Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 920
Filter
1.
Redox Biol ; 73: 103222, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38843767

ABSTRACT

BACKGROUND: Cystathionine ß-synthase (CBS)-deficient homocystinuria (HCU) is an inherited disorder of sulfur amino acid metabolism with varying severity and organ complications, and a limited knowledge about underlying pathophysiological processes. Here we aimed at getting an in-depth insight into disease mechanisms using a transgenic mouse model of HCU (I278T). METHODS: We assessed metabolic, proteomic and sphingolipidomic changes, and mitochondrial function in tissues and body fluids of I278T mice and WT controls. Furthermore, we evaluated the efficacy of methionine-restricted diet (MRD) in I278T mice. RESULTS: In WT mice, we observed a distinct tissue/body fluid compartmentalization of metabolites with up to six-orders of magnitude differences in concentrations among various organs. The I278T mice exhibited the anticipated metabolic imbalance with signs of an increased production of hydrogen sulfide and disturbed persulfidation of free aminothiols. HCU resulted in a significant dysregulation of liver proteome affecting biological oxidations, conjugation of compounds, and metabolism of amino acids, vitamins, cofactors and lipids. Liver sphingolipidomics indicated upregulation of the pro-proliferative sphingosine-1-phosphate signaling pathway. Liver mitochondrial function of HCU mice did not seem to be impaired compared to controls. MRD in I278T mice improved metabolic balance in all tissues and substantially reduced dysregulation of liver proteome. CONCLUSION: The study highlights distinct tissue compartmentalization of sulfur-related metabolites in normal mice, extensive metabolome, proteome and sphingolipidome disruptions in I278T mice, and the efficacy of MRD to alleviate some of the HCU-related biochemical abnormalities.


Subject(s)
Cystathionine beta-Synthase , Disease Models, Animal , Homocystinuria , Liver , Metabolomics , Mice, Transgenic , Proteomics , Sphingolipids , Animals , Mice , Homocystinuria/metabolism , Homocystinuria/genetics , Proteomics/methods , Cystathionine beta-Synthase/metabolism , Cystathionine beta-Synthase/deficiency , Cystathionine beta-Synthase/genetics , Liver/metabolism , Metabolomics/methods , Sphingolipids/metabolism , Mitochondria/metabolism , Lipidomics/methods , Proteome/metabolism
2.
Biochem J ; 481(8): 569-585, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38563463

ABSTRACT

Homocystinuria is a rare disease caused by mutations in the CBS gene that results in a deficiency of cystathionine ß-synthase (CBS). CBS is an essential pyridoxal 5'-phosphate (PLP)-dependent enzyme in the transsulfuration pathway, responsible for combining serine with homocysteine to produce cystathionine, whose activity is enhanced by the allosteric regulator S-adenosylmethionine (SAM). CBS also plays a role in generating hydrogen sulfide (H2S), a gaseous signaling molecule with diverse regulatory functions within the vascular, nervous, and immune systems. In this study, we present the clinical and biochemical characterization of two novel CBS missense mutations that do not respond to pyridoxine treatment, namely c.689T > A (L230Q) and 215A > T (K72I), identified in a Chinese patient. We observed that the disease-associated K72I genetic variant had no apparent effects on the spectroscopic and catalytic properties of the full-length enzyme. In contrast, the L230Q variant expressed in Escherichia coli did not fully retain heme and when compared with the wild-type enzyme, it exhibited more significant impairments in both the canonical cystathionine-synthesis and the alternative H2S-producing reactions. This reduced activity is consistent with both in vitro and in silico evidence, which indicates that the L230Q mutation significantly decreases the overall protein's stability, which in turn, may represent the underlying cause of its pathogenicity.


Subject(s)
Cystathionine beta-Synthase , Homocystinuria , Mutation, Missense , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/metabolism , Homocystinuria/genetics , Homocystinuria/metabolism , Homocystinuria/enzymology , Humans , Male , Female
3.
Plant J ; 118(6): 2154-2168, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38558071

ABSTRACT

Verticillium wilt (VW) is a devasting disease affecting various plants, including upland cotton, a crucial fiber crop. Despite its impact, the genetic basis underlying cotton's susceptibility or defense against VW remains unclear. Here, we conducted a genome-wide association study on VW phenotyping in upland cotton and identified a locus on A13 that is significantly associated with VW resistance. We then identified a cystathionine ß-synthase domain gene at A13 locus, GhCBSX3A, which was induced by Verticillium dahliae. Functional analysis, including expression silencing in cotton and overexpression in Arabidopsis thaliana, confirmed that GhCBSX3A is a causal gene at the A13 locus, enhancing SAR-RBOHs-mediated apoplastic oxidative burst. We found allelic variation on the TATA-box of GhCBSX3A promoter attenuated its expression in upland cotton, thereby weakening VW resistance. Interestingly, we discovered that altered artificial selection of GhCBSX3A_R (an elite allele for VW) under different VW pressures during domestication and other improved processes allows specific human needs to be met. Our findings underscore the importance of GhCBSX3A in response to VW, and we propose a model for defense-associated genes being selected depending on the pathogen's pressure. The identified locus and gene serve as promising targets for VW resistance enhancement in cotton through genetic engineering.


Subject(s)
Ascomycota , Disease Resistance , Gossypium , Plant Diseases , Plant Proteins , Gossypium/genetics , Gossypium/microbiology , Gossypium/immunology , Gossypium/metabolism , Disease Resistance/genetics , Plant Diseases/microbiology , Plant Diseases/immunology , Plant Diseases/genetics , Ascomycota/physiology , Plant Proteins/genetics , Plant Proteins/metabolism , Genome-Wide Association Study , Respiratory Burst , Gene Expression Regulation, Plant , Arabidopsis/genetics , Arabidopsis/microbiology , Arabidopsis/immunology , Arabidopsis/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Plants, Genetically Modified , Verticillium
4.
Clin Exp Hypertens ; 46(1): 2328147, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38488417

ABSTRACT

BACKGROUND: Several studies indicate that the cystathionine ß-synthase (CBS) gene T833C, G919A and 844ins68 polymorphisms in the 8th exon region may be correlated with coronary artery disease (CAD) susceptibility, but the results have been inconsistent and inconclusive. Thus, a meta-analysis was conducted to provide a comprehensive estimate of these associations. METHODS: On the basis of searches in the PubMed, EMBASE, Cochrane Library, Wanfang, VIP, and CNKI databases, we selected 14 case - control studies including 2123 cases and 2368 controls for this meta-analysis. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were calculated accordingly using a fixed-effect or random-effect model. RESULTS: The results indicated an increased risk between the CBS T833C gene polymorphisms and susceptibility to CAD under the dominant model (CC+CT vs. TT: OR = 1.92, 95% CI: 1.11 ~ 3.32), recessive model (CC vs. CT+TT: OR = 1.88, 95% CI: 1.17 ~ 3.03), and homozygous model (CC vs. TT: OR = 2.46, 95% CI: 1.04 ~ 5.83). In these three genetic models, no significant association was identified for CBS G919A (AA+AG vs. GG: OR = 1.48, 95% CI: 0.45 ~ 4.82),(AA vs. AG+GG: OR = 1.58, 95% CI: 0.93 ~ 2.70),(AA vs. GG: OR = 1.66, 95% CI: 0.40 ~ 6.92) or CBS 844ins68 (II+ID vs. DD: OR = 1.04, 95% CI: 0.80 ~ 1.35),(II vs. ID+DD: OR = 1.09, 95% CI: 0.51 ~ 2.36),(II vs. DD: OR = 1.10, 95% CI: 0.51 ~ 2.39). CONCLUSIONS: This meta-analysis suggests that the CBS T833C gene polymorphism is significantly associated with the risk of CAD and it shows a stronger association in Asian populations. Individuals with the C allele of the CBS gene T833C polymorphism might be particularly susceptible to CAD.


Subject(s)
Coronary Artery Disease , Humans , Coronary Artery Disease/genetics , Cystathionine beta-Synthase/genetics , Polymorphism, Genetic , Homozygote , Exons/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide/genetics
5.
Exp Cell Res ; 437(1): 114007, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38499142

ABSTRACT

Gastric cancer metastasis is a major cause of poor prognosis. Our previous research showed that methionine restriction (MR) lowers the invasiveness and motility of gastric carcinoma. In this study, we investigated the particular mechanisms of MR on gastric carcinoma metastasis. In vitro, gastric carcinoma cells (AGS, SNU-5, MKN7, KATO III, SNU-1, and MKN45) were grown in an MR medium for 24 h. In vivo, BALB/c mice were given a methionine-free (Met-) diet. Transwell assays were used to investigate cell invasion and migration. The amounts of Krüppel like factor 10 (KLF10) and cystathionine ß-synthase (CBS) were determined using quantitative real-time PCR and Western blot. To determine the relationship between KLF10 and CBS, chromatin immunoprecipitation and a dual-luciferase reporter experiment were used. Hematoxylin-eosin staining was used to detect lung metastasis. Liquid chromatography-mass spectrometry was used to determine cystathionine content. MR therapy had varying effects on the invasion and migration of gastric carcinoma cells AGS, SNU-5, MKN7, KATO III, SNU-1, and MKN45. KLF10 was highly expressed in AGS cells but poorly expressed in KATO III cells. KLF10 improved MR's ability to prevent gastric carcinoma cell invasion and migration. In addition, KLF10 may interact with CBS, facilitating transcription. Further detection revealed that inhibiting the KLF10/CBS-mediated trans-sulfur pathway lowered Met-'s inhibitory effect on lung metastasis development. KLF10 transcription activated CBS, accelerated the trans-sulfur pathway, and increased gastric carcinoma cells' susceptibility to MR.


Subject(s)
Carcinoma , Lung Neoplasms , Stomach Neoplasms , Mice , Animals , Methionine/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Stomach Neoplasms/pathology , Racemethionine , Sulfur , Lung Neoplasms/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Early Growth Response Transcription Factors/metabolism
6.
Commun Biol ; 7(1): 9, 2024 01 03.
Article in English | MEDLINE | ID: mdl-38172561

ABSTRACT

There are limited therapeutic options for patients with advanced prostate cancer (PCa). We previously found that heat shock factor 1 (HSF1) expression is increased in PCa and is an actionable target. In this manuscript, we identify that HSF1 regulates the conversion of homocysteine to cystathionine in the transsulfuration pathway by altering levels of cystathionine-ß-synthase (CBS). We find that HSF1 directly binds the CBS gene and upregulates CBS mRNA levels. Targeting CBS decreases PCa growth and induces tumor cell death while benign prostate cells are largely unaffected. Combined inhibition of HSF1 and CBS results in more pronounced inhibition of PCa cell proliferation and reduction of transsulfuration pathway metabolites. Combination of HSF1 and CBS knockout decreases tumor size for a small cell PCa xenograft mouse model. Our study thus provides new insights into the molecular mechanism of HSF1 function and an effective therapeutic strategy against advanced PCa.


Subject(s)
Cystathionine , Prostatic Neoplasms , Male , Humans , Mice , Animals , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cell Proliferation , Prostatic Neoplasms/genetics , Heat-Shock Response
7.
Mol Cell Biochem ; 479(4): 825-829, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37198322

ABSTRACT

One in 700 children is born with the down syndrome (DS). In DS, there is an extra copy of X chromosome 21 (trisomy). Interestingly, the chromosome 21 also contains an extra copy of the cystathionine beta synthase (CBS) gene. The CBS activity is known to contribute in mitochondrial sulfur metabolism via trans-sulfuration pathway. We hypothesize that due to an extra copy of the CBS gene there is hyper trans-sulfuration in DS. We believe that understanding the mechanism of hyper trans-sulfuration during DS will be important in improving the quality of DS patients and towards developing new treatment strategies. We know that folic acid "1-carbon" metabolism (FOCM) cycle transfers the "1-carbon" methyl group to DNA (H3K4) via conversion of s-adenosyl methionine (SAM) to s-adenosyl homocysteine (SAH) by DNMTs (the gene writers). The demethylation reaction is carried out by ten-eleven translocation methylcytosine dioxygenases (TETs; the gene erasers) through epigenetics thus turning the genes off/on and opening the chromatin by altering the acetylation/HDAC ratio. The S-adenosyl homocysteine hydrolase (SAHH) hydrolyzes SAH to homocysteine (Hcy) and adenosine. The Hcy is converted to cystathionine, cysteine and hydrogen sulfide (H2S) via CBS/cystathioneγ lyase (CSE)/3-mercaptopyruvate sulfurtransferase (3MST) pathways. Adenosine by deaminase is converted to inosine and then to uric acid. All these molecules remain high in DS patients. H2S is a potent inhibitor of mitochondrial complexes I-IV, and regulated by UCP1. Therefore, decreased UCP1 levels and ATP production can ensue in DS subjects. Interestingly, children born with DS show elevated levels of CBS/CSE/3MST/Superoxide dismutase (SOD)/cystathionine/cysteine/H2S. We opine that increased levels of epigenetic gene writers (DNMTs) and decreased in gene erasers (TETs) activity cause folic acid exhaustion, leading to an increase in trans-sulphuration by CBS/CSE/3MST/SOD pathways. Thus, it is important to determine whether SIRT3 (inhibitor of HDAC3) can decrease the trans-sulfuration activity in DS patients. Since there is an increase in H3K4 and HDAC3 via epigenetics in DS, we propose that sirtuin-3 (Sirt3) may decrease H3K4 and HDAC3 and hence may be able to decrease the trans-sulfuration in DS. It would be worth to determine whether the lactobacillus, a folic acid producing probiotic, mitigates hyper-trans-sulphuration pathway in DS subjects. Further, as we know that in DS patients the folic acid is exhausted due to increase in CBS, Hcy and re-methylation. In this context, we suggest that folic acid producing probiotics such as lactobacillus might be able to improve re-methylation process and hence may help decrease the trans-sulfuration pathway in the DS patients.


Subject(s)
Down Syndrome , Hydrogen Sulfide , Kidney Diseases , Sirtuin 3 , Child , Humans , Cystathionine/genetics , Cystathionine/metabolism , Down Syndrome/genetics , Trisomy , Cysteine , Sirtuin 3/genetics , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Hydrogen Sulfide/metabolism , S-Adenosylmethionine , Superoxide Dismutase/metabolism , Adenosine , Kidney Diseases/metabolism , Folic Acid , Homocysteine , Carbon , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism
8.
Free Radic Biol Med ; 210: 13-24, 2024 01.
Article in English | MEDLINE | ID: mdl-37951283

ABSTRACT

Cystathionine-ß-synthase (CBS) catalyzes the first step of the transsulfuration pathway. The role of host-derived CBS in Staphylococcus aureus (S. aureus)-induced udder infection remains elusive. Herein, we report that S. aureus infection enhances the expression of CBS in mammary epithelial cells in vitro and in vivo. A negative correlation is present between the expression of CBS and inflammation after employing a pharmacological inhibitor/agonist of CBS. In addition, CBS achieves a fine balance between eliciting sufficient protective innate immunity and preventing excessive damage to cells and tissues preserving the integrity of the blood-milk barrier (BMB). CBS/H2S reduces bacterial load by promoting the generation of antibacterial substances (ROS, RNS) and inhibiting apoptosis, as opposed to relying solely on intense inflammatory reactions. Conversely, H2S donor alleviate inflammation via S-sulfhydrating HuR. Finally, CBS/H2S promotes the expression of Abcb1b, which in turn strengthens the integrity of the BMB. The study described herein demonstrates the importance of CBS in regulating the mammary immune response to S. aureus. Increased CBS in udder tissue modulates excessive inflammation, which suggests a novel target for drug development in the battle against S. aureus and other infections.


Subject(s)
Cystathionine beta-Synthase , Hydrogen Sulfide , Animals , Humans , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Staphylococcus aureus/metabolism , Cystathionine , Mammary Glands, Animal/metabolism , Inflammation , Hydrogen Sulfide/metabolism
9.
Mol Cell Biol ; 43(12): 664-674, 2023.
Article in English | MEDLINE | ID: mdl-38051092

ABSTRACT

Homocystinuria (HCU), an inherited metabolic disorder caused by lack of cystathionine beta-synthase (CBS) activity, is chiefly caused by misfolding of single amino acid residue missense pathogenic variants. Previous studies showed that chemical, pharmacological chaperones or proteasome inhibitors could rescue function of multiple pathogenic CBS variants; however, the underlying mechanisms remain poorly understood. Using Chinese hamster DON fibroblasts devoid of CBS and stably overexpressing human WT or mutant CBS, we showed that expression of pathogenic CBS variant mostly dysregulates gene expression of small heat shock proteins HSPB3 and HSPB8 and members of HSP40 family. Endoplasmic reticulum stress sensor BiP was found upregulated with CBS I278T variant associated with proteasomes suggesting proteotoxic stress and degradation of misfolded CBS. Co-expression of the main effector HSP70 or master regulator HSF1 rescued steady-state levels of CBS I278T and R125Q variants with partial functional rescue of the latter. Pharmacological proteostasis modulators partially rescued expression and activity of CBS R125Q likely due to reduced proteotoxic stress as indicated by decreased BiP levels and promotion of refolding as indicated by induction of HSP70. In conclusion, targeted manipulation of cellular proteostasis may represent a viable therapeutic approach for the permissive pathogenic CBS variants causing HCU.


Subject(s)
Cystathionine beta-Synthase , Homocystinuria , Humans , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/metabolism , Homocystinuria/drug therapy , Homocystinuria/genetics , Homocystinuria/metabolism , Cystathionine/metabolism , Cystathionine/therapeutic use , Proteostasis , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism
10.
Int J Mol Sci ; 24(24)2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38138989

ABSTRACT

Regulatory adenine nucleotide-binding cystathionine ß-synthase (CBS) domains are widespread in proteins; however, information on the mechanism of their modulating effects on protein function is scarce. The difficulty in obtaining structural data for such proteins is ascribed to their unusual flexibility and propensity to form higher-order oligomeric structures. In this study, we deleted the most movable domain from the catalytic part of a CBS domain-containing bacterial inorganic pyrophosphatase (CBS-PPase) and characterized the deletion variant both structurally and functionally. The truncated CBS-PPase was inactive but retained the homotetrameric structure of the full-size enzyme and its ability to bind a fluorescent AMP analog (inhibitor) and diadenosine tetraphosphate (activator) with the same or greater affinity. The deletion stabilized the protein structure against thermal unfolding, suggesting that the deleted domain destabilizes the structure in the full-size protein. A "linear" 3D structure with an unusual type of domain swapping predicted for the truncated CBS-PPase by Alphafold2 was confirmed by single-particle electron microscopy. The results suggest a dual role for the CBS domains in CBS-PPase regulation: they allow for enzyme tetramerization, which impedes the motion of one catalytic domain, and bind adenine nucleotides to mitigate or aggravate this effect.


Subject(s)
Cystathionine beta-Synthase , Pyrophosphatases , Pyrophosphatases/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Catalytic Domain , Bacterial Proteins/metabolism , Nucleotides
11.
Redox Biol ; 68: 102958, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37948927

ABSTRACT

Astrocytic dysfunction is central to age-related neurodegenerative diseases. However, the mechanisms leading to astrocytic dysfunction are not well understood. We identify that among the diverse cellular constituents of the brain, murine and human astrocytes are enriched in the expression of CBS. Depleting CBS in astrocytes causes mitochondrial dysfunction, increases the production of reactive oxygen species (ROS) and decreases cellular bioenergetics that can be partially rescued by exogenous H2S supplementation or by re-expressing CBS. Conversely, the CBS/H2S axis, associated protein persulfidation and proliferation are decreased in astrocytes upon oxidative stress which can be rescued by exogenous H2S supplementation. Here we reveal that in the aging brain, the CBS/H2S axis is downregulated leading to decreased protein persulfidation, together augmenting oxidative stress. Our findings uncover an important protective role of the CBS/H2S axis in astrocytes that may be disrupted in the aged brain.


Subject(s)
Aging , Astrocytes , Brain , Cystathionine beta-Synthase , Aged , Animals , Humans , Mice , Aging/metabolism , Aging/pathology , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Cystathionine/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism
12.
Eur J Med Res ; 28(1): 540, 2023 Nov 25.
Article in English | MEDLINE | ID: mdl-38007457

ABSTRACT

Activating transcription factor 6 (ATF6) is an endoplasmic reticulum stress responsive gene. We previously reported that conditional knockout of hepatic ATF6 exacerbated liver metabolic damage by repressing autophagy through mTOR pathway. However, the mechanism by which ATF6 influence liver metabolism has not been well established. Hydrogen sulfide (H2S) is a gaseous signaling molecule that plays an important role in regulating inflammation, and suppress nonalcoholic fatty liver in mice. Based on the previous study, we assumed that ATF6 may regulate H2S production to participate in liver metabolism. In order to clarify the mechanism by which ATF6 regulates H2S synthesis to ameliorate liver steatosis and inflammatory environment, we conducted the present study. We used the liver specific ATF6 knockout mice and fed on high-fat-diet, and found that H2S level was significantly downregulated in hepatic ATF6 knockout mice. Restoring H2S by the administration of slow H2S releasing agent GYY4137 ameliorated the hepatic steatosis and glucose tolerance. ATF6 directly binds to the promoter of cystathionine ß synthetase (CBS), an important enzyme in H2S synthesis. Thus, ATF6 could upregulate H2S production through CBS. Sulfhydrated Sirtuin-1 (SIRT1) was downregulated in ATF6 knockout mice. The expression of pro-inflammatory factor IL-17A was upregulated and anti-inflammatory factor IL-10 was downregulated in ATF6 knockout mice. Our results suggest that ATF6 can transcriptionally enhance CBS expression as well as H2S synthesis. ATF6 increases SIRT1 sulfhydration and ameliorates lipogenesis and inflammation in the fatty liver. Therefore, ATF6 could be a novel therapeutic strategy for high-fat diet induced fatty liver metabolic abnormalities.


Subject(s)
Fatty Liver , Hydrogen Sulfide , Animals , Mice , Activating Transcription Factor 6/metabolism , Cystathionine/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Inflammation/metabolism , Ligases/metabolism , Liver/metabolism , Mice, Knockout , Sirtuin 1/metabolism
13.
J Biol Chem ; 299(12): 105449, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37949228

ABSTRACT

Cystathionine ß-synthase (CBS) catalyzes the committing step in the transsulfuration pathway, which is important for clearing homocysteine and furnishing cysteine. The transsulfuration pathway also generates H2S, a signaling molecule. CBS is a modular protein with a heme and pyridoxal phosphate-binding catalytic core, which is separated by a linker region from the C-terminal regulatory domain that binds S-adenosylmethionine (AdoMet), an allosteric activator. Recent cryo-EM structures reveal that CBS exists in a fibrillar form and undergoes a dramatic architectural rearrangement between the basal and AdoMet-bound states. CBS is the single most common locus of mutations associated with homocystinuria, and, in this study, we have characterized three clinical variants (K384E/N and M391I), which reside in the linker region. The native fibrillar form is destabilized in the variants, and differences in their limited proteolytic fingerprints also reveal conformational alterations. The crystal structure of the truncated K384N variant, lacking the regulatory domain, reveals that the overall fold of the catalytic core is unperturbed. M391I CBS exhibits a modest (1.4-fold) decrease while the K384E/N variants exhibit a significant (∼8-fold) decrease in basal activity, which is either unresponsive to or inhibited by AdoMet. Pre-steady state kinetic analyses reveal that the K384E/N substitutions exhibit pleiotropic effects and that the differences between them are expressed in the second half reaction, that is, homocysteine binding and reaction with the aminoacrylate intermediate. Together, these studies point to an important role for the linker in stabilizing the higher-order oligomeric structure of CBS and enabling AdoMet-dependent regulation.


Subject(s)
Cystathionine beta-Synthase , Mutation , Humans , Allosteric Regulation/genetics , Crystallography, X-Ray , Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Homocysteine/metabolism , Homocystinuria/enzymology , Homocystinuria/genetics , Kinetics , S-Adenosylmethionine/metabolism , Protein Conformation , Catalytic Domain
14.
Pharmacol Rep ; 75(6): 1610-1618, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37874531

ABSTRACT

BACKGROUND: Gaseous neurotransmitters have been thought to be novel factors involved in the mechanisms of mental disorders pathogenesis for quite some time. However, little is known about the potential crosstalk between neuronal gasotransmitter signaling and neuroleptics action. The present work was, therefore, focused on gene expression of H2S and CO-producing enzymes in the brains of rats chronically treated with olanzapine, an atypical antipsychotic drug. METHODS: Studies were carried out on adult, male Sprague-Dawley rats that were divided into 2 groups: control and experimental animals treated with olanzapine (28-day-long intraperitoneal injection, at a dose of 5 mg/kg daily). All individuals were sacrificed under anesthesia and the whole brains excised. Immunohistochemical procedure was used for histological assessment of the whole brain and for quantitative analysis of cystathionine ß-synthase (CBS) and heme oxygenase 2 (HO-2) protein distribution in selected brain structures. RESULTS: Long-term treatment with olanzapine is reflected in different changes in the number of enzymes-expressing cells in the rat brain. Olanzapine decreased the number of CBS-expressing cells and possibly reduced H2S synthesis in the hippocampus and striatum. The antipsychotic administration increased the number of HO-2 immunopositive cells and probably stimulated the CO production in the hippocampus. CONCLUSIONS: Modulatory effect of olanzapine on cellular mechanisms of gasotransmitter synthesis may be an alternative way of their pharmacological action.


Subject(s)
Antipsychotic Agents , Gasotransmitters , Hydrogen Sulfide , Animals , Male , Rats , Antipsychotic Agents/pharmacology , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Gasotransmitters/metabolism , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Hippocampus , Hydrogen Sulfide/metabolism , Olanzapine/pharmacology , Rats, Sprague-Dawley
15.
J Mol Neurosci ; 73(11-12): 921-931, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37864623

ABSTRACT

We aimed to investigate the mechanism underlying the roles of miRNA-377, Cystathionine-ß-synthase (CBS), and hydrogen sulfide (H2S) in the development of hypoxic-ischemic encephalopathy (HIE). We investigated the relationship between CBS, H2S, and miR-377 in both humans with HIE and animals with hypoxic-ischemic insult. An animal model of fetal rats with hypoxic-ischemic brain injury was established, and the fetal rats were randomly assigned to control and hypoxic-ischemic groups for 15 min (mild) and 30 min (moderate) groups. Human samples were collected from children diagnosed with HIE. Healthy or non-neurological disease children were selected as the control group. Hematoxylin-eosin (HE) staining, quantitative real-time polymerase chain reaction (qRT-PCR), enzyme-linked immunosorbent assay (ELISA), and western blot were used to conduct this study. Hypoxia-ischemia induced pathological alterations in brain tissue changes were more severe in groups with severe hypoxic insult. miRNA-377 expression levels were upregulated in brain tissue and serum of fetal rats and human samples with HIE compared to controls. Conversely, CBS and H2S expression levels were significantly decreased in both human and animal samples compared to controls. Our findings suggest that CBS is a target gene of miR-377 which may contribute to the development of HIE by regulating CBS/H2S. H2S has a protective effect against hypoxic damage in brain tissue. The study provides new insights into the potential mechanisms underlying the protective role of H2S in hypoxic brain damage and may contribute to the development of novel therapies for HIE.


Subject(s)
Hydrogen Sulfide , Hypoxia-Ischemia, Brain , MicroRNAs , Child , Humans , Rats , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Hypoxia-Ischemia, Brain/genetics , Cystathionine , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Rats, Sprague-Dawley , Hydrogen Sulfide/metabolism
16.
Biomolecules ; 13(10)2023 10 07.
Article in English | MEDLINE | ID: mdl-37892173

ABSTRACT

This study was performed on human primary (WM115) and metastatic (WM266-4) melanoma cell lines developed from the same individual. The expression of proteins involved in L-cysteine metabolism (sulfurtransferases, and cystathionine ß-synthase) and antioxidative processes (thioredoxin, thioredoxin reductase-1, glutathione peroxidase, superoxide dismutase 1) as well as the level of sufane sulfur, and cell proliferation under hypoxic conditions were investigated. Hypoxia in WM115 and WM266-4 cells was confirmed by induced expression of carbonic anhydrase IX and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 by the RT-PCR and Western blot methods. It was shown that, under hypoxic conditions the inhibition of WM115 and WM266-4 melanoma cell proliferation was associated with decreased expression of thioredoxin reductase-1 and cystathionine ß-synthase. These two enzymes may be important therapeutic targets in the treatment of melanoma. Interestingly, it was also found that in normoxia the expression and activity of 3-mercaptopyruvate sulfurtransferase in metastatic WM266-4 melanoma cells was significantly higher than in primary melanoma WM115 cells.


Subject(s)
Antioxidants , Melanoma , Humans , Cysteine/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Thioredoxin-Disulfide Reductase , Melanoma/pathology
17.
Int J Mol Sci ; 24(18)2023 Sep 21.
Article in English | MEDLINE | ID: mdl-37762687

ABSTRACT

Endogenous hydrogen sulfide (H2S) produced by cystathionine ß-synthase (CBS) and cystathionine-γ lyase (CSE) has emerged as a novel uterine vasodilator contributing to pregnancy-associated increases in uterine blood flow, which safeguard pregnancy health. Uterine artery (UA) H2S production is stimulated via exogenous estrogen replacement and is associated with elevated endogenous estrogens during pregnancy through the selective upregulation of CBS without altering CSE. However, how endogenous estrogens regulate uterine artery CBS expression in pregnancy is unknown. This study was conducted to test a hypothesis that endogenous estrogens selectively stimulate UA CBS expression via specific estrogen receptors (ER). Treatment with E2ß (0.01 to 100 nM) stimulated CBS but not CSE mRNA in organ cultures of fresh UA rings from both NP and P (gestational day 20, GD20) rats, with greater responses to all doses of E2ß tested in P vs. NP UA. ER antagonist ICI 182,780 (ICI, 1 µM) completely attenuated E2ß-stimulated CBS mRNA in both NP and P rat UA. Subcutaneous injection with ICI 182,780 (0.3 mg/rat) of GD19 P rats for 24 h significantly inhibited UA CBS but not mRNA expression, consistent with reduced endothelial and smooth muscle cell CBS (but not CSE) protein. ICI did not alter mesenteric and renal artery CBS and CSE mRNA. In addition, ICI decreased endothelial nitric oxide synthase mRNA in UA but not in mesenteric or renal arteries. Thus, pregnancy-augmented UA CBS/H2S production is mediated by the actions of endogenous estrogens via specific ER in pregnant rats.


Subject(s)
Cystathionine beta-Synthase , Fulvestrant , Hydrogen Sulfide , Animals , Female , Pregnancy , Rats , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Estrogens/metabolism , Fulvestrant/pharmacology , Hydrogen Sulfide/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation , Uterine Artery/metabolism
18.
Mol Genet Metab ; 139(4): 107653, 2023 08.
Article in English | MEDLINE | ID: mdl-37463544

ABSTRACT

Classical homocystinuria (HCU) is a rare inborn error of amino acid metabolism characterized by accumulation of homocysteine, an intermediate product of methionine metabolism, leading to significant systemic toxicities, particularly within the vascular, skeletal, and ocular systems. Most patients require lifelong dietary therapy with severe restriction of natural protein to minimize methionine intake, and many patients still struggle to maintain healthy homocysteine levels. Since eliminating methionine from the diet reduces homocysteine levels, we hypothesized that an enzyme that can degrade methionine within the gastrointestinal (GI) tract could help HCU patients maintain healthy levels while easing natural protein restrictions. We describe the preclinical development of CDX-6512, a methionine gamma lyase (MGL) enzyme that was engineered for stability and activity within the GI tract for oral administration to locally degrade methionine. CDX-6512 is stable to low pH and intestinal proteases, enabling it to survive the harsh GI environment without enteric coating and to degrade methionine freed from dietary protein within the small intestine. Administering CDX-6512 to healthy non-human primates following a high protein meal led to a dose-dependent suppression of plasma methionine. In Tg-I278T Cbs-/- mice, an animal model that recapitulates aspects of HCU disease including highly elevated serum homocysteine levels, oral dosing of CDX-6512 after a high protein meal led to suppression in serum levels of both methionine and homocysteine. When animals received a daily dose of CDX-6512 with a high protein meal for two weeks, the Tg-I278T Cbs-/- mice maintained baseline homocysteine levels, whereas homocysteine levels in untreated animals increased by 39%. These preclinical data demonstrate the potential of CDX-6512 as an oral enzyme therapy for HCU.


Subject(s)
Homocystinuria , Humans , Mice , Animals , Homocystinuria/drug therapy , Homocystinuria/genetics , Methionine/metabolism , Homocysteine , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Racemethionine , Gastrointestinal Tract/metabolism
19.
J Inherit Metab Dis ; 46(6): 1114-1130, 2023 11.
Article in English | MEDLINE | ID: mdl-37477632

ABSTRACT

The loss of cystathionine ß-synthase (CBS), an important homocysteine (Hcy)-metabolizing enzyme or the loss of PHF8, an important histone demethylase participating in epigenetic regulation, causes severe intellectual disability in humans. Similar neuropathies were also observed in Cbs-/- and Phf8-/- mice. How CBS or PHF8 depletion can cause neuropathy was unknown. To answer this question, we examined a possible interaction between PHF8 and CBS using Cbs-/- mouse and neuroblastoma cell models. We quantified gene expression by RT-qPCR and western blotting, mTOR-bound H4K20me1 by chromatin immunoprecipitation (CHIP) assay, and amyloid ß (Aß) by confocal fluorescence microscopy using anti-Aß antibody. We found significantly reduced expression of Phf8, increased H4K20me1, increased mTOR expression and phosphorylation, and increased App, both on protein and mRNA levels in brains of Cbs-/- mice versus Cbs+/- sibling controls. Autophagy-related Becn1, Atg5, and Atg7 were downregulated while p62, Nfl, and Gfap were upregulated on protein and mRNA levels, suggesting reduced autophagy and increased neurodegeneration in Cbs-/- brains. In mouse neuroblastoma N2a or N2a-APPswe cells, treatments with Hcy-thiolactone, N-Hcy-protein or Hcy, or Cbs gene silencing by RNA interference significantly reduced Phf8 expression and increased total H4K20me1 as well as mTOR promoter-bound H4K20me1. This led to transcriptional mTOR upregulation, autophagy downregulation, and significantly increased APP and Aß levels. The Phf8 gene silencing increased Aß, but not APP, levels. Taken together, our findings identify Phf8 as a regulator of Aß synthesis and suggest that neuropathy of Cbs deficiency is mediated by Hcy metabolites, which transcriptionally dysregulate the Phf8 → H4K20me1 → mTOR → autophagy pathway thereby increasing Aß accumulation.


Subject(s)
Cystathionine beta-Synthase , Neuroblastoma , Animals , Mice , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Autophagy/genetics , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Epigenesis, Genetic , Histone Demethylases/genetics , Histone Demethylases/metabolism , Neuroblastoma/genetics , RNA, Messenger , TOR Serine-Threonine Kinases , Transcription Factors/genetics
20.
PLoS One ; 18(6): e0286550, 2023.
Article in English | MEDLINE | ID: mdl-37319242

ABSTRACT

Classic homocystinuria is an inborn error of metabolism caused mainly by missense mutations leading to misfolded and/or unstable human cystathionine ß-synthase (CBS) protein, causing the accumulation of excess total homocysteine (tHcy) in tissues. Previously, it has been shown that certain missense containing human CBS proteins can be functionally rescued in mouse models of CBS deficiency by treatment with proteasome inhibitors. The rescue by proteasome inhibitors is thought to work both by inhibiting the degradation of misfolded CBS protein and by inducing the levels of heat-shock chaperone proteins in the liver. Here we examine the effectiveness of two FDA approved protease inhibitors, carfilzomib and bortezomib, on various transgenic mouse models of human CBS deficiency. Our results show that although both drugs are effective in inducing the liver chaperone proteins Hsp70 and Hsp27, and are effective in inhibiting proteasome function, bortezomib was somewhat more robust in restoring the mutant CBS function. Moreover, there was no significant correlation between proteasome inhibition and CBS activity, suggesting that some of bortezomib's effects are via other mechanisms. We also test the use of low-doses of bortezomib and carfilzomib on various mouse models for extended periods of time and find that while low-doses are less toxic, they are also less effective at restoring CBS function. Overall, these results show that while restoration of mutant CBS function is possible with proteasome inhibitors, the exact mechanism is complicated and it will likely be too toxic for long-term patient treatment.


Subject(s)
Cystathionine beta-Synthase , Homocystinuria , Humans , Mice , Animals , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Homocystinuria/drug therapy , Homocystinuria/genetics , Homocystinuria/metabolism , Bortezomib/pharmacology , Bortezomib/therapeutic use , Proteasome Endopeptidase Complex , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...