1
|
Jakubowski H, Witucki Ł. Homocysteine Metabolites, Endothelial Dysfunction, and Cardiovascular Disease. Int J Mol Sci 2025; 26:746. [PMID: 39859460 PMCID: PMC11765536 DOI: 10.3390/ijms26020746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Atherosclerosis is accompanied by inflammation that underlies cardiovascular disease (CVD) and its vascular manifestations, including acute stroke, myocardial infarction, and peripheral artery disease, the leading causes of morbidity/mortality worldwide. The monolayer of endothelial cells formed on the luminal surface of arteries and veins regulates vascular tone and permeability, which supports vascular homeostasis. Endothelial dysfunction, the first step in the development of atherosclerosis, is caused by mechanical and biochemical factors that disrupt vascular homeostasis and induce inflammation. Together with increased plasma levels of low-density lipoprotein (LDL), diabetes, hypertension, cigarette smoking, infectious microorganisms, and genetic factors, epidemiological studies established that dysregulated metabolism of homocysteine (Hcy) causing hyperhomocysteinemia (HHcy) is associated with CVD. Patients with severe HHcy exhibit severe CVD and die prematurely due to vascular complications. Biochemically, HHcy is characterized by elevated levels of Hcy and related metabolites such as Hcy-thiolactone and N-Hcy-protein, seen in genetic and nutritional deficiencies in Hcy metabolism in humans and animals. The only known source of Hcy in humans is methionine released in the gut from dietary protein. Hcy is generated from S-adenosylhomocysteine (AdoHcy) and metabolized to cystathionine by cystathionine β-synthase (CBS) and to Hcy-thiolactone by methionyl-tRNA synthetase. Hcy-thiolactone, a chemically reactive thioester, modifies protein lysine residues, generating N-homocysteinylated (N-Hcy)-protein. N-Hcy-proteins lose their normal native function and become cytotoxic, autoimmunogenic, proinflammatory, prothrombotic, and proatherogenic. Accumulating evidence, discussed in this review, shows that these Hcy metabolites can promote endothelial dysfunction, CVD, and stroke in humans by inducing pro-atherogenic changes in gene expression, upregulating mTOR signaling, and inhibiting autophagy through epigenetic mechanisms involving specific microRNAs, histone demethylase PHF8, and methylated histone H4K20me1. Clinical studies, also discussed in this review, show that cystathionine and Hcy-thiolactone are associated with myocardial infarction and ischemic stroke by influencing blood clotting. These findings contribute to our understanding of the complex mechanisms underlying endothelial dysfunction, atherosclerosis, CVD, and stroke and identify potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Microbiology, Biochemistry and Molecular Genetics, International Center for Public Health, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, 60-632 Poznań, Poland;
| | - Łukasz Witucki
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, 60-632 Poznań, Poland;
| |
Collapse
|
2
|
Marroncini G, Martinelli S, Menchetti S, Bombardiere F, Martelli FS. Hyperhomocysteinemia and Disease-Is 10 μmol/L a Suitable New Threshold Limit? Int J Mol Sci 2024; 25:12295. [PMID: 39596358 PMCID: PMC11594664 DOI: 10.3390/ijms252212295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Hyperhomocysteinemia (HHcy) is a medical condition characterized by an abnormally high level of homocysteine (Hcy) in the blood. Homocysteine is a toxic sulfur-containing amino acid that is produced during the metabolism of methionine. Under normal circumstances, Hcy is recycled back to methionine via the remethylation pathway, through the action of various enzymes and vitamins, particularly folic acid (vitamin B9) and B12 used when intracellular methionine levels are low, thus restoring the necessary levels to correctly maintain active protein synthesis. A second pathway, used in cases of intracellular methionine excess, (the trans-sulfuration pathway) is the one that recycles Hcy into cysteine (a precursor of glutathione), first passing through cystathionine (via the enzyme cystathionine beta-synthase), a reaction that requires vitamin B6 in its active form. HHcy has been identified as a risk factor for a variety of disorders, including cardiovascular diseases, multiple sclerosis, diabetes, Alzheimer's and Parkinson's diseases, osteoporosis and cancer. However, it remains unclear whether the slightly elevated concentration of Hcy (Hcy 7-10 μmol/L) is a causative factor or simply a marker of these pathologies. In human plasma, the concentration of Hcy ([Hcy]) is classified as mild (15 to 30 μmol/L), moderate (30 to 100 μmol/L), and severe (greater than 100 μmol/L). Interestingly, many laboratories continue to consider 25 μmol/L as normal. This review seeks to examine the controversial literature regarding the normal range of HHcy and emphasizes that even a [Hcy] level of 10 μmol/L may contribute to the development of several diseases, aiming to discuss whether it would be appropriate to lower the threshold of HHcy normal values.
Collapse
Affiliation(s)
- Giada Marroncini
- Biomolecular Diagnostic Laboratories, Via N. Porpora, 50144 Florence, Italy; (S.M.); (F.B.); (F.S.M.)
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy;
| | - Serena Martinelli
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy;
| | - Sara Menchetti
- Biomolecular Diagnostic Laboratories, Via N. Porpora, 50144 Florence, Italy; (S.M.); (F.B.); (F.S.M.)
| | - Francesco Bombardiere
- Biomolecular Diagnostic Laboratories, Via N. Porpora, 50144 Florence, Italy; (S.M.); (F.B.); (F.S.M.)
| | | |
Collapse
|
3
|
Ren L, Pushpakumar S, Almarshood H, Das SK, Sen U. Epigenetic DNA Methylation and Protein Homocysteinylation: Key Players in Hypertensive Renovascular Damage. Int J Mol Sci 2024; 25:11599. [PMID: 39519150 PMCID: PMC11546175 DOI: 10.3390/ijms252111599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Hypertension has been a threat to the health of people, the mechanism of which, however, remains poorly understood. It is clinically related to loss of nephron function, glomerular sclerosis, or necrosis, resulting in renal functional declines. The mechanisms underlying hypertension's development and progression to organ damage, including hypertensive renal damage, remain to be fully elucidated. As a developing approach, epigenetics has been postulated to elucidate the phenomena that otherwise cannot be explained by genetic studies. The main epigenetic hallmarks, such as DNA methylation, histone acetylation, deacetylation, noncoding RNAs, and protein N-homocysteinylation have been linked with hypertension. In addition to contributing to endothelial dysfunction and oxidative stress, biologically active gases, including NO, CO, and H2S, are crucial regulators contributing to vascular remodeling since their complex interplay conducts homeostatic functions in the renovascular system. Importantly, epigenetic modifications also directly contribute to the pathogenesis of kidney damage via protein N-homocysteinylation. Hence, epigenetic modulation to intervene in renovascular damage is a potential therapeutic approach to treat renal disease and dysfunction. This review illustrates some of the epigenetic hallmarks and their mediators, which have the ability to diminish the injury triggered by hypertension and renal disease. In the end, we provide potential therapeutic possibilities to treat renovascular diseases in hypertension.
Collapse
Affiliation(s)
- Lu Ren
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Hebah Almarshood
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Swapan K. Das
- Department of Internal Medicine, Section on Endocrinology and Metabolism, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| |
Collapse
|
4
|
Witucki Ł, Jakubowski H. Homocysteine metabolites impair the PHF8/H4K20me1/mTOR/autophagy pathway by upregulating the expression of histone demethylase PHF8-targeting microRNAs in human vascular endothelial cells and mice. FASEB J 2024; 38:e70072. [PMID: 39323294 DOI: 10.1096/fj.202302116r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/21/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
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.
Collapse
Affiliation(s)
- Łukasz Witucki
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| | - Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, International Center for Public Health, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
5
|
Ding N, Ma S, Chang Q, Xie L, Li G, Hao Y, Xiong J, Yang A, Yang X, Jiang Y, Zhang H. Novel long noncoding lncARF mediated hyperhomocysteinemia-induced atherosclerosis via autophagy inhibition in foam cells. J Adv Res 2024:S2090-1232(24)00373-4. [PMID: 39214417 DOI: 10.1016/j.jare.2024.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Homocysteine (Hcy) is well recognized to be an independent risk factor for atherosclerosis. Long non-coding RNAs (lncRNAs) are emerging regulators of pathophysiological processes including atherosclerosis, while the underlying mechanisms of its involvement in Hcy induced-atherosclerosis remain largely unknown. OBJECTIVES The primary aim of this study is to assess the role of lncARF (autophagy-related factor induced by Hcy) in Hcy induced-atherosclerosis and related mechanism. METHODS RNA sequencing of foam cells treated with Hcy revealed a novel specific long noncoding RNA called lncARF. Locked nucleic acid gapmeRs-mediated lncARF knockdown was used to explore the role of lncARF both in vivo and in vitro. Mass spectrometry, RNA pull-down and RNA immunoprecipitation (RIP) assays were employed to uncover a mechanistic role of lncARF. Mass array assay and chromatin immunoprecipitation (ChIP) were used to detect the transcriptional activation of lncARF mediated by transcription factor. Clinically, receiver operating characteristic (ROC) curve analysis was used to assess the diagnostic value of lncARF in atherosclerotic patients with hyperhomocysteinemia (HHcy). RESULTS We observed that the expression of lncARF was substantially upregulated in atherosclerotic plaques, and knockdown of lncARF decreased the formation of atherosclerotic lesions by promoting autophagy in foam cells. Mechanistically, lncARF physically binds to RRAGD and inhibits its ubiquitination, further activating the PI3K/Akt and MAPK signaling pathways. Moreover, in vitro experiments showed that transcription factor FosB inhibited the binding of DNMT1 at the lncARF promoter, leading to transcriptional activation through DNA hypomethylation. Clinically, lncARF expression was positively correlated with serum Hcy levels, and it could distinguish atherosclerotic patients with HHcy with a high area under the ROC curve, sensitivity and specificity. CONCLUSIONS Our study highlights the mechanisms of lncARF in protecting against the development of atherosclerosis involving the epigenetic modifications and RRAGD/PI3K/Akt and RRAGD/MAPK signaling pathways, which may provide novel diagnostic biomarkers to improve atherosclerosis treatment.
Collapse
Affiliation(s)
- Ning Ding
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China; School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Shengchao Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China; School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Qingning Chang
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China; General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Lin Xie
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China; School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Guizhong Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China; School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Yinju Hao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China; General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Jiantuan Xiong
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China; School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Anning Yang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China; School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xiaoling Yang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China; School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Yideng Jiang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China; School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China.
| | - Huiping Zhang
- Department of Medical Genetics, Hunan Provincial Maternal and Child Health Hospital, Changsha 410008, China; Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan 750004, China; General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
6
|
Jakubowski H. Homocysteine Thiolactone Detoxifying Enzymes and Alzheimer's Disease. Int J Mol Sci 2024; 25:8095. [PMID: 39125665 PMCID: PMC11312131 DOI: 10.3390/ijms25158095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
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.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, University of Life Sciences, 60-637 Poznań, Poland; ; Tel.: +48-973-972-8733; Fax: +48-973-972-8981
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, International Center for Public Health, Newark, NJ 07103, USA
| |
Collapse
|
7
|
Zhang Q, Wu C, Tan X, Li C, Liu Y, Hu S. Homocysteine Facilitates the Formation of Carotid Atherosclerotic Plaque Through Inflammatory and Noninflammatory Mechanisms. Metab Syndr Relat Disord 2024; 22:365-371. [PMID: 38422209 DOI: 10.1089/met.2023.0293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
Background: Elevated homocysteine (Hcy) was considered a significant risk factor in the development and progression of carotid atherosclerosis (CAS), which involves a combination of inflammatory and noninflammatory mechanisms. However, epidemiological surveys have presented conflicting results. In this study, we aim to offer an epidemiological viewpoint on how elevated Hcy impacts CAS and its potential mechanisms. Methods: Levels of high-sensitivity C-reactive protein (hsCRP) were measured to assess the inflammatory status. The estimation of CAS events was performed by assessing carotid intima-media thickness using Doppler ultrasonography. Univariate analysis was conducted to investigate the variations in biochemical parameters among three groups: normal, carotid atherosclerotic thickening (CAT), and carotid atherosclerotic plaque (CAP) formation. Logistic regression analysis was conducted to identify the risk factors associated with the progression of CAT and CAP. In addition, multivariate linear regression analysis was conducted to identify the independent factors that correlated with hsCRP levels. Results: The study encompassed 3897 participants, with 2992 (76.8%) being males and 905 (23.2%) being females. The incidence of CAT and CAP rose with higher Hcy levels, with an overall odds ratio (OR) of 2.04 [95% confidence intervals (CI) 1.69-2.40] for CAT and 2.68 (95% CI 2.32-3.05) for CAP. After adjusting for gender, age, and blood markers, the OR for CAT and CAP decreased, with an overall OR of 1.05 (95% CI 0.81-1.28) and OR of 1.24 (95% CI 1.02-1.46), respectively. CAP risk independently increased when Hcy level exceeded 19.7 μmol/L (P = 0.030), but not CAT risk (P = 0.299). The impact of hsCRP on CAS events is similar to that of Hcy, and a multiple linear analysis found a significant independent correlation between hsCRP and Hcy (P = 0.001). Conclusions: Elevated Hcy levels can facilitate the formation of CAP through both inflammatory and noninflammatory processes, but it does not independently influence CAT.
Collapse
Affiliation(s)
- Qiang Zhang
- Health Management Center, Chongqing General Hospital, Chongqing, China
| | - Chunxi Wu
- Department of Blood Transfusion, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xiaoqing Tan
- Health Management Center, Chongqing General Hospital, Chongqing, China
| | - Can Li
- Health Management Center, Chongqing General Hospital, Chongqing, China
| | - Ying Liu
- Health Management Center, Chongqing General Hospital, Chongqing, China
| | - Shixia Hu
- Health Management Center, Chongqing General Hospital, Chongqing, China
| |
Collapse
|
8
|
Sircana MC, Erre GL, Castagna F, Manetti R. Crosstalk between Inflammation and Atherosclerosis in Rheumatoid Arthritis and Systemic Lupus Erythematosus: Is There a Common Basis? Life (Basel) 2024; 14:716. [PMID: 38929699 PMCID: PMC11204900 DOI: 10.3390/life14060716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in patients with rheumatoid arthritis and systemic lupus erythematosus. Traditional cardiovascular risk factors, although present in lupus and rheumatoid arthritis, do not explain such a high burden of early cardiovascular disease in the context of these systemic connective tissue diseases. Over the past few years, our understanding of the pathophysiology of atherosclerosis has changed from it being a lipid-centric to an inflammation-centric process. In this review, we examine the pathogenesis of atherosclerosis in systemic lupus erythematosus and rheumatoid arthritis, the two most common systemic connective tissue diseases, and consider them as emblematic models of the effect of chronic inflammation on the human body. We explore the roles of the inflammasome, cells of the innate and acquired immune system, neutrophils, macrophages, lymphocytes, chemokines and soluble pro-inflammatory cytokines in rheumatoid arthritis and systemic lupus erythematosus, and the roles of certain autoantigens and autoantibodies, such as oxidized low-density lipoprotein and beta2-glycoprotein, which may play a pathogenetic role in atherosclerosis progression.
Collapse
Affiliation(s)
| | | | | | - Roberto Manetti
- Department of Medical, Surgical and Pharmacology, University of Sassari, 07100 Sassari, Italy; (G.L.E.); (F.C.)
| |
Collapse
|
9
|
Witucki Ł, Jakubowski H. Homocysteine metabolites inhibit autophagy by upregulating miR-21-5p, miR-155-5p, miR-216-5p, and miR-320c-3p in human vascular endothelial cells. Sci Rep 2024; 14:7151. [PMID: 38531978 DOI: 10.1038/s41598-024-57750-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 03/21/2024] [Indexed: 03/28/2024] Open
Abstract
Nutritional and genetic deficiencies in homocysteine (Hcy) metabolism lead to hyperhomocysteinemia (HHcy) and cause endothelial dysfunction, a hallmark of atherosclerosis, which is a major cause of cardiovascular disease (CVD). Impaired autophagy causes the accumulation of damaged proteins and organelles and is associated with CVD. Biochemically, HHcy is characterized by elevated levels of Hcy and its metabolites, Hcy-thiolactone and N-Hcy-protein. However, whether these metabolites can dysregulate mTOR signaling and autophagy in endothelial cells is not known. Here, we examined the influence of Hcy-thiolactone, N-Hcy-protein, and Hcy on autophagy human umbilical vein endothelial cells. We found that treatments with Hcy-thiolactone, N-Hcy-protein, or Hcy significantly downregulated beclin 1 (BECN1), autophagy-related 5 (ATG5), autophagy-related 7 (ATG7), and microtubule-associated protein 1 light chain 3 (LC3) mRNA and protein levels. We also found that these changes were mediated by upregulation by Hcy-thiolactone, N-Hcy-protein, and Hcy of autophagy-targeting microRNA (miR): miR-21, miR-155, miR-216, and miR-320c. The effects of these metabolites on levels of miR targeting autophagy as well as on the levels of BECN1, ATG5, ATG7, and LC3 mRNA and protein were abrogated by treatments with inhibitors of miR-21, miR-155, miR-216, and mir320c. Taken together, our findings show that Hcy metabolites can upregulate miR-21, miR-155, miR-216, and mir320c, which then downregulate autophagy in human endothelial cells, important for vascular homeostasis.
Collapse
Affiliation(s)
- Łukasz Witucki
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, 60-632, Poznań, Poland
| | - Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, 60-632, Poznań, Poland.
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, International Center for Public Health, Rutgers University, 225 Warren Street, Newark, NJ, 07103, USA.
| |
Collapse
|
10
|
Demir Özer E, Yildirim M. The modification of nisin with homocysteine thiolactone and its effect on antimicrobial activity. Braz J Microbiol 2024; 55:191-199. [PMID: 38082122 PMCID: PMC10920495 DOI: 10.1007/s42770-023-01207-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/02/2023] [Indexed: 03/09/2024] Open
Abstract
The aim of the present study is to make an important contribution to the literature by focusing on the preparation of the N-homocysteine conjugate of nisin and evaluating the effect of the N-homocysteinylation reaction on its antimicriobial activity. The modification process was monitored using both acetic acid urea polyacrylamide gel electrophoresis (AAU-PAGE) and tricine sodium dodecyl sulphate polyacrylamide gel electrophoresis (tricine SDS-PAGE). The antibacterial effectiveness of modified nisin was assessed against Staphylococcus aureus ATCC 6538, Enterococcus faecium ATCC 9097, Bacillus subtilis ATCC 6633, Lactococcus lactis ssp. cremoris AÜ, Listeria monocytogenes NCTC 5348, and Escherichia coli RSKK. Optimal conditions for achieving the highest N-homocysteinylation degree (6.30%) were determined as 6 mg/mL nisin, 150 mM homocysteine thiolactone, 150 rpm shaking rate, pH of 3.0, and a reaction time of 6 h. The modified nisin obtained did not have a significant inhibitory effect on the strains tested except E. faecium. E. faecium was inhibited by the modified nisin and its antibacterial activity was determined as approximately 10% of the antibacterial activity of unmodified nisin. On the other hand, hydrolysis of nisin by trypsin and thermolysin resulted in significant specific side chain modifications induced by the homocysteine-thiolactone reaction, especially at Lys12 and Lys22. The results provide valuable insights into the potential of N-homocysteinylation to improve the antibacterial properties of nisin and also suggest that the effects of specific modifications identified during the modification process should be investigated.
Collapse
Affiliation(s)
- Ezgi Demir Özer
- Department of Gastronomy and Culinary Arts, Cappadocia University, Nevşehir, Turkey.
| | - Metin Yildirim
- Department of Food Engineering, Niğde Ömer Halisdemir University, Niğde, Turkey
| |
Collapse
|
11
|
Witucki Ł, Jakubowski H. Homocysteine metabolites inhibit autophagy, elevate amyloid beta, and induce neuropathy by impairing Phf8/H4K20me1-dependent epigenetic regulation of mTOR in cystathionine β-synthase-deficient mice. J Inherit Metab Dis 2023; 46:1114-1130. [PMID: 37477632 DOI: 10.1002/jimd.12661] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/10/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
The loss of cystathionine β-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 β (Aβ) by confocal fluorescence microscopy using anti-Aβ 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 Aβ levels. The Phf8 gene silencing increased Aβ, but not APP, levels. Taken together, our findings identify Phf8 as a regulator of Aβ synthesis and suggest that neuropathy of Cbs deficiency is mediated by Hcy metabolites, which transcriptionally dysregulate the Phf8 → H4K20me1 → mTOR → autophagy pathway thereby increasing Aβ accumulation.
Collapse
Affiliation(s)
- Łukasz Witucki
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, New Jersey Medical School, Newark, New Jersey, USA
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| | - Hieronim Jakubowski
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, New Jersey Medical School, Newark, New Jersey, USA
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| |
Collapse
|
12
|
Petrović DJ, Jagečić D, Krasić J, Sinčić N, Mitrečić D. Effect of Fetal Bovine Serum or Basic Fibroblast Growth Factor on Cell Survival and the Proliferation of Neural Stem Cells: The Influence of Homocysteine Treatment. Int J Mol Sci 2023; 24:14161. [PMID: 37762465 PMCID: PMC10531752 DOI: 10.3390/ijms241814161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
In vitro cell culture is a routinely used method which is also applied for in vitro modeling of various neurological diseases. On the other hand, media used for cell culture are often not strictly standardized between laboratories, which hinders the comparison of the obtained results. Here, we compared the effects of homocysteine (Hcy), a molecule involved in neurodegeneration, on immature cells of the nervous system cultivated in basal medium or media supplemented by either fetal bovine serum or basic fibroblast growth factor. The number of cells in basal media supplemented with basic fibroblast growth factor (bFGF) was 2.5 times higher in comparison to the number of cells in basal media supplemented with fetal bovine serum (FBS). We also found that the neuron-specific β-3-tubulin protein expression dose dependently decreased with increasing Hcy exposure. Interestingly, bFGF exerts a protective effect on β-3-tubulin protein expression at a concentration of 1000 µM Hcy compared to FBS-treated neural stem cells on Day 7. Supplementation with bFGF increased SOX2 protein expression two-fold compared to FBS supplementation. GFAP protein expression increased five-fold on Day 3 in FBS-treated neural stem cells, whereas on Day 7, bFGF increased GFAP expression two-fold compared to FBS-treated neural stem cells. Here, we have clearly shown that the selection of culturing media significantly influences various cellular parameters, which, in turn, can lead to different conclusions in experiments based on in vitro models of pathological conditions.
Collapse
Affiliation(s)
- Dražen Juraj Petrović
- Laboratory for Stem Cells, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia; (D.J.P.); (D.J.)
- Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Glycoscience Research Laboratory, Genos Ltd., 10000 Zagreb, Croatia
- BIMIS—Biomedical Research Center Šalata, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Denis Jagečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia; (D.J.P.); (D.J.)
- Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- BIMIS—Biomedical Research Center Šalata, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Jure Krasić
- Laboratory for Stem Cells, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia; (D.J.P.); (D.J.)
- BIMIS—Biomedical Research Center Šalata, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Laboratory for Neurogenomics and In Situ Hybridization, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Nino Sinčić
- BIMIS—Biomedical Research Center Šalata, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers (epiMark), Department of Medical Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Dinko Mitrečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia; (D.J.P.); (D.J.)
- Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- BIMIS—Biomedical Research Center Šalata, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
13
|
Loubane G, Robert G, Firdaus SB, Venne P, Comeau C, Boudreault PL, Komba JE, Wagner JR, Naylor S, Klarskov K. Conundrum of dehydroascorbic acid and homocysteine thiolactone reaction products: Structural characterization and effect on peptide and protein N-homocysteinylation. Free Radic Biol Med 2023; 206:111-124. [PMID: 37385568 DOI: 10.1016/j.freeradbiomed.2023.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
An excessive blood level of homocysteine (HcySH) is associated with numerous cardiovascular and neurodegenerative disease conditions. It has been suggested that direct S-homocysteinylation, of proteins by HcySH, or N-homosteinylation by homocysteine thiolactone (HTL) could play a causative role in these maladies. In contrast, ascorbic acid (AA) plays a significant role in oxidative stress prevention. AA is oxidized to dehydroascorbic acid (DHA) and if not rapidly reduced back to AA may degrade to reactive carbonyl products. In the present work, DHA is shown to react with HTL to produce a spiro bicyclic ring containing a six-membered thiazinane-carboxylic acid moiety. This reaction product is likely formed by initial imine condensation and subsequent hemiaminal product followed by HTL ring opening and intramolecular nucleophilic attack of the resulting thiol anion to form the spiro product. The reaction product was determined to have an accurate mass of 291.0414 and a molecular composition C10H13NO7S containing five double bond equivalents. We structurally characterized the reaction product using a combination of accurate mass tandem mass spectrometry, 1D and 2D-nuclear magnetic resonance. We also demonstrated that formation of the reaction product prevented peptide and protein N-homocysteinylation by HTL using a model peptide and α-lactalbumin. Furthermore, the reaction product is formed in Jurkat cells when exposed to HTL and DHA.
Collapse
Affiliation(s)
- Ghizlane Loubane
- Département de Pharmacologie et Physiologie Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 2160 San Fernando Drive, Milwaukee, WI, 53122, USA
| | - Gabriel Robert
- Département de Médecine Nucléaire et Radiobiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 2160 San Fernando Drive, Milwaukee, WI, 53122, USA
| | - Syed Benazir Firdaus
- Département de Pharmacologie et Physiologie Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 2160 San Fernando Drive, Milwaukee, WI, 53122, USA
| | - Philippe Venne
- Département de Chimie, 2160 San Fernando Drive, Milwaukee, WI, 53122, USA
| | - Christian Comeau
- Département de Chimie, 2160 San Fernando Drive, Milwaukee, WI, 53122, USA
| | | | - Jeampy E Komba
- Département de Pharmacologie et Physiologie Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 2160 San Fernando Drive, Milwaukee, WI, 53122, USA
| | - J Richard Wagner
- Département de Médecine Nucléaire et Radiobiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 2160 San Fernando Drive, Milwaukee, WI, 53122, USA
| | - Stephen Naylor
- ReNeuroGen LLC, 2160 San Fernando Drive, Milwaukee, WI, 53122, USA
| | - Klaus Klarskov
- Département de Pharmacologie et Physiologie Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 2160 San Fernando Drive, Milwaukee, WI, 53122, USA.
| |
Collapse
|
14
|
Zhao R, Cao L, Gu WJ, Li L, Chen ZZ, Xiang J, Zhou ZY, Xu B, Zang WD, Zhou XY, Cao J, Sun K, Zhao JY. Gestational palmitic acid suppresses embryonic GATA-binding protein 4 signaling and causes congenital heart disease. Cell Rep Med 2023; 4:100953. [PMID: 36809766 PMCID: PMC10040382 DOI: 10.1016/j.xcrm.2023.100953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/13/2022] [Accepted: 01/31/2023] [Indexed: 02/22/2023]
Abstract
Dysregulated maternal fatty acid metabolism increases the risk of congenital heart disease (CHD) in offspring with an unknown mechanism, and the effect of folic acid fortification in preventing CHD is controversial. Using gas chromatography coupled to either a flame ionization detector or mass spectrometer (GC-FID/MS) analysis, we find that the palmitic acid (PA) concentration increases significantly in serum samples of pregnant women bearing children with CHD. Feeding pregnant mice with PA increased CHD risk in offspring and cannot be rescued by folic acid supplementation. We further find that PA promotes methionyl-tRNA synthetase (MARS) expression and protein lysine homocysteinylation (K-Hcy) of GATA4 and results in GATA4 inhibition and abnormal heart development. Targeting K-Hcy modification by either genetic ablation of Mars or using N-acetyl-L-cysteine (NAC) decreases CHD onset in high-PA-diet-fed mice. In summary, our work links maternal malnutrition and MARS/K-Hcy with the onset of CHD and provides a potential strategy in preventing CHD by targeting K-Hcy other than folic acid supplementation.
Collapse
Affiliation(s)
- Rui Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Li Cao
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, and Department of Materials Science, Fudan University, Shanghai 200438, China
| | - Wen-Jun Gu
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, and Department of Materials Science, Fudan University, Shanghai 200438, China
| | - Lei Li
- Department of Anatomy and Neuroscience Research Institute, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhong-Zhong Chen
- Urogenital Development Research Center, Department of Urology, Shanghai Children's Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Jie Xiang
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, and Department of Materials Science, Fudan University, Shanghai 200438, China
| | - Ze-Yu Zhou
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, and Department of Materials Science, Fudan University, Shanghai 200438, China
| | - Bo Xu
- Department of Anesthesiology, General Hospital of Southern Theatre Command of People's Liberation Army, Guangzhou 510030, China
| | - Wei-Dong Zang
- Department of Anatomy and Neuroscience Research Institute, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiang-Yu Zhou
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences, and Department of Materials Science, Fudan University, Shanghai 200438, China.
| | - Jing Cao
- Department of Anatomy and Neuroscience Research Institute, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Kun Sun
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Anatomy and Neuroscience Research Institute, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; International Human Phenome Institutes (Shanghai), Shanghai 200433, China.
| |
Collapse
|
15
|
Ishii I, Bhatia M. Amino Acids in Health and Disease: The Good, the Bad, and the Ugly. Int J Mol Sci 2023; 24:4931. [PMID: 36902358 PMCID: PMC10002494 DOI: 10.3390/ijms24054931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
The Special Issue "Amino Acid Metabolism and Regulation in Health and Disease 2 [...].
Collapse
Affiliation(s)
- Isao Ishii
- Department of Health Chemistry, Showa Pharmaceutical University, Tokyo 194-8543, Japan
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand
| |
Collapse
|
16
|
Guéant JL, Guéant-Rodriguez RM, Oussalah A, Zuily S, Rosenberg I. Hyperhomocysteinemia in Cardiovascular Diseases: Revisiting Observational Studies and Clinical Trials. Thromb Haemost 2023; 123:270-282. [PMID: 36170884 DOI: 10.1055/a-1952-1946] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Thromboembolic manifestations are relatively frequent in patients with intermediate/severe hyperhomocysteinemia (>30 µmol/L) related to inherited disorders and deficiencies in vitamin B12 and folate. In contrast, moderate hyperhomocysteinemia (15-30 µmol/L) is a modest predictor of cardiovascular risk. The recognition of homocysteine as a cardiovascular risk factor has been challenged by some but not all randomized clinical trials. We reviewed the main data of this controversy and formulated conclusions to be translated in clinical practice.Homocysteine-lowering trials have been performed in cardiovascular subjects with moderate but not intermediate/severe hyperhomocysteinemia despite the dose-effect risk association. The first meta-analyses found no benefit and led cardiology societies not recommending homocysteine in the assessment of cardiovascular risk. This guideline challenged the need to diagnose and treat the nutritional and genetic causes of intermediate/major hyperhomocysteinemia and was not revised when larger meta-analyses concluded to a reduced risk of stroke. In a recent observational study, 84% of consecutive cardiovascular patients assessed for homocysteine had intermediate or major hyperhomocysteinemia, which was properly assessed in only half of the cases and related to B12 and/or folate deficiency and Addison/Biermer disease in 55% of these cases.In conclusion, revisiting observational studies and clinical trials suggests that cardiovascular patients should be screened for hyperhomocysteinemia, when no other risk factor is found. Patients with intermediate/major hyperhomocysteinemia should be properly assessed and treated for B vitamin deficiencies and inherited disorders according to current guidelines. Further trials are needed to assess the effect of lowering homocysteine according to hyperhomocysteinemia categories at baseline.
Collapse
Affiliation(s)
- Jean-Louis Guéant
- Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, Departments of Hepato-Gastroenterology and Molecular Medicine, University Hospital of Nancy, F-54000 Nancy, France.,Reference Centre for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, F-54000 Nancy, France.,INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, F-54000 Nancy, France
| | - Rosa-Maria Guéant-Rodriguez
- Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, Departments of Hepato-Gastroenterology and Molecular Medicine, University Hospital of Nancy, F-54000 Nancy, France.,Reference Centre for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, F-54000 Nancy, France.,INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, F-54000 Nancy, France
| | - Abderrahim Oussalah
- Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, Departments of Hepato-Gastroenterology and Molecular Medicine, University Hospital of Nancy, F-54000 Nancy, France.,Reference Centre for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, F-54000 Nancy, France.,INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, F-54000 Nancy, France
| | - Stéphane Zuily
- Vascular Medicine Division and Regional Competence Center for Rare Auto-Immune Diseases, INSERM UMR_S 1116 DCAC and CHRU-Nancy, Université de Lorraine, F-54000 Nancy, France
| | - Irwin Rosenberg
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts, United States
| |
Collapse
|
17
|
Sobieszczuk-Nowicka E, Arasimowicz-Jelonek M, Tanwar UK, Floryszak-Wieczorek J. Plant homocysteine, a methionine precursor and plant's hallmark of metabolic disorders. FRONTIERS IN PLANT SCIENCE 2022; 13:1044944. [PMID: 36570932 PMCID: PMC9773845 DOI: 10.3389/fpls.2022.1044944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Homocysteine (Hcy) is a sulfur-containing non-proteinogenic amino acid, which arises from redox-sensitive methionine metabolism. In plants, Hcy synthesis involves both cystathionine β-lyase and S-adenosylhomocysteine hydrolase activities. Thus, Hcy itself is crucial for de novo methionine synthesis and S-adenosylmethionine recycling, influencing the formation of ethylene, polyamines, and nicotianamine. Research on mammalian cells has shown biotoxicity of this amino acid, as Hcy accumulation triggers oxidative stress and the associated lipid peroxidation process. In addition, the presence of highly reactive groups induces Hcy and Hcy derivatives to modify proteins by changing their structure and function. Currently, Hcy is recognized as a critical, independent hallmark of many degenerative metabolic diseases. Research results indicate that an enhanced Hcy level is also toxic to yeast and bacteria cells. In contrast, in the case of plants the metabolic status of Hcy remains poorly examined and understood. However, the presence of the toxic Hcy metabolites and Hcy over-accumulation during the development of an infectious disease seem to suggest harmful effects of this amino acid also in plant cells. The review highlights potential implications of Hcy metabolism in plant physiological disorders caused by environmental stresses. Moreover, recent research advances emphasize that recognizing the Hcy mode of action in various plant systems facilitates verification of the potential status of Hcy metabolites as bioindicators of metabolism disorders and thus may constitute an element of broadly understood biomonitoring.
Collapse
Affiliation(s)
- Ewa Sobieszczuk-Nowicka
- Department of Plant Physiology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | | | - Umesh Kumar Tanwar
- Department of Plant Physiology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | | |
Collapse
|
18
|
Tabibzadeh S. Resolving Geroplasticity to the Balance of Rejuvenins and Geriatrins. Aging Dis 2022; 13:1664-1714. [PMID: 36465174 PMCID: PMC9662275 DOI: 10.14336/ad.2022.0414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/14/2022] [Indexed: 09/29/2024] Open
Abstract
According to the cell centric hypotheses, the deficits that drive aging occur within cells by age dependent progressive damage to organelles, telomeres, biologic signaling pathways, bioinformational molecules, and by exhaustion of stem cells. Here, we amend these hypotheses and propose an eco-centric model for geroplasticity (aging plasticity including aging reversal). According to this model, youth and aging are plastic and require constant maintenance, and, respectively, engage a host of endogenous rejuvenating (rejuvenins) and gero-inducing [geriatrin] factors. Aging in this model is akin to atrophy that occurs as a result of damage or withdrawal of trophic factors. Rejuvenins maintain and geriatrins adversely impact cellular homeostasis, cell fitness, and proliferation, stem cell pools, damage response and repair. Rejuvenins reduce and geriatrins increase the age-related disorders, inflammatory signaling, and senescence and adjust the epigenetic clock. When viewed through this perspective, aging can be successfully reversed by supplementation with rejuvenins and by reducing the levels of geriatrins.
Collapse
Affiliation(s)
- Siamak Tabibzadeh
- Frontiers in Bioscience Research Institute in Aging and Cancer, Irvine, CA 92618, USA
| |
Collapse
|
19
|
Relevance of Plasma Homocysteine and Methylenetetrahydrofolate Reductase 677TT Genotype in Sickle Cell Disease: A Systematic Review and Meta-Analysis. Int J Mol Sci 2022; 23:ijms232314641. [PMID: 36498990 PMCID: PMC9736045 DOI: 10.3390/ijms232314641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022] Open
Abstract
We evaluated the relevance of plasma homocysteine (HC) and the TT genotype of the methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism (rs1801133) in sickle cell disease (SCD) and associated vaso-occlusive crisis (VOC) and ischemic stroke (IS). We identified in Embase and Medline 22 studies on plasma HC and 22 on MTHFR genotypes. Due to age-related HC differences, adult and paediatric SCD were separated: 879 adult SCD and 834 controls (CTR) yielded a neutral effect size; 427 paediatric SCD and 625 CTR favoured SCD (p = 0.001) with wide heterogeneity (I2 = 95.5%) and were sub-grouped by country: six studies (Dutch Antilles n = 1, USA n = 5) yielded a neutral effect size, four (India n = 1, Arab countries n = 3) favoured SCD (p < 0.0001). Moreover, 249 SCD in VOC and 419 out of VOC yielded a neutral effect size. The pooled prevalence of the MTHFR TT genotype in 267 SCD equalled that of 1199 CTR (4.26% vs. 2.86%, p = 0.45), and in 84 SCD with IS equalled that of 86 without IS (5.9% vs. 3.7%, p = 0.47); removal of one paediatric study yielded a significant effect size (p = 0.006). Plasma HC in paediatric SCD from Middle East and India was higher, possibly due to vitamin deficiencies. Despite its low prevalence in SCD, the MTHFR TT genotype relates to adult IS.
Collapse
|
20
|
Wusiman W, Zhang Z, Ding Q, Liu M. The pathophyiological role of aminoacyl-tRNA synthetases in digestive system diseases. Front Physiol 2022; 13:935576. [PMID: 36017335 PMCID: PMC9396140 DOI: 10.3389/fphys.2022.935576] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/05/2022] [Indexed: 12/24/2022] Open
Abstract
Aminoacyl-tRNA synthetases (ARSs) catalyze the ligation of amino acids to their cognate transfer RNAs and are indispensable enzymes for protein biosynthesis in all the cells. Previously, ARSs were considered simply as housekeeping enzymes, however, they are now known to be involved in a variety of physiological and pathological processes, such as tumorigenesis, angiogenesis, and immune response. In this review, we summarize the role of ARSs in the digestive system, including the esophagus, stomach, small intestine, colon, as well as the auxiliary organs such as the pancreas, liver, and the gallbladder. Furthermore, we specifically focus on the diagnostic and prognostic value of ARSs in cancers, aiming to provide new insights into the pathophysiological implications of ARSs in tumorigenesis.
Collapse
Affiliation(s)
- Wugelanmu Wusiman
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zerui Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Ding
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mei Liu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Mei Liu,
| |
Collapse
|
21
|
Chen SM, Tang XQ. Homocysteinylation and Sulfhydration in Diseases. Curr Neuropharmacol 2022; 20:1726-1735. [PMID: 34951391 PMCID: PMC9881069 DOI: 10.2174/1570159x20666211223125448] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/02/2021] [Accepted: 12/18/2021] [Indexed: 11/22/2022] Open
Abstract
Homocysteine (Hcy) is an important intermediate in methionine metabolism and generation of one-carbon units, and its dysfunction is associated with many pathological states. Although Hcy is a non-protein amino acid, many studies have demonstrated protein-related homocysteine metabolism and possible mechanisms underlying homocysteinylation. Homocysteinylated proteins lose their original biological function and have a negative effect on the various disease phenotypes. Hydrogen sulfide (H2S) has been recognized as an important gaseous signaling molecule with mounting physiological properties. H2S modifies small molecules and proteins via sulfhydration, which is supposed to be essential in the regulation of biological functions and signal transduction in human health and disorders. This review briefly introduces Hcy and H2S, further discusses pathophysiological consequences of homocysteine modification and sulfhydryl modification, and ultimately makes a prediction that H2S might exert a protective effect on the toxicity of homocysteinylation of target protein via sulfhydration. The highlighted information here yields new insights into the role of protein modification by Hcy and H2S in diseases.
Collapse
Affiliation(s)
- Si-Min Chen
- Emergency Intensive Care Unit, Department of Emergency, Xiangtan Central Hospital, Xiangtan, 411100, Hunan, P.R. China; ,The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, P.R. China; ,Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, P.R. China
| | - Xiao-Qing Tang
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, P.R. China; ,Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, P.R. China,Address correspondence to this author at the The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China 69 Chuanshan Road, Hengyang 421001, Hunan Province, P.R. China; E-mails: ;
| |
Collapse
|
22
|
Wu Z, Jankowski V, Jankowski J. Irreversible post-translational modifications - Emerging cardiovascular risk factors. Mol Aspects Med 2022; 86:101010. [PMID: 34404548 DOI: 10.1016/j.mam.2021.101010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/19/2021] [Accepted: 08/12/2021] [Indexed: 12/23/2022]
Abstract
Despite the introduction of lipid-lowering drugs, antihypertensives, antiplatelet and anticoagulation therapies for primary prevention of cardiovascular and heart diseases (CVD), it remains the number one cause of death globally, raising the question for novel/further essential factors besides traditional risk factors such as cholesterol, blood pressure and coagulation. With continuous identification and characterization of non-enzymatic post-translationally modified isoforms of proteins and lipoproteins, it is becoming increasingly clear that irreversible non-enzymatic post-translational modifications (nPTMs) alter the biological functions of native proteins and lipoproteins thereby transforming innate serum components into CVD mediators. In particular renal insufficiency and metabolic imbalance are major contributors to the systemically increased concentration of reactive metabolites and thus increased frequency of nPTMs, promoting multi-morbid disease development centering around cardiovascular disease. nPTMs are significantly involved in the onset and progression of cardiovascular disease and represent a significant and novel risk factor. These insights represent potentially new avenues for risk assessment, prevention and therapy. This review chapter summarizes all forms of nPTMs found in CKD and under metabolic imbalance and discusses the biochemical connections between molecular alterations and the pathological impact on increased cardiovascular risk, novel nPTM-associated non-traditional cardiovascular risk factors, and clinical implication of nPTM in cardiovascular disease.
Collapse
Affiliation(s)
- Zhuojun Wu
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, Maastricht, the Netherlands.
| |
Collapse
|
23
|
The Mighty Mitochondria Are Unifying Organelles and Metabolic Hubs in Multiple Organs of Obesity, Insulin Resistance, Metabolic Syndrome, and Type 2 Diabetes: An Observational Ultrastructure Study. Int J Mol Sci 2022; 23:ijms23094820. [PMID: 35563211 PMCID: PMC9101653 DOI: 10.3390/ijms23094820] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 12/25/2022] Open
Abstract
Mitochondria (Mt) are essential cellular organelles for the production of energy and thermogenesis. Mt also serve a host of functions in addition to energy production, which include cell signaling, metabolism, cell death, and aging. Due to the central role of Mt in metabolism as metabolic hubs, there has been renewed interest in how Mt impact metabolic pathways and multiple pathologies. This review shares multiple observational ultrastructural findings in multiple cells and organs to depict aberrant mitochondrial (aMt) remodeling in pre-clinical rodent models. Further, it is intended to show how remodeling of Mt are associated with obesity, insulin resistance, metabolic syndrome (MetS), and type 2 diabetes mellitus (T2DM). Specifically, Mt remodeling in hypertensive and insulin-resistant lean models (Ren2 rat models), lean mice with streptozotocin-induced diabetes, obesity models including diet-induced obesity, genetic leptin-deficient ob/ob, and leptin receptor-deficient db/db diabetic mice are examined. Indeed, aMt dysfunction and damage have been implicated in multiple pathogenic diseases. Manipulation of Mt such as the induction of Mt biogenesis coupled with improvement of mitophagy machinery may be helpful to remove leaky damaged aMt in order to prevent the complications associated with the generation of superoxide-derived reactive oxygen species and the subsequent reactive species interactome. A better understanding of Mt remodeling may help to unlock many of the mysteries in obesity, insulin resistance, MetS, T2DM, and the associated complications of diabetic end-organ disease.
Collapse
|
24
|
Hayden MR, Tyagi SC. Impaired Folate-Mediated One-Carbon Metabolism in Type 2 Diabetes, Late-Onset Alzheimer's Disease and Long COVID. MEDICINA (KAUNAS, LITHUANIA) 2021; 58:16. [PMID: 35056324 PMCID: PMC8779539 DOI: 10.3390/medicina58010016] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/25/2022]
Abstract
Impaired folate-mediated one-carbon metabolism (FOCM) is associated with many pathologies and developmental abnormalities. FOCM is a metabolic network of interdependent biosynthetic pathways that is known to be compartmentalized in the cytoplasm, mitochondria and nucleus. Currently, the biochemical mechanisms and causal metabolic pathways responsible for the initiation and/or progression of folate-associated pathologies have yet to be fully established. This review specifically examines the role of impaired FOCM in type 2 diabetes mellitus, Alzheimer's disease and the emerging Long COVID/post-acute sequelae of SARS-CoV-2 (PASC). Importantly, elevated homocysteine may be considered a biomarker for impaired FOCM, which is known to result in increased oxidative-redox stress. Therefore, the incorporation of hyperhomocysteinemia will be discussed in relation to impaired FOCM in each of the previously listed clinical diseases. This review is intended to fill gaps in knowledge associated with these clinical diseases and impaired FOCM. Additionally, some of the therapeutics will be discussed at this early time point in studying impaired FOCM in each of the above clinical disease states. It is hoped that this review will allow the reader to better understand the role of FOCM in the development and treatment of clinical disease states that may be associated with impaired FOCM and how to restore a more normal functional role for FOCM through improved nutrition and/or restoring the essential water-soluble B vitamins through oral supplementation.
Collapse
Affiliation(s)
- Melvin R. Hayden
- Departments of Internal Medicine, Endocrinology Diabetes and Metabolism Diabetes and Cardiovascular Disease Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Suresh C. Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| |
Collapse
|
25
|
Grazioli E, Romani A, Marrone G, Di Lauro M, Cerulli C, Urciuoli S, Murri A, Guerriero C, Tranchita E, Tesauro M, Parisi A, Di Daniele N, Noce A. Impact of Physical Activity and Natural Bioactive Compounds on Endothelial Dysfunction in Chronic Kidney Disease. Life (Basel) 2021; 11:841. [PMID: 34440585 PMCID: PMC8402113 DOI: 10.3390/life11080841] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/21/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic kidney disease (CKD) represents a world-wide public health problem. Inflammation, endothelial dysfunction (ED) and vascular calcifications are clinical features of CKD patients that increase cardiovascular (CV) mortality. CKD-related CV disease pathogenic mechanisms are not only associated with traditional factors such as arterial hypertension and dyslipidemia, but also with ED, oxidative stress and low-grade inflammation. The typical comorbidities of CKD contribute to reduce the performance and the levels of the physical activity in nephropathic patients compared to healthy subjects. Currently, the effective role of physical activity on ED is still debated, but the available few literature data suggest its positive contribution. Another possible adjuvant treatment of ED in CKD patients is represented by natural bioactive compounds (NBCs). Among these, minor polar compounds of extra virgin olive oil (hydroxytyrosol, tyrosol and oleocanthal), polyphenols, and vitamin D seem to exert a beneficial role on ED in CKD patients. The objective of the review is to evaluate the effectiveness of physical exercise protocols and/or NBCs on ED in CKD patients.
Collapse
Affiliation(s)
- Elisa Grazioli
- Department of Exercise, Human and Health Sciences, Foro Italico University of Rome, 00135 Rome, Italy; (E.G.); (C.C.); (A.M.); (E.T.); (A.P.)
| | - Annalisa Romani
- PHYTOLAB (Pharmaceutical, Cosmetic, Food Supplement, Technology and Analysis), DiSIA, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Florence, Italy; (A.R.); (S.U.)
| | - Giulia Marrone
- UOC of Internal Medicine—Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (C.G.); (N.D.D.)
| | - Manuela Di Lauro
- UOC of Internal Medicine—Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (C.G.); (N.D.D.)
| | - Claudia Cerulli
- Department of Exercise, Human and Health Sciences, Foro Italico University of Rome, 00135 Rome, Italy; (E.G.); (C.C.); (A.M.); (E.T.); (A.P.)
| | - Silvia Urciuoli
- PHYTOLAB (Pharmaceutical, Cosmetic, Food Supplement, Technology and Analysis), DiSIA, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Florence, Italy; (A.R.); (S.U.)
| | - Arianna Murri
- Department of Exercise, Human and Health Sciences, Foro Italico University of Rome, 00135 Rome, Italy; (E.G.); (C.C.); (A.M.); (E.T.); (A.P.)
| | - Cristina Guerriero
- UOC of Internal Medicine—Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (C.G.); (N.D.D.)
| | - Eliana Tranchita
- Department of Exercise, Human and Health Sciences, Foro Italico University of Rome, 00135 Rome, Italy; (E.G.); (C.C.); (A.M.); (E.T.); (A.P.)
| | - Manfredi Tesauro
- UOC of Internal Medicine—Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (C.G.); (N.D.D.)
| | - Attilio Parisi
- Department of Exercise, Human and Health Sciences, Foro Italico University of Rome, 00135 Rome, Italy; (E.G.); (C.C.); (A.M.); (E.T.); (A.P.)
| | - Nicola Di Daniele
- UOC of Internal Medicine—Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (C.G.); (N.D.D.)
| | - Annalisa Noce
- UOC of Internal Medicine—Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (C.G.); (N.D.D.)
| |
Collapse
|
26
|
Chu M, Teng J, Guo L, Wang Y, Zhang L, Gao J, Liu L. Mild hyperhomocysteinemia induces blood-brain barrier dysfunction but not neuroinflammation in the cerebral cortex and hippocampus of wild-type mice. Can J Physiol Pharmacol 2021; 99:847-856. [PMID: 34161158 DOI: 10.1139/cjpp-2020-0507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study explored the potential effects of mild hyperhomocysteinemia (HHcy) on the blood-brain barrier (BBB) and neuroinflammation. Seven-week-old male wild-type C57BL/6 mice were fed normal mouse chow (the control group) or a methionine-enriched diet (the HHcy group) for 14 weeks. Mice in the HHcy group exhibited a slight increase in serum Hcy levels (13.56 ± 0.61 μmol/L). Activation of the ERK signaling pathway, up-regulation of matrix metalloproteinase-9 (MMP-9), and degradation of tight junction proteins (occludin and claudin-5) were observed in both the cerebral cortex and hippocampus of mice with mild HHcy. However, microglia were not activated and the levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were not changed in either the cerebral cortex or hippocampus of mice with mild HHcy. Moreover, the signaling activity of STAT3 also did not differ significantly between the two groups. These findings demonstrate that the BBB is highly vulnerable to homocysteine insult. Even a slight increase in serum homocysteine levels up-regulates MMP-9 expression and disrupts the BBB integrity. Meanwhile, microglia activation or the STAT3 pathway might not contribute to the effects of mild HHcy on the brain.
Collapse
Affiliation(s)
- Min Chu
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Jijun Teng
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Lei Guo
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Yuyang Wang
- Department of Rehabilitation, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Liang Zhang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Jing Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Lijun Liu
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| |
Collapse
|
27
|
Morris G, Berk M, Walder K, O'Neil A, Maes M, Puri BK. The lipid paradox in neuroprogressive disorders: Causes and consequences. Neurosci Biobehav Rev 2021; 128:35-57. [PMID: 34118292 DOI: 10.1016/j.neubiorev.2021.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 04/27/2021] [Accepted: 06/06/2021] [Indexed: 02/07/2023]
Abstract
Chronic systemic inflammation is associated with an increased risk of cardiovascular disease in an environment of low low-density lipoprotein (LDL) and low total cholesterol and with the pathophysiology of neuroprogressive disorders. The causes and consequences of this lipid paradox are explored. Circulating activated neutrophils can release inflammatory molecules such as myeloperoxidase and the pro-inflammatory cytokines interleukin-1 beta, interleukin-6 and tumour necrosis factor-alpha. Since activated neutrophils are associated with atherosclerosis and cardiovascular disease and with major depressive disorder, bipolar disorder and schizophrenia, it seems reasonable to hypothesise that the inflammatory molecules released by them may act as mediators of the link between systemic inflammation and the development of atherosclerosis in neuroprogressive disorders. This hypothesis is tested by considering the association at a molecular level of systemic inflammation with increased LDL oxidation; increased small dense LDL levels; increased lipoprotein (a) concentration; secretory phospholipase A2 activation; cytosolic phospholipase A2 activation; increased platelet activation; decreased apolipoprotein A1 levels and function; decreased paroxonase-1 activity; hyperhomocysteinaemia; and metabolic endotoxaemia. These molecular mechanisms suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand
| | | |
Collapse
|
28
|
Fledderus J, Vanchin B, Rots MG, Krenning G. The Endothelium as a Target for Anti-Atherogenic Therapy: A Focus on the Epigenetic Enzymes EZH2 and SIRT1. J Pers Med 2021; 11:jpm11020103. [PMID: 33562658 PMCID: PMC7915331 DOI: 10.3390/jpm11020103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
Endothelial cell inflammatory activation and dysfunction are key events in the pathophysiology of atherosclerosis, and are associated with an elevated risk of cardiovascular events. Yet, therapies specifically targeting the endothelium and atherosclerosis are lacking. Here, we review how endothelial behaviour affects atherogenesis and pose that the endothelium may be an efficacious cellular target for antiatherogenic therapies. We discuss the contribution of endothelial inflammatory activation and dysfunction to atherogenesis and postulate that the dysregulation of specific epigenetic enzymes, EZH2 and SIRT1, aggravate endothelial dysfunction in a pleiotropic fashion. Moreover, we propose that commercially available drugs are available to clinically explore this postulation.
Collapse
Affiliation(s)
- Jolien Fledderus
- Medical Biology Section, Laboratory for Cardiovascular Regenerative Medicine, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands; (J.F.); (B.V.)
| | - Byambasuren Vanchin
- Medical Biology Section, Laboratory for Cardiovascular Regenerative Medicine, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands; (J.F.); (B.V.)
- Department Cardiology, School of Medicine, Mongolian National University of Medical Sciences, Jamyan St 3, Ulaanbaatar 14210, Mongolia
| | - Marianne G. Rots
- Epigenetic Editing, Medical Biology Section, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands;
| | - Guido Krenning
- Medical Biology Section, Laboratory for Cardiovascular Regenerative Medicine, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands; (J.F.); (B.V.)
- Correspondence: ; Tel.: +31-50-361-8043; Fax: +31-50-361-9911
| |
Collapse
|
29
|
Jung S, Joo NS, Kim YN, Choi BH. Cut-off value of serum homocysteine in relation to increase of coronary artery calcification. J Investig Med 2021; 69:345-350. [PMID: 33148632 PMCID: PMC7848052 DOI: 10.1136/jim-2020-001478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2020] [Indexed: 01/07/2023]
Abstract
A recent study reported that coronary artery calcification (CAC) and serum homocysteine were well associated; however, no report is available for the cut-off value of serum homocysteine according to increase of coronary-artery calcification volume score (CVS). The data of 469 out of 777 subjects in 1 health promotion center located in Seoul were selected after exclusion of the missing data of serum homocysteine and CVS. CVS was categorized into 2 groups: CVS=0 and CVS>0. Serum homocysteine according to the CVS groups was compared, and the cut-off value of serum homocysteine according to the increase of CVS (>0) was calculated using the receiver operating characteristic curve. Mean age was 54.5 years and the proportion of females was 22.2%. Mean serum homocysteine concentration and CVS were 11.2 μmol/L and 50.4, respectively. After adjustments for age and sex, serum homocysteine was associated with CVS (r=0.167, p=0.001), and Log(Homocysteine) also showed a significant difference according to the CVS groups. The cut-off value of serum homocysteine according to the increase of CVS (>0) was 9.45 μmol/L (area under the curve=0.569 (95% CI 0.512 to 0.625), p=0.015). The cut-off value of serum homocysteine was 9.45 μmol/L according to the increase of coronary-artery CVS.
Collapse
Affiliation(s)
- Susie Jung
- Department of Family Practice and Community Health, Ajou University School of Medicine, Suwon, South Korea
| | - Nam-Seok Joo
- Department of Family Practice and Community Health, Ajou University Hospital, Suwon, Gyeonggi-do, South Korea
| | - Yu-Na Kim
- Department of Family Practice and Community Health, Ajou University School of Medicine, Suwon, South Korea
| | - Beom-Hee Choi
- Functional Medicine Clinic, GCIMED, Seoul, South Korea
| |
Collapse
|
30
|
Jakubowski H. Proteomic exploration of cystathionine β-synthase deficiency: implications for the clinic. Expert Rev Proteomics 2021; 17:751-765. [PMID: 33320032 DOI: 10.1080/14789450.2020.1865160] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Homocystinuria due to cystathionine β-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.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences , Poznań, Poland.,Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University-New Jersey Medical School, International Center for Public Health , Newark, NJ USA
| |
Collapse
|
31
|
The importance of homocysteine in the development of cardiovascular complications in patients with rheumatoid arthritis. Reumatologia 2020; 58:282-288. [PMID: 33227081 PMCID: PMC7667944 DOI: 10.5114/reum.2020.99732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/16/2020] [Indexed: 12/26/2022] Open
Abstract
Rheumatoid arthritis (RA) not only leads to disability due to joint changes, but also significantly shortens the life expectancy of patients, mainly due to more frequent occurrence of heart attacks and strokes. Accelerated atherosclerosis in these patients is caused, among other factors, by high homocysteine (HCY) concentration in blood. Numerous studies have shown that treatment with vitamin B significantly reduces the concentration of HCY in blood, but does not reduce the risk of heart diseases. Recent studies have shown, however, that folic acid (FA) administration reduces the risk of stroke by 10–20%. Due to the fact that in patients with RA strokes are more frequent than in the general population and hyperhomocysteinemia (HHCY) is often found, determination of HCY concentration in blood is advisable, and in persons with HHCY it is recommended to use FA in primary and secondary stroke prevention.
Collapse
|
32
|
Burgess K, Bennett C, Mosnier H, Kwatra N, Bethel F, Jadavji NM. The Antioxidant Role of One-Carbon Metabolism on Stroke. Antioxidants (Basel) 2020; 9:E1141. [PMID: 33212887 PMCID: PMC7698340 DOI: 10.3390/antiox9111141] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
One-carbon (1C) metabolism is a metabolic network that is centered on folate, a B vitamin; it integrates nutritional signals with biosynthesis, redox homeostasis, and epigenetics. This metabolic pathway also reduces levels of homocysteine, a non-protein amino acid. High levels of homocysteine are linked to increased risk of hypoxic events, such as stroke. Several preclinical studies have suggested that 1C metabolism can impact stroke outcome, but the clinical data are unclear. The objective of this paper was to review preclinical and clinical research to determine whether 1C metabolism has an antioxidant role on stroke. To accomplish the objective, we searched for publications using the following medical subject headings (MeSH) keywords: antioxidants, hypoxia, stroke, homocysteine, one-carbon metabolism, folate, methionine, and dietary supplementation of one-carbon metabolism. Both pre-clinical and clinical studies were retrieved and reviewed. Our review of the literature suggests that deficiencies in 1C play an important role in the onset and outcome of stroke. Dietary supplementation of 1C provides beneficial effects on stroke outcome. For stroke-affected patients or individuals at high risk for stroke, the data suggest that nutritional modifications in addition to other therapies could be incorporated into a treatment plan.
Collapse
Affiliation(s)
- Kassidy Burgess
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA;
- Biomedical Sciences Program, Midwestern University, Glendale, AZ 85308, USA; (C.B.); (N.K.); (F.B.)
| | - Calli Bennett
- Biomedical Sciences Program, Midwestern University, Glendale, AZ 85308, USA; (C.B.); (N.K.); (F.B.)
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Hannah Mosnier
- School of Medicine, National University of Ireland Galway, H91 TK33, Ireland;
- College of Dental Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Neha Kwatra
- Biomedical Sciences Program, Midwestern University, Glendale, AZ 85308, USA; (C.B.); (N.K.); (F.B.)
- College of Dental Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Forrest Bethel
- Biomedical Sciences Program, Midwestern University, Glendale, AZ 85308, USA; (C.B.); (N.K.); (F.B.)
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Nafisa M. Jadavji
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA;
- Biomedical Sciences Program, Midwestern University, Glendale, AZ 85308, USA; (C.B.); (N.K.); (F.B.)
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
33
|
Mei X, Qi D, Zhang T, Zhao Y, Jin L, Hou J, Wang J, Lin Y, Xue Y, Zhu P, Liu Z, Huang L, Nie J, Si W, Ma J, Ye J, Finnell RH, Saiyin H, Wang H, Zhao J, Zhao S, Xu W. Inhibiting MARSs reduces hyperhomocysteinemia-associated neural tube and congenital heart defects. EMBO Mol Med 2020; 12:e9469. [PMID: 32003121 PMCID: PMC7059139 DOI: 10.15252/emmm.201809469] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 02/05/2023] Open
Abstract
Hyperhomocysteinemia is a common metabolic disorder that imposes major adverse health consequences. Reducing homocysteine levels, however, is not always effective against hyperhomocysteinemia-associated pathologies. Herein, we report the potential roles of methionyl-tRNA synthetase (MARS)-generated homocysteine signals in neural tube defects (NTDs) and congenital heart defects (CHDs). Increased copy numbers of MARS and/or MARS2 were detected in NTD and CHD patients. MARSs sense homocysteine and transmit its signal by inducing protein lysine (N)-homocysteinylation. Here, we identified hundreds of novel N-homocysteinylated proteins. N-homocysteinylation of superoxide dismutases (SOD1/2) provided new mechanistic insights for homocysteine-induced oxidative stress, apoptosis and Wnt signalling deregulation. Elevated MARS expression in developing and proliferating cells sensitizes them to the effects of homocysteine. Targeting MARSs using the homocysteine analogue acetyl homocysteine thioether (AHT) reversed MARS efficacy. AHT lowered NTD and CHD onsets in retinoic acid-induced and hyperhomocysteinemia-induced animal models without affecting homocysteine levels. We provide genetic and biochemical evidence to show that MARSs are previously overlooked genetic determinants and key pathological factors of hyperhomocysteinemia, and suggest that MARS inhibition represents an important medicinal approach for controlling hyperhomocysteinemia-associated diseases.
Collapse
|
34
|
Mechanisms of homocysteine-induced damage to the endothelial, medial and adventitial layers of the arterial wall. Biochimie 2020; 173:100-106. [PMID: 32105811 DOI: 10.1016/j.biochi.2020.02.012] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/20/2020] [Indexed: 11/23/2022]
Abstract
Homocysteine (Hcy) is a non-protein forming amino acid which is the direct metabolic precursor of methionine. Increased concentration of serum Hcy is considered a risk factor for cardiovascular disease and is specifically linked to various diseases of the vasculature. Serum Hcy is associated with atherosclerosis, hypertension and aneurysms of the aorta in humans, though the precise mechanisms by which Hcy contributes to these conditions remain elusive. Results from clinical trials that successfully lowered serum Hcy without reducing features of vascular disease in cardiovascular patients have cast doubt on whether or not Hcy directly impacts the vasculature. However, studies in animals and in cell culture suggest that Hcy has a vast array of toxic effects on the vasculature, with demonstrated roles in endothelial dysfunction, medial remodeling and adventitial inflammation. It is hypothesized that rather than serum Hcy, tissue-bound Hcy and the incorporation of Hcy into proteins could underlie the toxic effects of Hcy on the vasculature. In this review, we present evidence for Hcy-associated vascular disease in humans, and we critically examine the possible mechanisms by which Hcy specifically impacts the endothelial, medial and adventitial layers of the arterial wall. Deciphering the mechanisms by which Hcy interacts with proteins in the arterial wall will allow for a better understanding of the pathomechanisms of hyperhomocysteinemia and will help to define a better means of prevention at the appropriate window of life.
Collapse
|
35
|
Urquhart BL, House AA. Assessing Plasma Total Homocysteine in Patients with End-Stage Renal Disease. Perit Dial Int 2020. [DOI: 10.1177/089686080702700502] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Elevated plasma total homocysteine (tHcy) is a risk factor for cardiovascular disease; however, in light of several recent randomized trials, the issue of causality has been cast into doubt. Patients with end-stage renal disease are particularly interesting as they consistently have elevated tHcy and their leading causes of morbidity and mortality are related to cardiovascular disease. In the present article, we review the early evidence for the homocysteine theory of atherosclerosis, homocysteine metabolism, mechanisms of toxicity, and pertinent available clinical investigations. Where appropriate, the sparse evidence of homocysteine in peritoneal dialysis is reviewed. We conclude by addressing the difficulties associated with lowering plasma tHcy in patients with end-stage renal disease and suggest some novel methods for lowering tHcy in this resistant population. Finally, to address the issue of causality, we recommend that clinicians and scientists await the results of the FAVORIT trial before abandoning homocysteine as a modifiable risk factor for cardiovascular disease, as this study has recruited patients from a population with consistently elevated plasma tHcy who are known to respond to vitamin therapy.
Collapse
Affiliation(s)
- Bradley L. Urquhart
- Departments of Medicine The University of Western Ontario, London, Ontario, Canada
- Physiology/Pharmacology, The University of Western Ontario, London, Ontario, Canada
| | - Andrew A. House
- Departments of Medicine The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
36
|
Homocysteine and Asymmetrical Dimethylarginine in Diabetic Rats Treated with Docosahexaenoic Acid-Loaded Zinc Oxide Nanoparticles. Appl Biochem Biotechnol 2020; 191:1127-1139. [PMID: 31960366 DOI: 10.1007/s12010-020-03230-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/08/2020] [Indexed: 12/17/2022]
Abstract
Hyperglycemia, the hallmark of diabetes mellitus, is considered one of the endothelial dysfunction risk factors, the main reason of vascular complication. In this study, we aimed to evaluate homocysteine (Hcy) and asymmetrical dimethylarginine (ADMA) levels in diabetic rats and the possibility to attenuate the elevation of these two parameters by supplementation of docosahexaenoic acid (DHA) alone or loaded zinc oxide nanoparticles (ZnONPs) to improve endothelial dysfunction in streptozotocin (STZ)-induced diabetic rats. Forty male albino rats weighing 180-200 g were classified as control, diabetic, diabetic treated with DHA, and diabetic treated with DHA-loaded zinc oxide nanoparticles (DHA/ZnONPs) groups. Fasting blood glucose, insulin, ADMA, Hcy, and nitric oxide (NO) were estimated. Fatty acids (linoleic acid (LA), arachidonic acid (AA), DHA, α-linolenic acid (ALA), and oleic acid (OA)) were also evaluated by reversed phase HPLC using a UV detector. The results showed that fasting blood sugar, insulin resistance, LA, AA, OA, ADMA, and Hcy increased significantly in diabetic rats compared with control while fasting insulin, DHA, ALA, and NO decreased significantly in diabetic rats. In both treated groups, fasting blood sugar, insulin resistance, LA, AA, OA, ADMA, and Hcy significantly decreased as compared with the diabetic group while fasting insulin, DHA, ALA, and NO were significantly increased. In conclusion, DHA and DHA/ZnONP supplementation protect against diabetic complications and improve endothelial dysfunction as well as hyperhomocysteinemia in diabetes. DHA/ZnONP-treated group appeared more efficient than DHA alone.
Collapse
|
37
|
Kumar A, Pathak R, Palfrey HA, Stone KP, Gettys TW, Murthy SN. High levels of dietary methionine improves sitagliptin-induced hepatotoxicity by attenuating oxidative stress in hypercholesterolemic rats. Nutr Metab (Lond) 2020; 17:2. [PMID: 31921324 PMCID: PMC6945706 DOI: 10.1186/s12986-019-0422-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/29/2019] [Indexed: 02/06/2023] Open
Abstract
Background Both cholesterol (Cho) and methionine (Met, a precursor for homocysteine) are risk factors for fatty liver disease. Since Western diets are rich in Cho and Met, we investigated the hepatic effects of feeding a diet enriched in Met and Cho. Further, based on the reported anti-oxidative and lipid lowering properties of sitagliptin (an antidiabetic drug), we tested whether it could counteract the negative effects of high Cho and Met. We therefore hypothesized that sitagliptin would ameliorate the development of liver pathology that is produced by feeding diets rich in either Cho, Met, or both. Methods Male Sprague Dawley rats were fed ad libitum a) control diet, or b) high Met or c) high Cho, or d) high Met + high Cho diets for 35 days. From day 10 to 35, 50% of rats in each dietary group were gavaged with either vehicle or an aqueous suspension of sitagliptin (100 mg/kg/day). Liver samples were harvested for histological, molecular, and biochemical analyses. Results The high Cho diet produced significant hepatic steatosis which was unaffected by sitagliptin. Contrary to expectation, sitagliptin exacerbated expression of hepatic markers of oxidative stress and fibrosis in rats fed high Cho. Corresponding increases in 4-hydroxynonenal adducts and collagen deposition were demonstrated by immunohistochemistry and sirius red staining. These hepatic changes were absent in rats on the high Met diet and they were comparable to controls. The inclusion of Met in the high Cho diet resulted in significant reduction of the hepatic steatosis, oxidative stress, and fibrosis produced by high Cho alone. Conclusion Sitagliptin exacerbated the effects of high Cho on both oxidative stress and fibrosis, resulting in NASH like symptoms that were significantly reversed by the inclusion of Met.
Collapse
Affiliation(s)
- Avinash Kumar
- 1Environmental Toxicology Department, Southern University and A&M College, Baton Rouge, LA 70813 USA
| | - Rashmi Pathak
- 1Environmental Toxicology Department, Southern University and A&M College, Baton Rouge, LA 70813 USA
| | - Henry A Palfrey
- 1Environmental Toxicology Department, Southern University and A&M College, Baton Rouge, LA 70813 USA
| | - Kirsten P Stone
- 2Laboratory of Nutrient Sensing and Adipocyte Signaling, Pennington Biomedical Research Center, Baton Rouge, LA USA
| | - Thomas W Gettys
- 2Laboratory of Nutrient Sensing and Adipocyte Signaling, Pennington Biomedical Research Center, Baton Rouge, LA USA
| | - Subramanyam N Murthy
- 1Environmental Toxicology Department, Southern University and A&M College, Baton Rouge, LA 70813 USA
| |
Collapse
|
38
|
Wang D, Zhao R, Qu YY, Mei XY, Zhang X, Zhou Q, Li Y, Yang SB, Zuo ZG, Chen YM, Lin Y, Xu W, Chen C, Zhao SM, Zhao JY. Colonic Lysine Homocysteinylation Induced by High-Fat Diet Suppresses DNA Damage Repair. Cell Rep 2019; 25:398-412.e6. [PMID: 30304680 DOI: 10.1016/j.celrep.2018.09.022] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/15/2018] [Accepted: 09/07/2018] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) onset is profoundly affected by Western diet. Here, we report that high-fat (HF) diet-induced, organ-specific colonic lysine homocysteinylation (K-Hcy) increase might promote CRC onset by impeding DNA damage repair. HF chow induced elevated methionyl-tRNA synthetase (MARS) expression and K-Hcy levels and DNA damage accumulation in the mouse and rat colon, resulting in a phenotype identical to that of CRC tissues. Moreover, the increased copy number of MARS, whose protein product promotes K-Hcy, correlated with increased CRC risk in humans. Mechanistically, MARS preferentially bound to and modified ataxia-telangiectasia and Rad3-related protein (ATR), inhibited ATR and its downstream effectors checkpoint kinase-1 and p53, and relieved cell-cycle arrest and decreased DNA damage-induced apoptosis by disrupting the binding of ATR-interacting protein to ATR. Inhibiting K-Hcy by targeting MARS reversed these effects and suppressed oncogenic CRC cell growth. Our study reveals a mechanism of Western-diet-associated CRC and highlights an intervention approach for reversing diet-induced oncogenic effects.
Collapse
Affiliation(s)
- Dan Wang
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, China; Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center for Genetics and Development and Children's Hospital of Fudan University, Shanghai 200438, China; Department of Neonatology and Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Rui Zhao
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, China
| | - Yuan-Yuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200438, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200438, China
| | - Xin-Yu Mei
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, China
| | - Xuan Zhang
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, China
| | - Qian Zhou
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, China
| | - Yang Li
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, China
| | - Shao-Bo Yang
- Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center for Genetics and Development and Children's Hospital of Fudan University, Shanghai 200438, China
| | - Zhi-Gui Zuo
- Department of Neonatology and Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yi-Ming Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yan Lin
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, China; Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center for Genetics and Development and Children's Hospital of Fudan University, Shanghai 200438, China
| | - Wei Xu
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, China; Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center for Genetics and Development and Children's Hospital of Fudan University, Shanghai 200438, China; Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chao Chen
- Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center for Genetics and Development and Children's Hospital of Fudan University, Shanghai 200438, China
| | - Shi-Min Zhao
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, China; Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center for Genetics and Development and Children's Hospital of Fudan University, Shanghai 200438, China; Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Jian-Yuan Zhao
- Obstetrics & Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai 200438, China; Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center for Genetics and Development and Children's Hospital of Fudan University, Shanghai 200438, China; Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
39
|
Parkhitko AA, Jouandin P, Mohr SE, Perrimon N. Methionine metabolism and methyltransferases in the regulation of aging and lifespan extension across species. Aging Cell 2019; 18:e13034. [PMID: 31460700 PMCID: PMC6826121 DOI: 10.1111/acel.13034] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/11/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022] Open
Abstract
Methionine restriction (MetR) extends lifespan across different species and exerts beneficial effects on metabolic health and inflammatory responses. In contrast, certain cancer cells exhibit methionine auxotrophy that can be exploited for therapeutic treatment, as decreasing dietary methionine selectively suppresses tumor growth. Thus, MetR represents an intervention that can extend lifespan with a complementary effect of delaying tumor growth. Beyond its function in protein synthesis, methionine feeds into complex metabolic pathways including the methionine cycle, the transsulfuration pathway, and polyamine biosynthesis. Manipulation of each of these branches extends lifespan; however, the interplay between MetR and these branches during regulation of lifespan is not well understood. In addition, a potential mechanism linking the activity of methionine metabolism and lifespan is regulation of production of the methyl donor S-adenosylmethionine, which, after transferring its methyl group, is converted to S-adenosylhomocysteine. Methylation regulates a wide range of processes, including those thought to be responsible for lifespan extension by MetR. Although the exact mechanisms of lifespan extension by MetR or methionine metabolism reprogramming are unknown, it may act via reducing the rate of translation, modifying gene expression, inducing a hormetic response, modulating autophagy, or inducing mitochondrial function, antioxidant defense, or other metabolic processes. Here, we review the mechanisms of lifespan extension by MetR and different branches of methionine metabolism in different species and the potential for exploiting the regulation of methyltransferases to delay aging.
Collapse
Affiliation(s)
- Andrey A. Parkhitko
- Department of GeneticsBlavatnik InstituteHarvard Medical SchoolBostonMassachusetts
| | - Patrick Jouandin
- Department of GeneticsBlavatnik InstituteHarvard Medical SchoolBostonMassachusetts
| | - Stephanie E. Mohr
- Department of GeneticsBlavatnik InstituteHarvard Medical SchoolBostonMassachusetts
| | - Norbert Perrimon
- Department of GeneticsBlavatnik InstituteHarvard Medical SchoolBostonMassachusetts
- Howard Hughes Medical InstituteBostonMassachusetts
| |
Collapse
|
40
|
Song J, Zheng Q, Ma X, Zhang Q, Xu Z, Zou C, Wang Z. Expression levels of paraoxonase-1 in aortic valve tissue are associated with the progression of calcific aortic valve stenosis. J Thorac Dis 2019; 11:2890-2898. [PMID: 31463118 DOI: 10.21037/jtd.2019.07.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Paraoxonase-1 (PON1) participates in several vital steps of lipid metabolism, which is associated with calcific aortic valve stenosis (CAVS). Although a few studies have suggested that PON1 in blood could inhibit aortic valve calcification, they did not provide detailed descriptions. In this study, we hypothesized that PON1 is expressed in the aortic valve and that the PON1 level is related to the severity of CAVS. Methods A total of 118 consecutive patients with CAVS were enrolled in the study; 35 individuals without aortic valve calcification were included in the control group. Aortic valve tissue was obtained from postoperative pathologic specimens. PON1 was detected qualitatively using immunohistochemistry and quantitatively using an enzyme-linked immunosorbent assay. The severity of aortic stenosis was evaluated using echocardiography. Results We detected PON1 in the aortic valve and noticed that the PON1 level was significantly lower in the case group than in the control group (P<0.001). Furthermore, we found no significant difference between the mild and moderate stenosis groups (P=0.395). However, the PON1 levels were obviously higher in both the mild and moderate stenosis groups than in the severe stenosis group (both P<0.001). We also detected a significant negative correlation between PON1 level and the maximum aortic valve gradient in the case group. Conclusions We detected PON1 in the aortic valve for the first time, and our results suggest that the PON1 level in aortic valve tissue decreases with increasing severity of aortic valve stenosis.
Collapse
Affiliation(s)
- Jian Song
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, China
| | - Qiang Zheng
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, China
| | - Xiaochun Ma
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, China
| | - Qian Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, China
| | - Zhenqiang Xu
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, China
| | - Chengwei Zou
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, China
| | - Zhengjun Wang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, China
| |
Collapse
|
41
|
Dysregulation of Epigenetic Mechanisms of Gene Expression in the Pathologies of Hyperhomocysteinemia. Int J Mol Sci 2019; 20:ijms20133140. [PMID: 31252610 PMCID: PMC6651274 DOI: 10.3390/ijms20133140] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
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.
Collapse
|
42
|
Tinelli C, Di Pino A, Ficulle E, Marcelli S, Feligioni M. Hyperhomocysteinemia as a Risk Factor and Potential Nutraceutical Target for Certain Pathologies. Front Nutr 2019; 6:49. [PMID: 31069230 PMCID: PMC6491750 DOI: 10.3389/fnut.2019.00049] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/03/2019] [Indexed: 01/08/2023] Open
Abstract
Hyperhomocysteinemia is recognized as a risk factor for several diseases, including cardiovascular and neurological conditions. Homocysteine (HCys) is a key metabolite involved in the biosynthesis and metabolism of methionine (Met), which plays a pivotal role in the physiological cell's life cycle. The biochemistry of Met is finely regulated by several enzymes that control HCys concentration. Indeed, balanced activity among the enzymes is essential for the cell's well-being, while its malfunction could raise HCys concentration which can lead to the onset of several pathological conditions. The HCys concentration increase seems to be caused mainly by the widely diffused polymorphisms of several enzymes. Nowadays, a blood test can easily detect elevated concentrations of HCys, referred to as Hyperhomocysteinemia (HHCys). Prolonged exposure to this condition can lead to the onset of cardiovascular disease and can lead to the development of atherosclerosis, stroke, inflammatory syndromes like osteoporosis and rheumatism, as well as neuronal pathologies including Alzheimer's and Parkinson's diseases. In this review, we analyzed the literature of several pathological conditions in which the molecular pathways of HHCys are involved. Interestingly, several observations indicate that the calibrated assumption of correct doses of vitamins such as folic acid, vitamin B6, vitamin B12, and betaine may control HHCys-related conditions.
Collapse
Affiliation(s)
- Caterina Tinelli
- Golgi Cenci Foundation, Abbiategrasso, Italy
- Laboratory of Neuronal Cell Signaling, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
| | - Antonella Di Pino
- Laboratory of Neuronal Cell Signaling, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
| | - Elena Ficulle
- Laboratory of Neurobiology in Translational Medicine, Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | - Serena Marcelli
- Laboratory of Neuronal Cell Signaling, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
| | - Marco Feligioni
- Laboratory of Neuronal Cell Signaling, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
- Laboratory of Neurobiology in Translational Medicine, Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| |
Collapse
|
43
|
Esse R, Barroso M, Tavares de Almeida I, Castro R. The Contribution of Homocysteine Metabolism Disruption to Endothelial Dysfunction: State-of-the-Art. Int J Mol Sci 2019; 20:E867. [PMID: 30781581 PMCID: PMC6412520 DOI: 10.3390/ijms20040867] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/05/2019] [Accepted: 02/12/2019] [Indexed: 02/07/2023] Open
Abstract
Homocysteine (Hcy) is a sulfur-containing non-proteinogenic amino acid formed during the metabolism of the essential amino acid methionine. Hcy is considered a risk factor for atherosclerosis and cardiovascular disease (CVD), but the molecular basis of these associations remains elusive. The impairment of endothelial function, a key initial event in the setting of atherosclerosis and CVD, is recurrently observed in hyperhomocysteinemia (HHcy). Various observations may explain the vascular toxicity associated with HHcy. For instance, Hcy interferes with the production of nitric oxide (NO), a gaseous master regulator of endothelial homeostasis. Moreover, Hcy deregulates the signaling pathways associated with another essential endothelial gasotransmitter: hydrogen sulfide. Hcy also mediates the loss of critical endothelial antioxidant systems and increases the intracellular concentration of reactive oxygen species (ROS) yielding oxidative stress. ROS disturb lipoprotein metabolism, contributing to the growth of atherosclerotic vascular lesions. Moreover, excess Hcy maybe be indirectly incorporated into proteins, a process referred to as protein N-homocysteinylation, inducing vascular damage. Lastly, cellular hypomethylation caused by build-up of S-adenosylhomocysteine (AdoHcy) also contributes to the molecular basis of Hcy-induced vascular toxicity, a mechanism that has merited our attention in particular. AdoHcy is the metabolic precursor of Hcy, which accumulates in the setting of HHcy and is a negative regulator of most cell methyltransferases. In this review, we examine the biosynthesis and catabolism of Hcy and critically revise recent findings linking disruption of this metabolism and endothelial dysfunction, emphasizing the impact of HHcy on endothelial cell methylation status.
Collapse
Affiliation(s)
- Ruben Esse
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Madalena Barroso
- University Children's Research@Kinder-UKE, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Isabel Tavares de Almeida
- Laboratory of Metabolism and Genetics, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
| | - Rita Castro
- Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
44
|
Jakubowski H. Homocysteine Modification in Protein Structure/Function and Human Disease. Physiol Rev 2019; 99:555-604. [PMID: 30427275 DOI: 10.1152/physrev.00003.2018] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
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.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers-New Jersey Medical School, International Center for Public Health , Newark, New Jersey ; and Department of Biochemistry and Biotechnology, Poznań University of Life Sciences , Poznań , Poland
| |
Collapse
|
45
|
Borowczyk K, Piechocka J, Głowacki R, Dhar I, Midtun Ø, Tell GS, Ueland PM, Nygård O, Jakubowski H. Urinary excretion of homocysteine thiolactone and the risk of acute myocardial infarction in coronary artery disease patients: the WENBIT trial. J Intern Med 2019; 285:232-244. [PMID: 30193001 PMCID: PMC6378604 DOI: 10.1111/joim.12834] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES No individual homocysteine (Hcy) metabolite has been studied as a risk marker for coronary artery disease (CAD). Our objective was to examine Hcy-thiolactone, a chemically reactive metabolite generated by methionyl-tRNA synthetase and cleared by the kidney, as a risk predictor of incident acute myocardial infarction (AMI) in the Western Norway B-Vitamin Intervention Trial. DESIGN Single centre, prospective double-blind clinical intervention study, randomized in a 2 × 2 factorial design. SUBJECTS AND METHODS Patients with suspected CAD (n = 2049, 69.8% men; 61.2-year-old) were randomized to groups receiving daily (i) folic acid (0.8 mg)/vitamin B12 (0.4 mg)/vitamin B6 (40 mg); (ii) folic acid/vitamin B12 ; (iii) vitamin B6 or (iv) placebo. Urinary Hcy-thiolactone was quantified at baseline, 12 and 38 months. RESULTS Baseline urinary Hcy-thiolactone/creatinine was significantly associated with plasma tHcy, ApoA1, glomerular filtration rate, potassium and pyridoxal 5'-phosphate (positively) and with age, hypertension, smoking, urinary creatinine, plasma bilirubin and kynurenine (negatively). During median 4.7-years, 183 patients (8.9%) suffered an AMI. In Cox regression analysis, Hcy-thiolactone/creatinine was associated with AMI risk (hazard ratio = 1.58, 95% confidence interval = 1.10-2.26, P = 0.012 for trend; adjusted for age, gender, tHcy). This association was confined to patients with pyridoxic acid below median (adjusted HR = 2.72, 95% CI = 1.47-5.03, P = 0.0001; Pinteraction = 0.020). B-vitamin/folate treatments did not affect Hcy-thiolactone/creatinine and its AMI risk association. CONCLUSIONS Hcy-thiolactone/creatinine ratio is a novel AMI risk predictor in patients with suspected CAD, independent of traditional risk factors and tHcy, but modified by vitamin B6 catabolism. These findings lend a support to the hypothesis that Hcy-thiolactone is mechanistically involved in cardiovascular disease.
Collapse
Affiliation(s)
- K Borowczyk
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers-New Jersey Medical School, International Center for Public Health, Newark, NJ, USA.,Department of Environmental Chemistry, Faculty of Chemistry, University of Łódź, Łódź, Poland
| | - J Piechocka
- Department of Environmental Chemistry, Faculty of Chemistry, University of Łódź, Łódź, Poland
| | - R Głowacki
- Department of Environmental Chemistry, Faculty of Chemistry, University of Łódź, Łódź, Poland
| | - I Dhar
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - G S Tell
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway.,Division of Mental and Physical Health, Norwegian Institute of Public Health, Bergen, Norway
| | - P M Ueland
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - O Nygård
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Heart Disease, Haukeland University Hospital, Institute of Medicine, University of Bergen, Bergen, Norway
| | - H Jakubowski
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers-New Jersey Medical School, International Center for Public Health, Newark, NJ, USA.,Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| |
Collapse
|
46
|
Zhu X, Zhang L, Hu Y, Zhang J. Identification of suitable reference genes for real-time qPCR in homocysteine-treated human umbilical vein endothelial cells. PLoS One 2018; 13:e0210087. [PMID: 30596787 PMCID: PMC6312244 DOI: 10.1371/journal.pone.0210087] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 12/17/2018] [Indexed: 12/14/2022] Open
Abstract
The imbalance in homocysteine (Hcy) metabolism has been implicated in the pathogenesis of human diseases, including cardiovascular and neurodegenerative disorders. When attempting to identify gene expression profiles using quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR), the selection of suitable reference genes is important. Here, the expression levels of 10 commonly used reference genes were assessed for normalization of RT-qPCR in Hcy-treated human umbilical vein endothelial cells (HUVECs) and control cells. The suitability of eight selected candidate genes was comparatively analyzed across the tested samples and separately ranked by four programs, geNorm, NormFinder, BestKeeper, and the ΔCt method. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was the most stable gene in the final ranking using the RankAggreg package. Surprisingly, the β-actin (ACTB) levels decreased significantly in Hcy-treated HUVECs compared with control HUVECs (P<0.05), and further study indicated that Hcy suppressed the expression of ACTB by upregulating the miR-145-5p level in Hcy-treated HUVECs. Our data suggest that GAPDH can be used as a reliable reference gene, while ACTB cannot; normalization of gene expression in RT-qPCR experiments in Hcy-treated HUVECs. The data, which identifies a suitable reference gene in Hcy-treated HUVECs, will contribute to the design of an effective and accurate method for quantitation of gene expression.
Collapse
Affiliation(s)
- Xia Zhu
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Cardiology, Urumqi Friendship Hospital, Urumqi, China
| | - Lujun Zhang
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
- * E-mail: (LZ); (JZ)
| | - Yangxi Hu
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianliang Zhang
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
- * E-mail: (LZ); (JZ)
| |
Collapse
|
47
|
Price BR, Wilcock DM, Weekman EM. Hyperhomocysteinemia as a Risk Factor for Vascular Contributions to Cognitive Impairment and Dementia. Front Aging Neurosci 2018; 10:350. [PMID: 30429785 PMCID: PMC6220027 DOI: 10.3389/fnagi.2018.00350] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022] Open
Abstract
Behind only Alzheimer's disease, vascular contributions to cognitive impairment and dementia (VCID) is the second most common cause of dementia, affecting roughly 10-40% of dementia patients. While there is no cure for VCID, several risk factors for VCID, such as diabetes, hypertension, and stroke, have been identified. Elevated plasma levels of homocysteine, termed hyperhomocysteinemia (HHcy), are a major, yet underrecognized, risk factor for VCID. B vitamin deficiency, which is the most common cause of HHcy, is common in the elderly. With B vitamin supplementation being a relatively safe and inexpensive therapeutic, the treatment of HHcy-induced VCID would seem straightforward; however, preclinical and clinical data shows it is not. Clinical trials using B vitamin supplementation have shown conflicting results about the benefits of lowering homocysteine and issues have arisen over proper study design within the trials. Studies using cell culture and animal models have proposed several mechanisms for homocysteine-induced cognitive decline, providing other targets for therapeutics. For this review, we will focus on HHcy as a risk factor for VCID, specifically, the different mechanisms proposed for homocysteine-induced cognitive decline and the clinical trials aimed at lowering plasma homocysteine.
Collapse
Affiliation(s)
| | | | - Erica M. Weekman
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
48
|
Ni K, Feng L, Ling M, Yin S, Zhang H, Zhu Y, Zhou Z, Zhou Z, Zhou X. The photocatalytic reaction of imperatorin and cysteine. MAIN GROUP CHEMISTRY 2018. [DOI: 10.3233/mgc-180263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Ke Ni
- Department of Chemical Engineering, School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, PR China
| | - Lin Feng
- Department of Chemical Engineering, School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, PR China
| | - Mengchen Ling
- Department of Chemical Engineering, School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, PR China
| | - Sili Yin
- Department of Chemical Engineering, School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, PR China
| | - Hailong Zhang
- Department of Chemical Engineering, School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, PR China
| | - Yunyang Zhu
- Department of Chemical Engineering, School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, PR China
| | - Zhen Zhou
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, PR China
| | - Zhiming Zhou
- Department of Chemical Engineering, School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, PR China
| | - Xiaohua Zhou
- Department of Chemical Engineering, School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, PR China
| |
Collapse
|
49
|
Majumder A, Singh M, George AK, Behera J, Tyagi N, Tyagi SC. Hydrogen sulfide improves postischemic neoangiogenesis in the hind limb of cystathionine-β-synthase mutant mice via PPAR-γ/VEGF axis. Physiol Rep 2018; 6:e13858. [PMID: 30175474 PMCID: PMC6119702 DOI: 10.14814/phy2.13858] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022] Open
Abstract
Neoangiogenesis is a fundamental process which helps to meet energy requirements, tissue growth, and wound healing. Although previous studies showed that Peroxisome proliferator-activated receptor (PPAR-γ) regulates neoangiogenesis via upregulation of vascular endothelial growth factor (VEGF), and both VEGF and PPAR-γ expressions were inhibited during hyperhomocysteinemic (HHcy), whether these two processes could trigger pathological effects in skeletal muscle via compromising neoangiogenesis has not been studied yet. Unfortunately, there are no treatment options available to date for ameliorating HHcy-mediated neoangiogenic defects. Hydrogen sulfide (H2 S) is a novel gasotransmitter that can induce PPAR-γ levels. However, patients with cystathionine-β-synthase (CBS) mutation(s) cannot produce a sufficient amount of H2 S. We hypothesized that exogenous supplementation of H2 S might improve HHcy-mediated poor neoangiogenesis via the PPAR-γ/VEGF axis. To examine this, we created a hind limb femoral artery ligation (FAL) in CBS+/- mouse model and treated them with GYY4137 (a long-acting H2 S donor compound) for 21 days. To evaluate neoangiogenesis, we used barium sulfate angiography and laser Doppler blood flow measurements in the ischemic hind limbs of experimental mice post-FAL to assess blood flow. Proteins and mRNAs levels were studied by Western blots and qPCR analyses. HIF1-α, VEGF, PPAR-γ and p-eNOS expressions were attenuated in skeletal muscle of CBS+/- mice after 21 days of FAL in comparison to wild-type (WT) mice, that were improved via GYY4137 treatment. We also found that the collateral vessel density and blood flow were significantly reduced in post-FAL CBS+/- mice compared to WT mice and these effects were ameliorated by GYY4137. Moreover, we found that plasma nitrite levels were decreased in post-FAL CBS+/- mice compared to WT mice, which were mitigated by GYY4137 supplementation. These results suggest that HHcy can inhibit neoangiogenesis via antagonizing the angiogenic signal pathways encompassing PPAR-γ/VEGF axis and that GYY4137 could serve as a potential therapeutic to alleviate the harmful metabolic effects of HHcy conditions.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of PhysiologyUniversity of Louisville School of MedicineLouisvilleKentucky40202USA
- Department of Biochemistry and Molecular GeneticsUniversity of Louisville School of MedicineLouisvilleKentucky40202USA
| | - Mahavir Singh
- Department of PhysiologyUniversity of Louisville School of MedicineLouisvilleKentucky40202USA
| | - Akash K. George
- Department of PhysiologyUniversity of Louisville School of MedicineLouisvilleKentucky40202USA
| | - Jyotirmaya Behera
- Department of PhysiologyUniversity of Louisville School of MedicineLouisvilleKentucky40202USA
| | - Neetu Tyagi
- Department of PhysiologyUniversity of Louisville School of MedicineLouisvilleKentucky40202USA
| | - Suresh C. Tyagi
- Department of PhysiologyUniversity of Louisville School of MedicineLouisvilleKentucky40202USA
| |
Collapse
|
50
|
Akbarian M, Ghasemi Y, Uversky VN, Yousefi R. Chemical modifications of insulin: Finding a compromise between stability and pharmaceutical performance. Int J Pharm 2018; 547:450-468. [DOI: 10.1016/j.ijpharm.2018.06.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 02/07/2023]
|