Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Publication year range
1.
Physiol Rev ; 99(1): 555-604, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30427275

ABSTRACT

Epidemiological studies established that elevated homocysteine, an important intermediate in folate, vitamin B12, and one carbon metabolism, is associated with poor health, including heart and brain diseases. Earlier studies show that patients with severe hyperhomocysteinemia, first identified in the 1960s, exhibit neurological and cardiovascular abnormalities and premature death due to vascular complications. Although homocysteine is considered to be a nonprotein amino acid, studies over the past 2 decades have led to discoveries of protein-related homocysteine metabolism and mechanisms by which homocysteine can become a component of proteins. Homocysteine-containing proteins lose their biological function and acquire cytotoxic, proinflammatory, proatherothrombotic, and proneuropathic properties, which can account for the various disease phenotypes associated with hyperhomocysteinemia. This review describes mechanisms by which hyperhomocysteinemia affects cellular proteostasis, provides a comprehensive account of the biological chemistry of homocysteine-containing proteins, and discusses pathophysiological consequences and clinical implications of their formation.


Subject(s)
Cardiovascular Diseases/metabolism , Homocysteine/metabolism , Hyperhomocysteinemia/metabolism , Vitamin B 12/metabolism , Animals , Folic Acid/metabolism , Homocysteine/chemistry , Humans , Risk Factors
2.
FASEB J ; 38(18): e70072, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39323294

ABSTRACT

The inability to efficiently metabolize homocysteine (Hcy) due to nutritional and genetic deficiencies, leads to hyperhomocysteinemia (HHcy) and endothelial dysfunction, a hallmark of atherosclerosis which underpins cardiovascular disease (CVD). PHF8 is a histone demethylase that demethylates H4K20me1, which affects the mammalian target of rapamycin (mTOR) signaling and autophagy, processes that play important roles in CVD. PHF8 is regulated by microRNA (miR) such as miR-22-3p and miR-1229-3p. Biochemically, HHcy is characterized by elevated levels of Hcy, Hcy-thiolactone and N-Hcy-protein. Here, we examined the effects of these metabolites on miR-22-3p, miR-1229-3p, and their target PHF8, as well as on the downstream consequences of these effects on H4K20me1, mTOR-, and autophagy-related proteins and mRNAs expression in human umbilical vein endothelial cells (HUVEC). We found that treatments with N-Hcy-protein, Hcy-thiolactone, or Hcy upregulated miR-22-3p and miR-1229-3p, attenuated PHF8 expression, upregulated H4K20me1, mTOR, and phospho-mTOR. Autophagy-related proteins (BECN1, ATG5, ATG7, lipidated LC3-II, and LC3-II/LC3-I ratio) were significantly downregulated by at least one of these metabolites. We also found similar changes in the expression of miR-22-3p, Phf8, mTOR- and autophagy-related proteins/mRNAs in vivo in hearts of Cbs-/- mice, which show severe HHcy and endothelial dysfunction. Treatments with inhibitors of miR-22-3p or miR-1229-3p abrogated the effects of Hcy-thiolactone, N-Hcy-protein, and Hcy on miR expression and on PHF8, H4K20me1, mTOR-, and autophagy-related proteins/mRNAs in HUVEC. Taken together, these findings show that Hcy metabolites upregulate miR-22-3p and miR-1229-3p expression, which then dysregulate the PHF8/H4K20me1/mTOR/autophagy pathway, important for vascular homeostasis.


Subject(s)
Autophagy , Histone Demethylases , Homocysteine , Human Umbilical Vein Endothelial Cells , MicroRNAs , TOR Serine-Threonine Kinases , Transcription Factors , Animals , Female , Humans , Mice , Histone Demethylases/metabolism , Histone Demethylases/genetics , Homocysteine/metabolism , Homocysteine/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Up-Regulation
3.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125665

ABSTRACT

Elevated levels of homocysteine (Hcy) and related metabolites are associated with Alzheimer's disease (AD). Severe hyperhomocysteinemia causes neurological deficits and worsens behavioral and biochemical traits associated with AD. Although Hcy is precluded from entering the Genetic Code by proofreading mechanisms of aminoacyl-tRNA synthetases, and thus is a non-protein amino acid, it can be attached to proteins via an N-homocysteinylation reaction mediated by Hcy-thiolactone. Because N-homocysteinylation is detrimental to a protein's function and biological integrity, Hcy-thiolactone-detoxifying enzymes-PON1, BLMH, BPHL-have evolved. This narrative review provides an account of the biological function of these enzymes and of the consequences of their impairments, leading to the phenotype characteristic of AD. Overall, accumulating evidence discussed in this review supports a hypothesis that Hcy-thiolactone contributes to neurodegeneration associated with a dysregulated Hcy metabolism.


Subject(s)
Alzheimer Disease , Homocysteine , Humans , Alzheimer Disease/metabolism , Homocysteine/metabolism , Homocysteine/analogs & derivatives , Animals , Hyperhomocysteinemia/metabolism
4.
FASEB J ; 36(10): e22547, 2022 10.
Article in English | MEDLINE | ID: mdl-36098436

ABSTRACT

Reproductive success in mice depends on sexually dimorphic major urinary proteins (Mup) that facilitate interactions between females and males. Deletion of cystathionine ß-synthase (Cbs) gene, a metabolic gene important for homeostasis of one-carbon metabolism, impairs reproduction by causing female infertility in mice. Here, we examined Mup biogenesis and sexual signaling in Cbs-/- versus Cbs+/- mice. We found significantly reduced levels of total urinary Mup protein in male and female Cbs-/- versus Cbs+/- mice. SDS-PAGE/Western blot, ESI-MS, and RT-qPCR analyses of the liver, plasma, and urinary proteins identified a male-specific Mup20 in Cbs-/- , but not in Cbs+/- females. The 18 893 Da Mup20 became the most abundant in urine of Cbs-/- females and males. Effects of Cbs genotype on 18 645 Da, 18 693 Da, and 18 709 Da Mup species abundance were Mup- and sex-specific. Cbs genotype-dependent changes in hepatic Mups and Mup20 expression were similar at the protein and mRNA level. Changes in Mups, but not in Mup20, can be explained by downregulation of hepatic Zhx2 and Ghr receptors in Cbs-/- mice. Behavioral testing showed that Cbs+/- females ignored Cbs-/- male urine but were attracted to Cbs+/- male urine. Cbs+/- males ignored urine of Cbs-/- males but countermarked urine of other Cbs+/- males and were attracted to urines of Cbs-/- as well as Cbs+/- females. Cbs-/- males did not countermark urine of Cbs+/- males but were attracted to urines of Cbs+/- females. Taken together, these findings show that Cbs, a metabolic gene, interacts with the processes involved in Mup biogenesis that are essential for the maintenance of sexual dimorphism and signaling and suggest that dysregulation of these interactions impairs reproductive fitness in mice.


Subject(s)
DNA Repair Enzymes/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Biosynthesis , Animals , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , DNA Repair Enzymes/genetics , Female , Gene Silencing , Male , Mice , Poly-ADP-Ribose Binding Proteins/genetics , Proteins/metabolism
5.
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
6.
Int J Mol Sci ; 24(9)2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37175471

ABSTRACT

High-density lipoprotein (HDL) exhibits cardio- and neuro-protective properties, which are thought to be promoted by paraoxonase 1 (PON1), a hydrolytic enzyme associated with an HDL subfraction also enriched with an anticoagulant protein (PROS1) and amyloid beta-transport protein clusterin (CLU, APOJ). Reduced levels of PON1 activity, characterized biochemically by elevated levels of homocysteine (Hcy)-thiolactone, oxidized lipids, and proteins modified by these metabolites in humans and mice, are associated with pathological abnormalities affecting the cardiovascular system (atherothrombosis) and the central nervous system (cognitive impairment, Alzheimer's disease). The molecular bases of these abnormalities have been largely unknown. Proteomic and metabolic studies over the past decade have significantly contributed to our understanding of PON1 function and the mechanisms by which PON1 deficiency can lead to disease. Recent studies discussed in this review highlight the involvement of dysregulated proteostasis in the pro-oxidative, pro-atherothrombotic, and pro-amyloidogenic phenotypes associated with low PON1 activity.


Subject(s)
Alzheimer Disease , Aryldialkylphosphatase , Humans , Mice , Animals , Aryldialkylphosphatase/metabolism , Amyloid beta-Peptides , Proteomics , Lipoproteins, HDL/metabolism , Alzheimer Disease/metabolism
7.
Int J Mol Sci ; 23(8)2022 Apr 10.
Article in English | MEDLINE | ID: mdl-35456998

ABSTRACT

Dysregulation of one-carbon metabolism affects a wide range of biological processes and is associated with a number of diseases, including cardiovascular disease, dementia, neural tube defects, and cancer. Accumulating evidence suggests that one-carbon metabolism plays an important role in COVID-19. The symptoms of long COVID-19 are similar to those presented by subjects suffering from vitamin B12 deficiency (pernicious anemia). The metabolism of a cell infected by the SARS-CoV-2 virus is reshaped to fulfill the need for massive viral RNA synthesis, which requires de novo purine biosynthesis involving folate and one-carbon metabolism. Many aspects of host sulfur amino acid metabolism, particularly glutathione metabolism underlying antioxidant defenses, are also taken over by the SARS-CoV-2 virus. The purpose of this review is to summarize recent findings related to one-carbon metabolism and sulfur metabolites in COVID-19 and discuss how they inform strategies to combat the disease.


Subject(s)
COVID-19 , COVID-19/complications , Carbon/metabolism , Folic Acid/metabolism , Homocysteine , Humans , Methionine/metabolism , SARS-CoV-2 , Vitamin B 12/metabolism , Post-Acute COVID-19 Syndrome
8.
J Proteome Res ; 20(5): 2458-2476, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33797904

ABSTRACT

Protein N-homocysteinylation by a homocysteine (Hcy) metabolite, Hcy-thiolactone, is an emerging post-translational modification (PTM) that occurs in all tested organisms and has been linked to human diseases. The yeast Saccharomyces cerevisiae is widely used as a model eukaryotic organism in biomedical research, including studies of protein PTMs. However, patterns of global protein N-homocysteinylation in yeast are not known. Here, we identified 68 in vivo and 197 in vitro N-homocysteinylation sites at protein lysine residues (N-Hcy-Lys). Some of the N-homocysteinylation sites overlap with other previously identified PTM sites. Protein N-homocysteinylation in vivo, induced by supplementation of yeast cultures with Hcy, which elevates Hcy-thiolactone levels, was accompanied by significant changes in the levels of 70 yeast proteins (38 up-regulated and 32 down-regulated) involved in the ribosomal structure, amino acid biosynthesis, and basic cellular pathways. Our study provides the first global survey of N-homocysteinylation and accompanying changes in the yeast proteome caused by elevated Hcy level. These findings suggest that protein N-homocysteinylation and dysregulation of cellular proteostasis may contribute to the toxicity of Hcy in yeast. Homologous proteins and N-homocysteinylation sites are likely to be involved in Hcy-related pathophysiology in humans and experimental animals. Data are available via ProteomeXchange with identifier PXD020821.


Subject(s)
Lysine , Saccharomyces cerevisiae , Animals , Homocysteine , Humans , Lysine/metabolism , Protein Processing, Post-Translational , Proteome , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
9.
Expert Rev Proteomics ; 17(10): 751-765, 2020 10.
Article in English | MEDLINE | ID: mdl-33320032

ABSTRACT

Introduction: Homocystinuria due to cystathionine ß-synthase (CBS) deficiency, the most frequent inborn error of sulfur amino acid metabolism, is characterized biochemically by severely elevated homocysteine (Hcy) and related metabolites, such as Hcy-thiolactone and N-Hcy-protein. CBS deficiency reduces life span and causes pathological abnormalities affecting most organ systems in the human body, including the cardiovascular (thrombosis, stroke), skeletal/connective tissue (osteoporosis, thin/non-elastic skin, thin hair), and central nervous systems (mental retardation, seizures), as well as the liver (fatty changes), and the eye (ectopia lentis, myopia). Molecular basis of these abnormalities were largely unknown and available treatments remain ineffective. Areas covered: Proteomic and transcriptomic studies over the past decade or so, have significantly contributed to our understanding of mechanisms by which the CBS enzyme deficiency leads to clinical manifestations associated with it. Expert opinion: Recent findings, discussed in this review, highlight the involvement of dysregulated proteostasis in pathologies associated with CBS deficiency, including thromboembolism, stroke, neurologic impairment, connective tissue/collagen abnormalities, hair defects, and hepatic toxicity. To ameliorate these pathologies, pharmacological, enzyme replacement, and gene transfer therapies are being developed.


Subject(s)
Cystathionine beta-Synthase/deficiency , Cystathionine beta-Synthase/metabolism , Fatty Liver/enzymology , Fatty Liver/metabolism , Animals , Autophagy/physiology , Fibrinogen/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Proteomics/methods , Transcriptome/genetics , Transcriptome/physiology
10.
Int J Mol Sci ; 21(7)2020 Apr 05.
Article in English | MEDLINE | ID: mdl-32260476

ABSTRACT

Cystathionine ß-synthase (CBS) is a housekeeping enzyme that catalyzes the first step of the homocysteine to cysteine transsulfuration pathway. Homozygous deletion of the Cbs gene in mice causes severe hyperhomocysteinemia and reduces life span. Here, we examined a possible involvement of senescence, mitochondrial DNA, and telomeres in the reduced life span of Cbs-/- mice. We found that senescence-related p21, Pai-1, Mcp1, and Il-6 mRNAs were significantly upregulated (2-10-fold) in liver, while p21 was upregulated in the brain of Cbs-/- mice (n = 20) compared with control Cbs+/- siblings (n = 20) in a sex- and age-dependent manner. Telomere length in blood (n = 80), liver (n = 40), and brain (n = 40) was not affected by the Cbs-/- genotype, but varied with sex and/or age. Levels of mitochondrial DNA tended to be reduced in livers, but not brains and blood, of Cbs-/- females (n = 20-40). The Cbs-/- genotype significantly reduced Tert mRNA expression in brain, but not liver, in a sex- and age-dependent manner. Multiple regression analysis showed that the senescence-related liver (but not brain) mRNAs and liver (but not brain or blood) mitochondrial DNA were associated with the Cbs genotype. In contrast, telomere length in blood, brain, and liver was not associated with the Cbs genotype or hyperhomocysteinemia, but was associated with sex (in brain and liver) and age (in brain and blood). Taken together, these findings suggest that the changes in senescence marker expression and mtDNA levels, but not telomere shortening, could account for the reduced life span of Cbs-/- mice.


Subject(s)
Cystathionine beta-Synthase/genetics , DNA, Mitochondrial/genetics , Gene Dosage , Longevity/genetics , Telomere Homeostasis , Animals , Brain/growth & development , Brain/metabolism , Female , Genetic Loci , Kidney/growth & development , Kidney/metabolism , Liver/growth & development , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Telomerase/genetics , Telomerase/metabolism
11.
FASEB J ; : fj201800346R, 2018 May 21.
Article in English | MEDLINE | ID: mdl-29782204

ABSTRACT

Genetic or nutritional deficiencies in 1 carbon and homocysteine (Hcy) metabolism elevate Hcy-thiolactone levels and are associated with cardiovascular and neurologic diseases. Hcy-thiolactone causes protein damage, cellular toxicity, and proatherogenic changes in gene expression in human cells and tissues. A polymorphic cardio-protective enzyme, paraoxonase 1 (PON1), hydrolyzes Hcy-thiolactone in vitro. However, whether Hcy-thiolactone hydrolysis is a physiologic function of the PON1 protein and whether polymorphisms in the PON1 gene affect Hcy-thiolactone levels in humans was unknown. Here we show that the PON1-192 genotype, which affects the enzymatic activity of the PON1 protein, also affected urinary Hcy-thiolactone levels, normalized to creatinine. Carriers of the PON1-192R allele had significantly lower Hcy-thiolactone/creatinine levels than individuals carrying the PON1-192Q allele. Individuals with low serum PON1 paraoxonase activity had significantly higher Hcy-thiolactone/creatinine levels compared with individuals with high paraoxonase activity. In contrast, Hcy-thiolactone/creatinine levels were unaffected by serum PON1 arylesterase activity or by PON1 protein levels. Taken together, these findings suggest that PON1 hydrolyzes Hcy-thiolactone in humans and that the interindividual variations in PON1 genotype/activity can modulate the pathology of hyperhomocysteinemia.-Perla-Kaján, J., Borowczyk, K., Glowacki, R., Nygård, O., Jakubowski, H. Paraoxonase 1 Q192r genotype and activity affect homocysteine thiolactone levels in humans.

12.
Int J Mol Sci ; 20(13)2019 Jun 27.
Article in English | MEDLINE | ID: mdl-31252610

ABSTRACT

Hyperhomocysteinemia (HHcy) exerts a wide range of biological effects and is associated with a number of diseases, including cardiovascular disease, dementia, neural tube defects, and cancer. Although mechanisms of HHcy toxicity are not fully uncovered, there has been a significant progress in their understanding. The picture emerging from the studies of homocysteine (Hcy) metabolism and pathophysiology is a complex one, as Hcy and its metabolites affect biomolecules and processes in a tissue- and sex-specific manner. Because of their connection to one carbon metabolism and editing mechanisms in protein biosynthesis, Hcy and its metabolites impair epigenetic control of gene expression mediated by DNA methylation, histone modifications, and non-coding RNA, which underlies the pathology of human disease. In this review we summarize the recent evidence showing that epigenetic dysregulation of gene expression, mediated by changes in DNA methylation and histone N-homocysteinylation, is a pathogenic consequence of HHcy in many human diseases. These findings provide new insights into the mechanisms of human disease induced by Hcy and its metabolites, and suggest therapeutic targets for the prevention and/or treatment.


Subject(s)
Epigenesis, Genetic , Hyperhomocysteinemia/genetics , Animals , DNA Methylation , Histone Code , Homocysteine/metabolism , Humans , Hyperhomocysteinemia/metabolism , RNA, Untranslated/genetics
13.
Int J Mol Sci ; 20(12)2019 Jun 25.
Article in English | MEDLINE | ID: mdl-31242583

ABSTRACT

Ischemic stroke induces brain injury via thrombotic or embolic mechanisms involving large or small vessels. Cystathionine ß-synthase deficiency (CBS), an inborn error of metabolism, is associated with vascular thromboembolism, the major cause of morbidity and mortality in affected patients. Because thromboembolism involves the brain vasculature in these patients, we hypothesize that CBS deficiency and ischemic stroke have similar molecular phenotypes. We used label-free mass spectrometry for quantification of changes in serum proteomes in CBS-deficient patients (n = 10) and gender/age-matched unaffected controls (n = 14), as well as in patients with cardioembolic (n = 17), large-vessel (n = 26), or lacunar (n = 25) ischemic stroke subtype. In CBS-deficient patients, 40 differentially expressed serum proteins were identified, of which 18 were associated with elevated homocysteine (Hcy) and 22 were Hcy-independent. We also identified Hcy-independent differentially expressed serum proteins in ischemic stroke patients, some of which were unique to a specific subtype: 10 of 32 for cardioembolic vs. large-vessel, six of 33 for cardioembolic vs. lacunar, and six of 23 for large-vessel vs. lacunar. There were significant overlaps between proteins affected by CBS deficiency and ischemic stroke, particularly the cardioembolic subtype, similar to protein overlaps between ischemic stroke subtypes. Top molecular pathways affected by CBS deficiency and ischemic stroke subtypes included acute phase response signaling and coagulation system. Similar molecular networks centering on NFκB were affected by CBS deficiency and stroke subtypes. These findings suggest common mechanisms involved in the pathologies of CBS deficiency and ischemic stroke subtypes.


Subject(s)
Biomarkers/blood , Brain Ischemia/complications , Cystathionine beta-Synthase/deficiency , Proteome , Proteomics , Stroke/blood , Stroke/etiology , Adult , Computational Biology/methods , Female , Humans , Male , Middle Aged , Proteomics/methods , Signal Transduction
14.
PLoS Pathog ; 12(10): e1005949, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27760199

ABSTRACT

The methylfolate trap, a metabolic blockage associated with anemia, neural tube defects, Alzheimer's dementia, cardiovascular diseases, and cancer, was discovered in the 1960s, linking the metabolism of folate, vitamin B12, methionine and homocysteine. However, the existence or physiological significance of this phenomenon has been unknown in bacteria, which synthesize folate de novo. Here we identify the methylfolate trap as a novel determinant of the bacterial intrinsic death by sulfonamides, antibiotics that block de novo folate synthesis. Genetic mutagenesis, chemical complementation, and metabolomic profiling revealed trap-mediated metabolic imbalances, which induced thymineless death, a phenomenon in which rapidly growing cells succumb to thymine starvation. Restriction of B12 bioavailability, required for preventing trap formation, using an "antivitamin B12" molecule, sensitized intracellular bacteria to sulfonamides. Since boosting the bactericidal activity of sulfonamides through methylfolate trap induction can be achieved in Gram-negative bacteria and mycobacteria, it represents a novel strategy to render these pathogens more susceptible to existing sulfonamides.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/metabolism , Drug Resistance, Microbial/physiology , Folic Acid/metabolism , Homocysteine/metabolism , Methionine/metabolism , Microbial Sensitivity Tests , Vitamin B 12/metabolism
15.
FASEB J ; 31(2): 598-609, 2017 02.
Article in English | MEDLINE | ID: mdl-28148781

ABSTRACT

The molecular mechanisms leading to and responsible for age-related, sporadic Alzheimer's disease (AD) remain largely unknown. It is well documented that aging patients with elevated levels of the amino acid metabolite homocysteine (Hcy) are at high risk of developing AD. We investigated the impact of Hcy on molecular clearance pathways in mammalian cells, including in vitro cultured induced pluripotent stem cell-derived forebrain neurons and in vivo neurons in mouse brains. Exposure to Hcy resulted in up-regulation of the mechanistic target of rapamycin complex 1 (mTORC1) activity, one of the major kinases in cells that is tightly linked to anabolic and catabolic pathways. Hcy is sensed by a constitutive protein complex composed of leucyl-tRNA-synthetase and folliculin, which regulates mTOR tethering to lysosomal membranes. In hyperhomocysteinemic human cells and cystathionine ß-synthase-deficient mouse brains, we find an acute and chronic inhibition of the molecular clearance of protein products resulting in a buildup of abnormal proteins, including ß-amyloid and phospho-Tau. Formation of these protein aggregates leads to AD-like neurodegeneration. This pathology can be prevented by inhibition of mTORC1 or by induction of autophagy. We conclude that an increase of intracellular Hcy levels predisposes neurons to develop abnormal protein aggregates, which are hallmarks of AD and its associated onset and pathophysiology with age.-Khayati, K., Antikainen, H., Bonder, E. M., Weber, G. F., Kruger, W. D., Jakubowski, H., Dobrowolski, R. The amino acid metabolite homocysteine activates mTORC1 to inhibit autophagy and form abnormal proteins in human neurons and mice.


Subject(s)
Autophagy/physiology , Gene Expression Regulation/physiology , Homocysteine/metabolism , Multiprotein Complexes/metabolism , Neurons/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , TOR Serine-Threonine Kinases/genetics
16.
Amino Acids ; 50(5): 537-546, 2018 05.
Article in English | MEDLINE | ID: mdl-29480334

ABSTRACT

Growing human head hair contains a history of keratin and provides a unique model for studies of protein damage. Here, we examined mechanism of homocysteine (Hcy) accumulation and keratin damage in human hair. We found that the content of Hcy-keratin increased along the hair fiber, with levels 5-10-fold higher levels in older sections at the hair's tip than in younger sections at hair's base. The accumulation of Hcy led to a complete loss of keratin solubility in sodium dodecyl sulfate. The increase in Hcy-keratin was accompanied by a decrease in methionine-keratin. Levels of Hcy-keratin were correlated with hair copper and iron in older hair. These relationships were recapitulated in model experiments in vitro, in which Hcy generation from Met exhibited a similar dependence on copper or iron. Taken together, these findings suggest that Hcy-keratin accumulation is due to copper/iron-catalyzed demethylation of methionine residues and contributes to keratin damage in human hair.


Subject(s)
Aging/metabolism , Hair/growth & development , Keratins/metabolism , Methionine/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Copper/metabolism , Female , Homocysteine/metabolism , Humans , Infant , Iron/metabolism , Male , Methylation , Middle Aged
17.
FASEB J ; 30(11): 3810-3821, 2016 11.
Article in English | MEDLINE | ID: mdl-27530978

ABSTRACT

Cystathionine ß-synthase (CBS) deficiency, a genetic disorder in homocysteine (Hcy) metabolism in humans, elevates plasma Hcy-thiolactone and leads to connective tissue abnormalities that affect the cardiovascular and skeletal systems. However, the underlying mechanism of these abnormalities is not understood. Hcy-thiolactone has the ability to form isopeptide bonds with protein lysine residues, which generates N-homocysteinylated protein. Because lysine residues are involved in collagen cross-linking, N-homocysteinylation of these lysines should impair cross-linking. Using a Tg-I278T Cbs-/- mouse model of hyperhomocysteinemia (HHcy) which replicates the connective tissue abnormalities observed in CBS-deficient patients, we found that N-Hcy-collagen was elevated in bone, tail, and heart of Cbs-/- mice, whereas pyridinoline cross-links were significantly reduced. Plasma deoxypyridinoline cross-link and cross-linked carboxyterminal telopeptide of type I collagen were also significantly reduced in the Cbs-/- mice. Lysine oxidase activity and mRNA level were not reduced by the Cbs-/- genotype. We also showed that collagen carries S-linked Hcy bound to the thiol of N-linked Hcy. In vitro experiments showed that Hcy-thiolactone modifies lysine residues in collagen type I α-1 chain. Residue K160, located in the nonhelical N-telopeptide region and involved in pyridinoline cross-link formation, was also N-homocysteinylated in vivo Taken together, our findings showed that N-homocysteinylation of collagen in Cbs-/- mice impairs its cross-linking. These findings explain, at least in part, connective tissue abnormalities observed in HHcy.-Perla-Kajan, J., Utyro, O., Rusek, M., Malinowska, A., Sitkiewicz, E., Jakubowski, H. N-Homocysteinylation impairs collagen cross-linking in cystathionine ß-synthase-deficient mice: a novel mechanism of connective tissue abnormalities.


Subject(s)
Collagen Type I/metabolism , Connective Tissue/metabolism , Cystathionine beta-Synthase/metabolism , Homocysteine/analogs & derivatives , Hyperhomocysteinemia/metabolism , Animals , Collagen Type I, alpha 1 Chain , Cystathionine beta-Synthase/genetics , Homocysteine/metabolism , Homocystinuria/genetics , Lysine/metabolism , Mice, Knockout , Peptides/metabolism
18.
Anal Biochem ; 508: 118-23, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27293214

ABSTRACT

Homocysteine (Hcy) and its metabolites Hcy-thiolactone, N-Hcy-protein, and S-Hcy-protein are implicated in vascular and neurological diseases. However, quantification of these metabolites remains challenging. Here I describe streamlined assays for these metabolites based on their conversion to Hcy-thiolactone. Free Hcy-thiolactone is extracted from the urine with chloroform/methanol. Total Hcy is converted to Hcy-thiolactone in the presence of 1 N HCl. Major urinary protein (MUP)-bound S-linked Hcy is liberated from the protein by reduction with dithiothreitol and converted to Hcy-thiolactone. Acid hydrolysis of MUP with 6 N HCl liberates N-linked Hcy as Hcy-thiolactone, which is then extracted with chloroform/methanol. Ferritin is used as an N-Hcy-protein standard and an authentic Hcy-thiolactone is used to monitor the efficiency of extraction. Hcy-thiolactone (free, derived from total Hcy, or from MUP-bound N-linked or S-linked Hcy) is separated by a cation exchange high-performance liquid chromatography, post-column derivatized with o-phthaldialdehyde, and quantified by fluorescence. Using these assays with as little as 2-20 µL of urine I show that MUP carry N-linked and S-linked Hcy and that N-Hcy-MUP and S-Hcy-MUP and Hcy-thiolactone are severely elevated in cystathionine ß-synthase-deficient mice. These assays will facilitate examination of the role of protein-related Hcy metabolites in health and disease.


Subject(s)
Cystathionine beta-Synthase/urine , Homocysteine/analogs & derivatives , Homocysteine/urine , Urinalysis/methods , Animals , Chromatography, High Pressure Liquid , Mice
19.
Anal Bioanal Chem ; 408(7): 1935-41, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26794212

ABSTRACT

A simple and rapid assay using pyridoxal 5'-phosphate (PLP) as a derivatizing reagent was developed for the simultaneous determination of homocysteine (Hcy) and cysteine (Cys) in human plasma. Derivatization with PLP affords UV-absorbing tetrahydrothiazine and thiazolidine derivatives of Hcy and Cys, respectively. Separation of these derivatives was achieved in 5 min using a hydrophilic interaction liquid chromatography, followed by UV detection at 330 nm. Linearity in detector response was observed over the range of 0.25-20 µM for Hcy and 10-300 µM for Cys. The limit of quantification (LOQ) values for Hcy and Cys were 0.25 and 2.5 µM, respectively. The method was successfully applied to plasma samples donated by apparently healthy volunteers.


Subject(s)
Chromatography, High Pressure Liquid/methods , Cysteine/blood , Homocysteine/blood , Cysteine/analysis , Homocysteine/analysis , Humans , Hydrophobic and Hydrophilic Interactions , Limit of Detection , Pyridoxal Phosphate/chemistry , Reproducibility of Results
20.
Biomed Environ Sci ; 29(10): 706-712, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27927270

ABSTRACT

OBJECTIVE: Obesity is associated with kidney defects. Physical activity is a key element in the treatment of obesity. The aim of this study was to compare the effect of endurance and endurance-strength training on kidney function in abdominally obese women. METHODS: Forty-four abdominally obese women were randomized to endurance training or endurance-strength training, three times a week for 3 months. Before and after the intervention, kidney function was assessed by measuring blood creatinine, urine creatinine, and urine albumin levels, and the albumin-to-creatinine ratio and glomerular filtration rate (GFR) were calculated. RESULTS: Renal hyperperfusion was present in both groups before the study. Following both types of physical activity, similar modifications of the investigated parameters were observed, but with no significant between-group differences. Both courses of training led to a significant increase in blood creatinine and a subsequent decrease in the GFR. A significant increase in urine creatinine and album levels, though not exceeding the range for microalbuminuria, was not accompanied by any difference in the albumin-to-creatinine ratio after endurance-strength training alone. CONCLUSION: Three months of either endurance or endurance-strength training has a favorable and comparable effect on renal function in abdominally obese women with renal hyperfiltration.


Subject(s)
Kidney/physiopathology , Obesity/therapy , Resistance Training , Adolescent , Adult , Aged , Creatinine/blood , Female , Glomerular Filtration Rate , Humans , Middle Aged , Obesity/blood , Obesity/physiopathology , Physical Endurance , Prospective Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL