergistically stimulates lysosome proteolysis and methionine adenosyltransferase to increase cysteine pool that drives the production of coenzyme A and glutathione, which support metabolic adaptation and antioxidant defense during increased lipid influx. Intriguingly, mice consuming an isocaloric protein-deficient Western diet exhibit selective hepatic cysteine, coenzyme A and glutathione deficiency and acylcarnitine accumulation, which are reversed by cystine supplementation without normalizing dietary protein intake. These findings support a pathogenic link of dysregulated sulfur amino acid metabolism to metabolic inflexibility that underlies both overnutrition and protein malnutrition-associated fatty liver development.


Subject(s)
Amino Acids, Sulfur , Fatty Liver , Amino Acids, Sulfur/metabolism , Animals , Antioxidants/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Coenzyme A/metabolism , Cysteine/metabolism , Cystine/metabolism , Dietary Proteins/metabolism , Fatty Liver/metabolism , Glutathione/metabolism , Homeostasis , Lipids , Liver/metabolism , Methionine/metabolism , Methionine Adenosyltransferase/metabolism , Mice , Oxidation-Reduction
9.
Cells ; 11(10)2022 05 10.
Article in English | MEDLINE | ID: mdl-35626640

ABSTRACT

Ferroptosis, a type of iron-dependent necrotic cell death, is triggered by the accumulation of excessive lipid peroxides in cells. Glutathione (GSH), a tripeptide redox molecule that contains a cysteine (Cys) unit in the center, plays a pivotal role in protection against ferroptosis. When the transsulfuration pathway is activated, the sulfur atom of methionine (Met) is utilized to generate Cys, which can then suppress Cys-starvation-induced ferroptosis. In the current study, we cultured HeLa cells in Met- and/or cystine (an oxidized Cys dimer)- deprived medium and investigated the roles of Met in ferroptosis execution. The results indicate that, in the absence of cystine or Met, ferroptosis or cell cycle arrest, respectively, occurred. Contrary to our expectations, however, the simultaneous deprivation of both Met and cystine failed to induce ferroptosis, although the intracellular levels of Cys and GSH were maintained at low levels. Supplementation with S-adenosylmethionine (SAM), a methyl group donor that is produced during the metabolism of Met, caused the cell cycle progression to resume and lipid peroxidation and the subsequent induction of ferroptosis was also restored under conditions of Met/cystine double deprivation. DNA methylation appeared to be involved in the resumption in the SAM-mediated cell cycle because its downstream metabolite S-adenosylhomocysteine failed to cause either cell cycle progression or ferroptosis to be induced. Taken together, our results suggest that elevated lipid peroxidation products that are produced during cell cycle progression are involved in the execution of ferroptosis under conditions of Cys starvation.


Subject(s)
Ferroptosis , Cell Cycle , Cysteine/metabolism , Cystine/metabolism , Glutathione/metabolism , HeLa Cells , Humans , Methionine/pharmacology , Reactive Oxygen Species/metabolism , S-Adenosylmethionine
10.
Arch Biochem Biophys ; 726: 109157, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35276211

ABSTRACT

Selenium was initially considered a toxic element found in plants growing in soils rich in this element. However, a few years later, selenocysteine was recognized as the 21st amino acid. Huber and Criddle's article has been crucial in discovering selenium-containing proteins and other related works on selenocysteine.


Subject(s)
Selenium , Selenocysteine , Cystine/analogs & derivatives , Cystine/metabolism , Organoselenium Compounds , Selenium/chemistry , Selenocysteine/chemistry , Selenocysteine/metabolism , Sulfur
11.
Eur J Nutr ; 61(5): 2331-2339, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35106632

ABSTRACT

PURPOSE: Although acute prolonged strenuous exercise has been shown to increase markers of gastrointestinal permeability and damage, little is known regarding the efficacy of nutritional supplement interventions on the attenuation of exercise-induced gastrointestinal syndrome. This study addressed the effects of oral amino acid supplementation on markers of gastrointestinal permeability and damage in response to exercise. METHODS: Sixteen active men aged 22.7 ± 2.6 years (mean ± standard deviation) completed placebo or cystine and glutamine supplementation trials in random order. Participants received either a placebo or cystine and glutamine supplements, three times a day for 5 days, separated by a 2-week washout period. On day 6, participants took their designated supplements 30 min before running at a speed corresponding to 75% of maximal oxygen uptake for 1 h, followed by a 4-h rest period. Blood samples were collected pre-exercise, immediately post-exercise, 30 min post-exercise, and 1, 2 and 4 h post-exercise on day 6. The plasma lactulose to mannitol ratio (L:M) and plasma intestinal fatty acid-binding protein (I-FABP) were used as markers of gastrointestinal permeability and damage, respectively. RESULTS: Plasma L:M (linear mixed model, coefficient ± standard error: - 0.011 ± 0.004, P = 0.0090) and changes (i.e., from pre-exercise) in plasma I-FABP (linear mixed model, - 195.3 ± 65.7 coefficient ± standard error (pg/mL), P = 0.0035) were lower in the cystine and glutamine supplementation trial than in the placebo trial. CONCLUSION: Oral cystine and glutamine supplementation attenuated the markers of gastrointestinal permeability and damage after 1 h of strenuous running in young men. TRIAL REGISTRATION NUMBER: UMIN000026008. DATE OF REGISTRATION: 13 December 2018.


Subject(s)
Glutamine , Running , Biomarkers , Cystine/metabolism , Cystine/pharmacology , Dietary Supplements , Gastrointestinal Tract/metabolism , Glutamine/pharmacology , Humans , Male , Permeability , Running/physiology , Young Adult
12.
Mol Oncol ; 16(9): 1777-1794, 2022 05.
Article in English | MEDLINE | ID: mdl-34856072

ABSTRACT

Nutritional intervention is becoming more prevalent as adjuvant therapy for many cancers in view of the tumor dependence on external sources for some nutrients. However, little is known about the mechanisms that make cancer cells require certain nutrients from the microenvironment. Herein, we report the dependence of glioma cells on exogenous cysteine/cystine, despite this amino acid being nonessential. Using several 13 C-tracers and analysis of cystathionine synthase and cystathioninase levels, we revealed that glioma cells were not able to support glutathione synthesis through the transsulfuration pathway, which allows methionine to be converted to cysteine in cysteine/cystine-deprived conditions. Therefore, we explored the nutritional deprivation in a mouse model of glioma. Animals subjected to a cysteine/cystine-free diet survived longer, although this increase did not attain statistical significance, with concomitant reductions in plasma glutathione and cysteine levels. At the end point, however, tumors displayed the ability to synthesize glutathione, even though higher levels of oxidative stress were detected. We observed a compensation from the nutritional intervention revealed as the recovery of cysteine-related metabolite levels in plasma. Our study highlights a time window where cysteine deprivation can be exploited for additional therapeutic strategies.


Subject(s)
Cysteine , Glioma , Animals , Cell Proliferation , Cysteine/metabolism , Cystine/metabolism , Glutathione/metabolism , Humans , Mice , Tumor Microenvironment
13.
Theriogenology ; 167: 24-31, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33743505

ABSTRACT

Sperm are redox-regulated cells, and deregulation of their redox status is considered to affect male fertility and to reduce their fertilizing ability following biotechnological procedures, such as cryopreservation. Cystine (CysS), after incorporation in sperm via SLC7A11 antiporter, has been demonstrated to increase intracellular GSH content, the most important non enzymatic antioxidant. This study was aimed at investigating the role of SLC7A11 antiporter on frozen-thawed stallion sperm ability to respond to in vitro capacitating environment after post-thaw incubation with CysS and/or Sulfasalazine (SS), a specific inhibitor of SLC7A11 antiporter. Viability, motility, immunolocalization of tyrosine phosphorylated proteins and the ability to bind to heterologous zonae pellucidae were evaluated. Thawed sperm from seven stallions (2 ejaculates/stallion) was washed and resuspended in Tyrodes media; each thawed ejaculate was divided in Control (CTR) and 3 samples supplemented with: 0.5 mM Cystine (CysS), 500 µM Sulfasalazine (SS) and 0.5 mM CysS + 500 µM SS (CysS + SS). After 1 h of incubation at 37 °C, samples were washed twice, resuspended in capacitating BWW medium and incubated at 38 °C under 5% CO2. After 30 and 60 min, sperm motility, viability and tyrosine phosphorylated protein immunolocalization, used as capacitation status index, were evaluated. After 30 min of capacitation, 4 × 105 sperm were co-incubated with denuded pig oocytes in capacitation medium for 30 min for the heterologous binding assay. None of the sperm parameters studied (motility, viability and tyrosine phosphorylation) showed any difference respective to control. The number of sperm bound per oocyte (mean ± SEM) tended to increase in CysS group (44.0 ± 12.3) respect CTR (40.8 ± 10.8) while decreased in SS group (32.4 ± 7.8) (p < 0.01). Moreover, CysS + SS group showed a lower binding rate (32.0 ± 10.0) compared to CysS (p < 0.001). Our results suggest that CysS supplementation of thawed stallion sperm can influence their ability to bind to heterologous zona pellucidae as the inhibition of CysS incorporation by SLC7A11 reduced the number of sperm bound per oocyte. This effect does not seem to be ascribed to a modification of sperm motility, membrane integrity and tyrosine phosphorylation.


Subject(s)
Amino Acid Transport System y+/antagonists & inhibitors , Semen Preservation , Animals , Antiporters , Cryopreservation/veterinary , Cystine/metabolism , Glutamic Acid , Horses , Male , Semen Preservation/veterinary , Sperm Motility , Spermatozoa/metabolism , Swine
14.
Protein Cell ; 12(8): 599-620, 2021 08.
Article in English | MEDLINE | ID: mdl-33000412

ABSTRACT

The cystine/glutamate antiporter SLC7A11 (also commonly known as xCT) functions to import cystine for glutathione biosynthesis and antioxidant defense and is overexpressed in multiple human cancers. Recent studies revealed that SLC7A11 overexpression promotes tumor growth partly through suppressing ferroptosis, a form of regulated cell death induced by excessive lipid peroxidation. However, cancer cells with high expression of SLC7A11 (SLC7A11high) also have to endure the significant cost associated with SLC7A11-mediated metabolic reprogramming, leading to glucose- and glutamine-dependency in SLC7A11high cancer cells, which presents potential metabolic vulnerabilities for therapeutic targeting in SLC7A11high cancer. In this review, we summarize diverse regulatory mechanisms of SLC7A11 in cancer, discuss ferroptosis-dependent and -independent functions of SLC7A11 in promoting tumor development, explore the mechanistic basis of SLC7A11-induced nutrient dependency in cancer cells, and conceptualize therapeutic strategies to target SLC7A11 in cancer treatment. This review will provide the foundation for further understanding SLC7A11 in ferroptosis, nutrient dependency, and tumor biology and for developing novel effective cancer therapies.


Subject(s)
Amino Acid Transport System y+/genetics , Antineoplastic Agents/therapeutic use , Cystine/metabolism , Ferroptosis/drug effects , Neoplasms/drug therapy , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/metabolism , Cystine/antagonists & inhibitors , DNA Methylation/drug effects , Ferroptosis/genetics , Gene Expression Regulation, Neoplastic , Glucose/antagonists & inhibitors , Glucose/metabolism , Glutamine/antagonists & inhibitors , Glutamine/metabolism , Glutathione/antagonists & inhibitors , Glutathione/metabolism , Histones/genetics , Histones/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Piperazines/therapeutic use , Signal Transduction , Sorafenib/therapeutic use , Sulfasalazine/therapeutic use
15.
Arch Anim Nutr ; 74(5): 414-427, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32840134

ABSTRACT

The objective of this study was to estimate the sulphur amino acid (methionine + cystine) requirements and nitrogen endogenous losses in kittens aged 150 to 240 d. Thirty-six cats were distributed in six treatments (six cats per treatment) consisting of different concentrations of methionine + cystine (M + C): T1, 6.5 g/kg; T2, 8.8 g/kg; T3, 11.3 g/kg; T4, 13.6 g/kg; T5, 16.0 g/kg; and control, 6.5 g/kg. Diets were formulated by serial dilution of T5 (a diet relatively deficient in M + C but containing high protein concentrations) with a minimal nitrogen diet (MND). Thus, crude protein and amino acid concentrations in diets T1-T5 decreased by the same factor. The control diet was the T1 diet supplemented with adequate concentrations of M + C (6.5 g/kg; 8.8 g/kg; 11.3 g/kg; 13.6 g/kg and 16.0 g/kg). All diets were based on ingredients commonly used in extruded cat diets. Digestibility assays were performed for the determination of nitrogen balance. Nitrogen intake (NI) and nitrogen excretion (NEX) results data were fitted with an exponential equation to estimate nitrogen maintenance requirement (NMR), theoretical maximum for daily nitrogen retention (NRmaxT), and protein quality (b). M + C requirements were calculated from the limiting amino acid intake (LAAI) equation assuming a nitrogen retention of 45 to 65% NRmaxT. The NMR of kittens aged 150, 195, and 240 d was estimated at 595, 559, and 455 mg/kg body weight (BW)0.67 per day, respectively, and M + C requirements were estimated at 517, 664, and 301 mg/kg BW0.67 per day, respectively.


Subject(s)
Cats/metabolism , Cystine/metabolism , Methionine/metabolism , Nitrogen/metabolism , Animal Feed/analysis , Animal Nutritional Physiological Phenomena/drug effects , Animals , Cystine/administration & dosage , Diet/veterinary , Dietary Supplements/analysis , Female , Male , Methionine/administration & dosage
16.
Nutrients ; 12(6)2020 Jun 21.
Article in English | MEDLINE | ID: mdl-32575864

ABSTRACT

The ESPGHAN/ESPEN/ESPR-Guidelines on pediatric parenteral nutrition (PPN) recommend the administration of the semiessential amino acid (AA) cysteine to preterm neonates due to their biochemical immaturity resulting in an inability to sufficiently synthetize endogenous cysteine. The soluble precursor N-acetylcysteine (NAC) is easily converted into bioavailable cysteine. Its dimer N,N-diacetylcystine (DAC) is almost unconvertable to cysteine when given intravenously resulting in a diminished bioavailability of cysteine. This study aims to understand the triggers and oxidation process of NAC to DAC to evaluate possibilities of reducing DAC formation in standardized PPN. Therefore, different air volumes (21% O2) were injected into the AA compartment of a standardized dual-chamber PPN. O2 concentrations were measured in the AA solution and the headspaces of the primary and secondary packaging. NAC and DAC concentrations were analyzed simultaneously. The analysis showed that O2 is principally delivered from the primary headspace. NAC oxidation exclusively delivers DAC, depending on the O2 amount in the solution and the headspaces. The reaction of NAC to DAC being containable by limiting the O2 concentration, the primary headspace must be minimized during manufacturing, and oxygen absorbers must be added into the secondary packaging for a long-term storage of semipermeable containers.


Subject(s)
Acetylcysteine/administration & dosage , Amino Acids/administration & dosage , Cystine/analogs & derivatives , Drug Stability , Infant, Premature , Parenteral Nutrition Solutions/chemistry , Parenteral Nutrition , Acetylcysteine/metabolism , Amino Acids/metabolism , Biological Availability , Cysteine/administration & dosage , Cysteine/metabolism , Cystine/metabolism , Drug Industry , Drug Storage , Humans , Infant Nutritional Physiological Phenomena , Infant, Newborn , Nutritional Requirements , Oxidation-Reduction , Oxygen
17.
Pediatrics ; 145(5)2020 05.
Article in English | MEDLINE | ID: mdl-32245805

ABSTRACT

Cystinuria is an autosomal recessive disorder characterized by excessive urinary excretion of cystine, resulting in recurrent cystine kidney stones, often presenting in childhood. Current treatment options for cystinuria include dietary and/or fluid measures and potassium citrate to reduce cystine excretion and/or increase solubility. Tiopronin and D-penicillamine are used in refractory cases to bind cystine in urine, albeit with serious side effects. A recent study revealed efficacy of nutritional supplement α-lipoic acid (ALA) treatment in preventing kidney stones in a mouse model of cystinuria. Here, we report 2 pediatric patients (6 and 15 years old) with cystinuria who received regular doses of ALA in addition to conventional therapy with potassium citrate. Both patients tolerated ALA without any adverse effects and had reduced frequency of symptomatic and asymptomatic kidney stones with disappearance of existing kidney stones in 1 patient after 2 months of ALA therapy. ALA treatment markedly improved laboratory markers of cystine solubility in urine with increased cystine capacity (-223 to -1 mg/L in patient 1 and +140 to +272 mg/L in patient 2) and decreased cystine supersaturation (1.7 to 0.88 in patient 1 and 0.64 to 0.48 in patient 2) without any changes in cystine excretion or urine pH. Our findings suggest that ALA improves solubility of cystine in urine and prevents stone formation in patients with cystinuria who do not respond to diet and citrate therapy.


Subject(s)
Antioxidants/therapeutic use , Cystine/metabolism , Cystinuria/drug therapy , Cystinuria/urine , Thioctic Acid/therapeutic use , Adolescent , Child , Female , Humans
18.
Infect Immun ; 88(3)2020 02 20.
Article in English | MEDLINE | ID: mdl-31843961

ABSTRACT

Staphylococcus aureus is a significant human pathogen due to its capacity to cause a multitude of diseases. As such, S. aureus efficiently pillages vital nutrients from the host; however, the molecular mechanisms that support sulfur acquisition during infection have not been established. One of the most abundant extracellular sulfur-containing metabolites within the host is cysteine, which acts as the major redox buffer in the blood by transitioning between reduced and oxidized (cystine) forms. We therefore hypothesized that S. aureus acquires host-derived cysteine and cystine as sources of nutrient sulfur during systemic infection. To test this hypothesis, we used the toxic cystine analogue selenocystine to initially characterize S. aureus homologues of the Bacillus subtilis cystine transporters TcyABC and TcyP. We found that genetic inactivation of both TcyA and TcyP induced selenocystine resistance. The double mutant also failed to proliferate in medium supplemented with cystine, cysteine, or N-acetyl cysteine as the sole sulfur source. However, only TcyABC was necessary for proliferation in defined medium containing homocystine as the sulfur source. Using a murine model of systemic infection, we observed tcyP-dependent competitive defects in the liver and heart, indicating that this sulfur acquisition strategy supports proliferation of S. aureus in these organs. Phylogenetic analyses identified TcyP homologues in many pathogenic species, implying that this sulfur procurement strategy is conserved. In total, this study is the first to experimentally validate sulfur acquisition systems in S. aureus and establish their importance during pathogenesis.


Subject(s)
Cystine/metabolism , Membrane Transport Proteins/physiology , Staphylococcal Infections/metabolism , Staphylococcus aureus/physiology , Sulfur/metabolism , Animals , Mice
19.
J Anim Physiol Anim Nutr (Berl) ; 104(1): 224-229, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31709659

ABSTRACT

The aim of this study was to develop a method to determine the efficiency of utilization of Met and Cys using stable isotopes in order to reduce the number of sacrificed animals relative to the comparative slaughter technique. Met and Cys efficiencies were obtained separately and as total SAA values. Twenty-one 14- to 28-day-old broiler chickens were fed experimental diets containing different Met:Cys ratios (44:56, 50:50 and 56:44). Birds were given diets with daily supplements of L-(15 N) Met (60 mmol/kg) or L-(15 N2 ) Cys (35 mmol/kg) throughout the entire experimental period. Excreta were collected daily, and birds were euthanized at the end of the trial to collect feather-free bodies and feathers. Samples were analysed for 15 N and 15 N-Met content. The utilization efficiency for Met, Cys and Met + Cys for feather-free bodies was 55%, 75%, and 60%, while the efficiencies for feathers were estimated at 96%, 77% and 84% respectively.


Subject(s)
Chickens/metabolism , Cystine/metabolism , Methionine/metabolism , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Cystine/administration & dosage , Diet/veterinary , Dietary Supplements , Feathers , Isotope Labeling , Methionine/administration & dosage , Nitrogen Isotopes , Nutritional Requirements
20.
ACS Appl Mater Interfaces ; 11(30): 26722-26730, 2019 Jul 31.
Article in English | MEDLINE | ID: mdl-31276364

ABSTRACT

An effective steady-state redox balance is maintained in cancer cells, allowing for protection against oxidative stress and thereby enhancing cell proliferation and tumor growth. Disruption of this redox balance would increase the cellular content of reactive oxygen species (ROS) and potentiate oxidative stress-induced cell death in tumor cells, thus representing an effective strategy for cancer treatment. Glutathione (GSH) is a major reducing agent, and its cellular levels are determined at least partly by the availability of cysteine via xCT (SLC7A11)-mediated entry of cystine into cells. We developed a nanoplatform using ZnO nanoparticles (NPs) as a carrier, loaded with salicylazosulfapyridine (SASP), and stabilized with DSPE-PEG, to form ultra-small NPs (SASP/ZnO NPs). The goal of this NP strategy is to disrupt the redox balance in cells by two mechanisms: increased generation of ROS and decreased synthesis of GSH. Such an approach would be effective in killing tumor cells. As expected, the SASP/ZnO NPs enhanced ROS production because of ZnO and impaired GSH synthesis because of SASP-induced inhibition of xCT (SLC7A11) transport function. As a consequence, treatment of tumor cells with SASP/ZnO NPs in vitro and in vivo resulted in a synergistic disruptive effect on redox balance in tumor cells and induced cell death and decreased tumor growth. This ambidextrous approach has potential in cancer therapy by combining two complementary pathways to disrupt the redox balance in tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , Nanoparticles/chemistry , Neoplasms/drug therapy , Oxidative Stress/drug effects , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cysteine/chemistry , Cystine/metabolism , Glutathione/metabolism , Humans , Nanoparticles/administration & dosage , Neoplasms/pathology , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Sulfasalazine/chemistry , Sulfasalazine/pharmacology , Zinc Oxide/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL