51
|
Paul R, Borah A. L-DOPA-induced hyperhomocysteinemia in Parkinson's disease: Elephant in the room. Biochim Biophys Acta Gen Subj 2016; 1860:1989-97. [DOI: 10.1016/j.bbagen.2016.06.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 05/20/2016] [Accepted: 06/14/2016] [Indexed: 02/08/2023]
|
52
|
Cotard S, Nouni A, Jaquinandi V, Gladu G, Kaladji A, Mahé G. [Peripheral artery disease in patients younger than 50 years old: Which etiology?]. Ann Cardiol Angeiol (Paris) 2016; 65:275-285. [PMID: 27319272 DOI: 10.1016/j.ancard.2016.04.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 04/29/2016] [Indexed: 06/06/2023]
Abstract
Peripheral arterial disease (PAD) encompasses disease of all arteries of the body except the coronary arteries. The main etiology whatever the patient's age is atherosclerosis. Different etiologies can induce PAD especially when patients are younger than 50 years old and have no cardiovascular risk factors (smoking, hypertension, diabetes…). PAD that appears before 50 years old can be named juvenile PAD (JPAD) although there is no consensus about the definition. The aim of this work is to present the different etiologies of JPAD according to their hereditary, acquired or mixed origins. The following hereditary causes are addressed: Marfan syndrome, Ehlers-Danlos syndrome, homocystinuria, pseudoxanthoma elasticum, osteogenesis imperfecta "mid-aortic" syndrome. Among the acquired etiologies, inflammatory JPADs without extravascular signs such as atherosclerosis and Buerger's disease, inflammatory JPADs with extravascular signs as Takayasu's disease, Behçet's disease and Cogan's syndrome, JPADs like aortitis, embolic JPADs, iatrogenic JPADs, and mechanical or traumatic JPADs are described. Finally, mixed origins as thrombotic disease and fibromuscular dysplasia are presented. This work will assist clinicians in the diagnosis of JPAD.
Collapse
Affiliation(s)
- S Cotard
- Imagerie cœur-vaisseaux, centre hospitalier universitaire, hôpital Pontchaillou, 2, rue Henri-Le-Guilloux, 35033 Rennes, France
| | - A Nouni
- Centre hospitalier centre Bretagne, Kério, 56306 Pontivy, France
| | - V Jaquinandi
- Imagerie cœur-vaisseaux, centre hospitalier universitaire, hôpital Pontchaillou, 2, rue Henri-Le-Guilloux, 35033 Rennes, France; Université de Rennes 1, Inserm, centre d'investigation clinique CIC 1414, 35033 Rennes, France
| | - G Gladu
- Clinique du Ter, 56270 Plœmeur, France
| | - A Kaladji
- Service de chirurgie vasculaire, centre hospitalier universitaire, hôpital Pontchaillou, 2, rue Henri-Le-Guilloux, 35033 Rennes, France
| | - G Mahé
- Imagerie cœur-vaisseaux, centre hospitalier universitaire, hôpital Pontchaillou, 2, rue Henri-Le-Guilloux, 35033 Rennes, France; Université de Rennes 1, Inserm, centre d'investigation clinique CIC 1414, 35033 Rennes, France.
| |
Collapse
|
53
|
Katsouda A, Bibli SI, Pyriochou A, Szabo C, Papapetropoulos A. Regulation and role of endogenously produced hydrogen sulfide in angiogenesis. Pharmacol Res 2016; 113:175-185. [PMID: 27569706 DOI: 10.1016/j.phrs.2016.08.026] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 08/05/2016] [Accepted: 08/24/2016] [Indexed: 01/16/2023]
Abstract
Recent studies have implicated endogenously produced H2S in the angiogenic process. On one hand, pharmacological inhibition and silencing of the enzymes involved in H2S synthesis attenuate the angiogenic properties of endothelial cells, including proliferation, migration and tube-like structure network formation. On the other hand, enhanced production of H2S by substrate supplementation or over-expression of H2S-producing enzymes leads to enhanced angiogenic responses in cultured endothelial cells. Importantly, H2S up-regulates expression of the key angiogenic factor vascular endothelial growth factor (VEGF) and contributes to the angiogenic signaling in response to VEGF. The signaling pathways mediating H2S-induced angiogenesis include mitogen-activated protein kinases, phosphoinositide-3 kinase, nitric oxide/cGMP-regulated cascades and ATP-sensitive potassium channels. Endogenously produced H2S has also been shown to facilitate neovascularization in prototypical model systems in vivo, and to contribute to wound healing, post-ischemic angiogenesis in the heart and other tissues, as well as in tumor angiogenesis. Targeting of H2S synthesizing enzymes might offer novel therapeutic opportunities for angiogenesis-related diseases.
Collapse
Affiliation(s)
- Antonia Katsouda
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece, Greece; Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Greece
| | - Sofia-Iris Bibli
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece, Greece
| | - Anastasia Pyriochou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece, Greece; Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Greece.
| |
Collapse
|
54
|
Hackfort BT, Mishra PK. Emerging role of hydrogen sulfide-microRNA crosstalk in cardiovascular diseases. Am J Physiol Heart Circ Physiol 2016; 310:H802-12. [PMID: 26801305 PMCID: PMC4867357 DOI: 10.1152/ajpheart.00660.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/18/2016] [Indexed: 12/15/2022]
Abstract
Despite an obnoxious smell and toxicity at a high dose, hydrogen sulfide (H2S) is emerging as a cardioprotective gasotransmitter. H2S mitigates pathological cardiac remodeling by regulating several cellular processes including fibrosis, hypertrophy, apoptosis, and inflammation. These encouraging findings in rodents led to initiation of a clinical trial using a H2S donor in heart failure patients. However, the underlying molecular mechanisms by which H2S mitigates cardiac remodeling are not completely understood. Empirical evidence suggest that H2S may regulate signaling pathways either by directly influencing a gene in the cascade or interacting with nitric oxide (another cardioprotective gasotransmitter) or both. Recent studies revealed that H2S may ameliorate cardiac dysfunction by up- or downregulating specific microRNAs. MicroRNAs are noncoding, conserved, regulatory RNAs that modulate gene expression mostly by translational inhibition and are emerging as a therapeutic target for cardiovascular disease (CVD). Few microRNAs also regulate H2S biosynthesis. The inter-regulation of microRNAs and H2S opens a new avenue for exploring the H2S-microRNA crosstalk in CVD. This review embodies regulatory mechanisms that maintain the physiological level of H2S, exogenous H2S donors used for increasing the tissue levels of H2S, H2S-mediated regulation of CVD, H2S-microRNAs crosstalk in relation to the pathophysiology of heart disease, clinical trials on H2S, and future perspectives for H2S as a therapeutic agent for heart failure.
Collapse
Affiliation(s)
- Bryan T Hackfort
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
55
|
Saha S, Chakraborty PK, Xiong X, Dwivedi SKD, Mustafi SB, Leigh NR, Ramchandran R, Mukherjee P, Bhattacharya R. Cystathionine β-synthase regulates endothelial function via protein S-sulfhydration. FASEB J 2016; 30:441-56. [PMID: 26405298 PMCID: PMC4684530 DOI: 10.1096/fj.15-278648] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/14/2015] [Indexed: 12/31/2022]
Abstract
Deficiencies of the human cystathionine β-synthase (CBS) enzyme are characterized by a plethora of vascular disorders and hyperhomocysteinemia. However, several clinical trials demonstrated that despite reduction in homocysteine levels, disease outcome remained unaffected, thus the mechanism of endothelial dysfunction is poorly defined. Here, we show that the loss of CBS function in endothelial cells (ECs) leads to a significant down-regulation of cellular hydrogen sulfide (H2S) by 50% and of glutathione (GSH) by 40%. Silencing CBS in ECs compromised phenotypic and signaling responses to the VEGF that were potentiated by decreased transcription of VEGF receptor (VEGFR)-2 and neuropilin (NRP)-1, the primary receptors regulating endothelial function. Transcriptional down-regulation of VEGFR-2 and NRP-1 was mediated by a lack in stability of the transcription factor specificity protein 1 (Sp1), which is a sulfhydration target of H2S at residues Cys68 and Cys755. Reinstating H2S but not GSH in CBS-silenced ECs restored Sp1 levels and its binding to the VEGFR-2 promoter and VEGFR-2, NRP-1 expression, VEGF-dependent proliferation, and migration phenotypes. Thus, our study emphasizes the importance of CBS-mediated protein S-sulfhydration in maintaining vascular health and function.-Saha, S., Chakraborty, P. K., Xiong, X., Dwivedi, S. K. D., Mustafi, S. B., Leigh, N. R., Ramchandran, R., Mukherjee, P., Bhattacharya, R. Cystathionine β-synthase regulates endothelial function via protein S-sulfhydration.
Collapse
Affiliation(s)
- Sounik Saha
- *Peggy and Charles Stephenson Cancer Center, Department of Obstetrics and Gynecology, Department of Pathology, and Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and Developmental Vascular Biology Program and Zebrafish Drug Screening Core, Department of Obstetrics and Gynecology, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Prabir K Chakraborty
- *Peggy and Charles Stephenson Cancer Center, Department of Obstetrics and Gynecology, Department of Pathology, and Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and Developmental Vascular Biology Program and Zebrafish Drug Screening Core, Department of Obstetrics and Gynecology, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Xunhao Xiong
- *Peggy and Charles Stephenson Cancer Center, Department of Obstetrics and Gynecology, Department of Pathology, and Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and Developmental Vascular Biology Program and Zebrafish Drug Screening Core, Department of Obstetrics and Gynecology, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Shailendra Kumar Dhar Dwivedi
- *Peggy and Charles Stephenson Cancer Center, Department of Obstetrics and Gynecology, Department of Pathology, and Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and Developmental Vascular Biology Program and Zebrafish Drug Screening Core, Department of Obstetrics and Gynecology, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Soumyajit Banerjee Mustafi
- *Peggy and Charles Stephenson Cancer Center, Department of Obstetrics and Gynecology, Department of Pathology, and Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and Developmental Vascular Biology Program and Zebrafish Drug Screening Core, Department of Obstetrics and Gynecology, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Noah R Leigh
- *Peggy and Charles Stephenson Cancer Center, Department of Obstetrics and Gynecology, Department of Pathology, and Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and Developmental Vascular Biology Program and Zebrafish Drug Screening Core, Department of Obstetrics and Gynecology, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ramani Ramchandran
- *Peggy and Charles Stephenson Cancer Center, Department of Obstetrics and Gynecology, Department of Pathology, and Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and Developmental Vascular Biology Program and Zebrafish Drug Screening Core, Department of Obstetrics and Gynecology, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Priyabrata Mukherjee
- *Peggy and Charles Stephenson Cancer Center, Department of Obstetrics and Gynecology, Department of Pathology, and Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and Developmental Vascular Biology Program and Zebrafish Drug Screening Core, Department of Obstetrics and Gynecology, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Resham Bhattacharya
- *Peggy and Charles Stephenson Cancer Center, Department of Obstetrics and Gynecology, Department of Pathology, and Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, USA; and Developmental Vascular Biology Program and Zebrafish Drug Screening Core, Department of Obstetrics and Gynecology, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
56
|
Dugbartey GJ, Talaei F, Houwertjes MC, Goris M, Epema AH, Bouma HR, Henning RH. Dopamine treatment attenuates acute kidney injury in a rat model of deep hypothermia and rewarming – The role of renal H2S-producing enzymes. Eur J Pharmacol 2015; 769:225-33. [DOI: 10.1016/j.ejphar.2015.11.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/12/2015] [Accepted: 11/13/2015] [Indexed: 01/11/2023]
|
57
|
Sponholz C, Kramer M, Schöneweck F, Menzel U, Inanloo Rahatloo K, Giamarellos-Bourboulis EJ, Papavassileiou V, Lymberopoulou K, Pavlaki M, Koutelidakis I, Perdios I, Scherag A, Bauer M, Platzer M, Huse K. Polymorphisms of cystathionine beta-synthase gene are associated with susceptibility to sepsis. Eur J Hum Genet 2015; 24:1041-8. [PMID: 26508567 DOI: 10.1038/ejhg.2015.231] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 09/14/2015] [Accepted: 09/29/2015] [Indexed: 11/09/2022] Open
Abstract
Sepsis is the systemic inflammatory host response to infection. Cystathionine beta-synthase (CBS)-dependent homocysteine (Hcy) pathway was demonstrated to affect disease severity and mortality in patients with severe sepsis/septic shock. Independent studies identified a single-nucleotide polymorphism (SNP, rs6586282, hg19 chr21:g.44478497C>T) in intron 14 of the CBS-coding gene (CBS) associated with Hcy plasma levels. We aimed to describe the association of this SNP and variants of a splice donor-affecting variable-number tandem repeat (VNTR, NG_008938.1:g.22763_22793[16_22]) 243 bp downstream of rs6586282 with severe human sepsis. We analyzed the VNTR structure and genotyped variants of rs6586282 and a neighboring SNP (rs34758144, hg19 chr21:g.44478582G>A) in two case-control studies including patients with severe sepsis/septic shock from Germany (n=168) and Greece (n=237). In both studies, we consistently observed an association of CBS VNTR alleles with sepsis susceptibility. Risk linearly increased with number of tandem repeats (per allele odds ratio in the adjusted analysis 1.34; 95% confidence interval (CI)=1.17-1.55; P<0.001). Association had also been shown for rs34758144 whose risk allele is in linkage disequilibrium with one long VNTR allele (19 repeat). In contrast, we observed no evidence for an effect on 28-day survival in patients with severe sepsis/septic shock (per allele hazard ratio in the adjusted analysis for VNTR 1.10; 95% CI=0.95-1.28; P=0.20). In a minigene approach, we demonstrated alternative splicing in distinct VNTR alleles, which, however, was independent of the number of tandem units. In conclusion, there is no ordinary conjunction between human CBS and severe sepsis/septic shock, but CBS genotypes are involved in disease susceptibility.
Collapse
Affiliation(s)
- Christoph Sponholz
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Genome Analysis, Leibniz Institute for Age Research-Fritz Lipmann Institute, Jena, Germany.,Department of Anaesthesiology and Intensive Care Therapy, Jena University Hospital, Jena, Germany
| | - Marcel Kramer
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Genome Analysis, Leibniz Institute for Age Research-Fritz Lipmann Institute, Jena, Germany
| | - Franziska Schöneweck
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Research group Clinical Epidemiology, CSCC, Jena University Hospital, Jena, Germany
| | - Uwe Menzel
- Systems Biology and Bioinformatics Group, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena, Germany
| | - Kolsoum Inanloo Rahatloo
- Genome Analysis, Leibniz Institute for Age Research-Fritz Lipmann Institute, Jena, Germany.,School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Evangelos J Giamarellos-Bourboulis
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,4th Department of Internal Medicine, University of Athens, Medical School, Athens, Greece
| | | | - Korina Lymberopoulou
- 2nd Department of Internal Medicine, Sismanogleion General Hospital, Athens, Greece
| | - Maria Pavlaki
- Department of Internal Medicine, Argos General Hospital, Argos, Greece
| | - Ioannis Koutelidakis
- 2nd Department of Surgery, University of Thessaloniki, Medical School, Thessaloniki, Greece
| | - Ioannis Perdios
- 1st Department of Internal Medicine, 'G. Gennimatas' General Hospital, Athens, Greece
| | - André Scherag
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Research group Clinical Epidemiology, CSCC, Jena University Hospital, Jena, Germany
| | - Michael Bauer
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Department of Anaesthesiology and Intensive Care Therapy, Jena University Hospital, Jena, Germany
| | - Matthias Platzer
- Genome Analysis, Leibniz Institute for Age Research-Fritz Lipmann Institute, Jena, Germany
| | - Klaus Huse
- Genome Analysis, Leibniz Institute for Age Research-Fritz Lipmann Institute, Jena, Germany
| |
Collapse
|
58
|
Lin VS, Chen W, Xian M, Chang CJ. Chemical probes for molecular imaging and detection of hydrogen sulfide and reactive sulfur species in biological systems. Chem Soc Rev 2015; 44:4596-4618. [PMID: 25474627 PMCID: PMC4456340 DOI: 10.1039/c4cs00298a] [Citation(s) in RCA: 740] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hydrogen sulfide (H2S), a gaseous species produced by both bacteria and higher eukaryotic organisms, including mammalian vertebrates, has attracted attention in recent years for its contributions to human health and disease. H2S has been proposed as a cytoprotectant and gasotransmitter in many tissue types, including mediating vascular tone in blood vessels as well as neuromodulation in the brain. The molecular mechanisms dictating how H2S affects cellular signaling and other physiological events remain insufficiently understood. Furthermore, the involvement of H2S in metal-binding interactions and formation of related RSS such as sulfane sulfur may contribute to other distinct signaling pathways. Owing to its widespread biological roles and unique chemical properties, H2S is an appealing target for chemical biology approaches to elucidate its production, trafficking, and downstream function. In this context, reaction-based fluorescent probes offer a versatile set of screening tools to visualize H2S pools in living systems. Three main strategies used in molecular probe development for H2S detection include azide and nitro group reduction, nucleophilic attack, and CuS precipitation. Each of these approaches exploits the strong nucleophilicity and reducing potency of H2S to achieve selectivity over other biothiols. In addition, a variety of methods have been developed for the detection of other reactive sulfur species (RSS), including sulfite and bisulfite, as well as sulfane sulfur species and related modifications such as S-nitrosothiols. Access to this growing chemical toolbox of new molecular probes for H2S and related RSS sets the stage for applying these developing technologies to probe reactive sulfur biology in living systems.
Collapse
Affiliation(s)
- Vivian S Lin
- Department of Chemistry, University of California, Berkeley, California, USA
| | - Wei Chen
- Department of Chemistry, Washington State University, Pullman, Washington, USA
| | - Ming Xian
- Department of Chemistry, Washington State University, Pullman, Washington, USA
| | - Christopher J Chang
- Department of Chemistry, University of California, Berkeley, California, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
- Howard Hughes Medical Institute, University of California, Berkeley, California, USA
| |
Collapse
|
59
|
Li JJ, Li Q, Du HP, Wang YL, You SJ, Wang F, Xu XS, Cheng J, Cao YJ, Liu CF, Hu LF. Homocysteine Triggers Inflammatory Responses in Macrophages through Inhibiting CSE-H2S Signaling via DNA Hypermethylation of CSE Promoter. Int J Mol Sci 2015; 16:12560-77. [PMID: 26047341 PMCID: PMC4490461 DOI: 10.3390/ijms160612560] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/23/2015] [Accepted: 05/20/2015] [Indexed: 12/31/2022] Open
Abstract
Hyperhomocysteinemia (HHcy) is an independent risk factor of atherosclerosis and other cardiovascular diseases. Unfortunately, Hcy-lowering strategies were found to have limited effects in reducing cardiovascular events. The underlying mechanisms remain unclear. Increasing evidence reveals a role of inflammation in the pathogenesis of HHcy. Homocysteine (Hcy) is a precursor of hydrogen sulfide (H2S), which is formed via the transsulfuration pathway catalyzed by cystathionine β-synthase and cystathionine γ-lyase (CSE) and serves as a novel modulator of inflammation. In the present study, we showed that methionine supplementation induced mild HHcy in mice, associated with the elevations of TNF-α and IL-1β in the plasma and reductions of plasma H2S level and CSE expression in the peritoneal macrophages. H2S-releasing compound GYY4137 attenuated the increases of TNF-α and IL-1β in the plasma of HHcy mice and Hcy-treated raw264.7 cells while CSE inhibitor PAG exacerbated it. Moreover, the in vitro study showed that Hcy inhibited CSE expression and H2S production in macrophages, accompanied by the increases of DNA methyltransferase (DNMT) expression and DNA hypermethylation in cse promoter region. DNMT inhibition or knockdown reversed the decrease of CSE transcription induced by Hcy in macrophages. In sum, our findings demonstrate that Hcy may trigger inflammation through inhibiting CSE-H2S signaling, associated with increased promoter DNA methylation and transcriptional repression of cse in macrophages.
Collapse
Affiliation(s)
- Jiao-Jiao Li
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215004, China.
| | - Qian Li
- Institute of Neuroscience, Soochow University, Suzhou 215123, China.
- Department of Pharmacology, School of Pharmaceutical Science, Soochow University, Suzhou 215123, China.
| | - Hua-Ping Du
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215004, China.
| | - Ya-Li Wang
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215004, China.
- Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| | - Shou-Jiang You
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215004, China.
| | - Fen Wang
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215004, China.
- Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| | - Xing-Shun Xu
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215004, China.
| | - Jian Cheng
- Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| | - Yong-Jun Cao
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215004, China.
| | - Chun-Feng Liu
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215004, China.
- Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| | - Li-Fang Hu
- Department of Neurology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215004, China.
- Institute of Neuroscience, Soochow University, Suzhou 215123, China.
- Department of Pharmacology, School of Pharmaceutical Science, Soochow University, Suzhou 215123, China.
| |
Collapse
|
60
|
Koning AM, Frenay ARS, Leuvenink HG, van Goor H. Hydrogen sulfide in renal physiology, disease and transplantation – The smell of renal protection. Nitric Oxide 2015; 46:37-49. [DOI: 10.1016/j.niox.2015.01.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/19/2015] [Accepted: 01/22/2015] [Indexed: 01/08/2023]
|
61
|
Neuronal regulation of expression of hydrogen sulfide-producing enzyme cystathionine β-synthase in rat spinal cord astrocytes. Neurosci Res 2015; 97:52-9. [PMID: 25797494 DOI: 10.1016/j.neures.2015.03.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 03/10/2015] [Accepted: 03/10/2015] [Indexed: 12/13/2022]
Abstract
Cystathionine β-synthase (CBS), expressed in astrocytes, generates a gaseous neuromodulator, hydrogen sulfide (H2S) in the central nervous system (CNS). However, little is known about the regulatory mechanisms of astrocytic CBS expression and activity. This study evaluated the influence of neurons on astrocytic CBS expression by employing multiple culture systems. Substantial CBS expression was observed in the intact neonatal rat spinal cord, while CBS content was markedly reduced in an astrocyte-enriched culture prepared from the neonatal spinal cord. Immunofluorescence analysis confirmed the localization of spinal cord CBS in astrocytes, but not in neurons. Although CBS expression was weak in the embryonic rat spinal cord, enzyme levels were time-dependently increased in a neuron/astrocyte mixed culture originating from embryonic spinal cord. The reduced CBS expression in isolated neonatal astrocytes was restored by co-culture with embryonic neurons. Together with the observed CBS expression levels, H2S production was relatively low in astrocytes cultured alone, but was considerably higher in astrocytes cultured with neurons. These results indicate that neurons are essential for maintaining the expression and H2S-producing activity of astrocytic CBS in the rat spinal cord.
Collapse
|
62
|
Jung J, Jeong NY. Hydrogen sulfide controls peripheral nerve degeneration and regeneration: a novel therapeutic strategy for peripheral demyelinating disorders or nerve degenerative diseases. Neural Regen Res 2015; 9:2119-21. [PMID: 25657730 PMCID: PMC4316442 DOI: 10.4103/1673-5374.147940] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2014] [Indexed: 11/12/2022] Open
Affiliation(s)
- Junyang Jung
- Department of Anatomy and Neurobiology, School of Medicine, Biomedical Science Institution, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Na Young Jeong
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan 602-714, Republic of Korea
| |
Collapse
|
63
|
Thorson MK, Van Wagoner RM, Harper MK, Ireland CM, Majtan T, Kraus JP, Barrios AM. Marine natural products as inhibitors of cystathionine beta-synthase activity. Bioorg Med Chem Lett 2015; 25:1064-6. [PMID: 25666819 DOI: 10.1016/j.bmcl.2015.01.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 01/05/2015] [Accepted: 01/07/2015] [Indexed: 10/24/2022]
Abstract
A library consisting of characterized marine natural products as well as synthetic derivatives was screened for compounds capable of inhibiting the production of hydrogen sulfide (H2S) by cystathionine beta-synthase (CBS). Eight hits were validated and shown to inhibit CBS activity with IC50 values ranging from 83 to 187μM. The majority of hits came from a series of synthetic polyandrocarpamine derivatives. In addition, a modified fluorogenic probe for H2S detection with improved solubility in aqueous solutions is reported.
Collapse
Affiliation(s)
- Megan K Thorson
- Department of Medicinal Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - Ryan M Van Wagoner
- Department of Medicinal Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - Mary Kay Harper
- Department of Medicinal Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - Chris M Ireland
- Department of Medicinal Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - Tomas Majtan
- Department of Pediatrics, University of Colorado, Aurora, CO 80045, USA
| | - Jan P Kraus
- Department of Pediatrics, University of Colorado, Aurora, CO 80045, USA
| | - Amy M Barrios
- Department of Medicinal Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA.
| |
Collapse
|
64
|
Winter G, Cordente AG, Curtin C. Formation of hydrogen sulfide from cysteine in Saccharomyces cerevisiae BY4742: genome wide screen reveals a central role of the vacuole. PLoS One 2014; 9:e113869. [PMID: 25517415 PMCID: PMC4269451 DOI: 10.1371/journal.pone.0113869] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 10/31/2014] [Indexed: 11/24/2022] Open
Abstract
Discoveries on the toxic effects of cysteine accumulation and, particularly, recent findings on the many physiological roles of one of the products of cysteine catabolism, hydrogen sulfide (H2S), are highlighting the importance of this amino acid and sulfur metabolism in a range of cellular activities. It is also highlighting how little we know about this critical part of cellular metabolism. In the work described here, a genome-wide screen using a deletion collection of Saccharomyces cerevisiae revealed a surprising set of genes associated with this process. In addition, the yeast vacuole, not previously associated with cysteine catabolism, emerged as an important compartment for cysteine degradation. Most prominent among the vacuole-related mutants were those involved in vacuole acidification; we identified each of the eight subunits of a vacuole acidification sub-complex (V1 of the yeast V-ATPase) as essential for cysteine degradation. Other functions identified included translation, RNA processing, folate-derived one-carbon metabolism, and mitochondrial iron-sulfur homeostasis. This work identified for the first time cellular factors affecting the fundamental process of cysteine catabolism. Results obtained significantly contribute to the understanding of this process and may provide insight into the underlying cause of cysteine accumulation and H2S generation in eukaryotes.
Collapse
Affiliation(s)
- Gal Winter
- School of Biomedical and Health Sciences, College of Health and Science, University of Western Sydney, Parramatta, NSW, Australia
- The Australian Wine Research Institute, Adelaide, SA, Australia
- Centre for Microbial Electrosynthesis (CEMES), University of Queensland, Brisbane, Queensland, Australia
- * E-mail:
| | | | - Chris Curtin
- The Australian Wine Research Institute, Adelaide, SA, Australia
| |
Collapse
|
65
|
Ried K, Fakler P. Potential of garlic (Allium sativum) in lowering high blood pressure: mechanisms of action and clinical relevance. Integr Blood Press Control 2014; 7:71-82. [PMID: 25525386 PMCID: PMC4266250 DOI: 10.2147/ibpc.s51434] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Garlic supplements have shown promise in the treatment of uncontrolled hypertension, lowering blood pressure (BP) by about 10 mmHg systolic and 8 mmHg diastolic, similar to standard BP medication. Aged garlic extract, which contains S-allylcysteine as the bioactive sulfur compound, in particular is standardizable and highly tolerable, with little or no known harmful interaction when taken with other BP-reducing or blood-thinning medication. Here we describe biologically plausible mechanisms of garlic's BP-lowering effect. Garlic-derived polysulfides stimulate the production of the vascular gasotransmitter hydrogen sulfide (H2S) and enhance the regulation of endothelial nitric oxide (NO), which induce smooth muscle cell relaxation, vasodilation, and BP reduction. Several dietary and genetic factors influence the efficiency of the H2S and NO signaling pathways and may contribute to the development of hypertension. Sulfur deficiency might play a part in the etiology of hypertension, and could be alleviated with supplementation of organosulfur compounds derived from garlic.
Collapse
Affiliation(s)
- Karin Ried
- National Institute of Integrative Medicine, Melbourne, VIC, Australia
| | - Peter Fakler
- National Institute of Integrative Medicine, Melbourne, VIC, Australia
| |
Collapse
|
66
|
Debreceni B, Debreceni L. The role of homocysteine-lowering B-vitamins in the primary prevention of cardiovascular disease. Cardiovasc Ther 2014; 32:130-8. [PMID: 24571382 DOI: 10.1111/1755-5922.12064] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality in the Western world. The effort of research should aim at the primary prevention of CVD. Alongside statin therapy, which is maintained to be an effective method of CVD prevention, there are alternative methods such as vitamin B substitution therapy with folic acid (FA), and vitamins B12 and B6 . B-vitamins may inhibit atherogenesis by decreasing the plasma level of homocysteine (Hcy)-a suspected etiological factor for atherosclerosis-and by other mechanisms, primarily through their antioxidant properties. Although Hcy-lowering vitamin trials have failed to demonstrate beneficial effects of B-vitamins in the prevention of CVD, a meta-analysis and stratification of a number of large vitamin trials have suggested their effectiveness in cardiovascular prevention (CVP) in some aspects. Furthermore, interpretation of the results from these large vitamin trials has been troubled by statin/aspirin therapy, which was applied along with the vitamin substitution, and FA fortification, both of which obscured the separate effects of vitamins in CVP. Recent research results have accentuated a new approach to vitamin therapy for CVP. Studies undertaken with the aim of primary prevention have shown that vitamin B substitution may be effective in the primary prevention of CVD and may also be an option in the secondary prevention of disease if statin therapy is accompanied by serious adverse effects. Further investigations are needed to determine the validity of vitamin substitution therapy before its introduction in the protocol of CVD prevention.
Collapse
Affiliation(s)
- Balazs Debreceni
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, Pecs, Hungary
| | | |
Collapse
|
67
|
Kalani A, Kamat PK, Givvimani S, Brown K, Metreveli N, Tyagi SC, Tyagi N. Nutri-epigenetics ameliorates blood-brain barrier damage and neurodegeneration in hyperhomocysteinemia: role of folic acid. J Mol Neurosci 2014; 52:202-15. [PMID: 24122186 DOI: 10.1007/s12031-013-0122-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 09/10/2013] [Indexed: 01/01/2023]
Abstract
Epigenetic mechanisms underlying nutrition (nutrition epigenetics) are important in understanding human health. Nutritional supplements, for example folic acid, a cofactor in one-carbon metabolism, regulate epigenetic alterations and may play an important role in the maintenance of neuronal integrity. Folic acid also ameliorates hyperhomocysteinemia, which is a consequence of elevated levels of homocysteine. Hyperhomocysteinemia induces oxidative stress that may epigenetically mediate cerebrovascular remodeling and leads to neurodegeneration; however, the mechanisms behind such alterations remain unclear. Therefore, the present study was designed to observe the protective effects of folic acid against hyperhomocysteinemia-induced epigenetic and molecular alterations leading to neurotoxic cascades. To test this hypothesis, we employed 8-weeks-old male wild-type (WT) cystathionine-beta-synthase heterozygote knockout methionine-fed (CBS+/− + Met), WT, and CBS+/− + Met mice supplemented with folic acid (FA) [WT + FA and CBS+/− + Met + FA, respectively, 0.0057-μg g−1 day−1 dose in drinking water/4 weeks]. Hyperhomocysteinemia in CBS+/− + Met mouse brain was accompanied by a decrease in methylenetetrahydrofolate reductase and an increase in S-adenosylhomocysteine hydrolase expression, symptoms of oxidative stress, upregulation of DNA methyltransferases, rise in matrix metalloproteinases, a drop in the tissue inhibitors of metalloproteinases, decreased expression of tight junction proteins, increased permeability of the blood-brain barrier, neurodegeneration, and synaptotoxicity. Supplementation of folic acid to CBS+/− + Met mouse brain led to a decrease in the homocysteine level and rescued pathogenic and epigenetic alterations, showing its protective efficacy against homocysteine-induced neurotoxicity.
Collapse
|
68
|
Park BS, Kim HW, Rhyu IJ, Park C, Yeo SG, Huh Y, Jeong NY, Jung J. Hydrogen sulfide is essential for Schwann cell responses to peripheral nerve injury. J Neurochem 2014; 132:230-42. [PMID: 25123509 DOI: 10.1111/jnc.12932] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 08/06/2014] [Accepted: 08/08/2014] [Indexed: 11/29/2022]
Abstract
Hydrogen sulfide (H2 S) functions as a physiological gas transmitter in both normal and pathophysiological cellular events. H2 S is produced from substances by three enzymes: cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (MST). In human tissues, these enzymes are involved in tissue-specific biochemical pathways for H2 S production. For example, CBS and cysteine aminotransferase/MST are present in the brain, but CSE is not. Thus, we examined the expression of H2 S production-related enzymes in peripheral nerves. Here, we found that CSE and MST/cysteine aminotransferase, but not CBS, were present in normal peripheral nerves. In addition, injured sciatic nerves in vivo up-regulated CSE in Schwann cells during Wallerian degeneration (WD); however, CSE was not up-regulated in peripheral axons. Using an ex vivo sciatic nerve explant culture, we found that the inhibition of H2 S production broadly prevented the process of nerve degeneration, including myelin fragmentation, axonal degradation, Schwann cell dedifferentiation, and Schwann cell proliferation in vitro and in vivo. Thus, these results indicate that H2 S signaling is essential for Schwann cell responses to peripheral nerve injury. Hydrogen sulfide (H2 S) functions as a physiological gas transmitter in both normal and pathophysiological cellular events. H2 S is produced from cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfur transferase (MST). Here, we found that CSE and MST/CAT were present in normal peripheral nerves. Injured static nerves in vivo up-regulated CSE in Schwann cells during Wallerian degeneration, but CSE was not up-regulated in peripheral axons.
Collapse
Affiliation(s)
- Byung Sun Park
- Department of Anatomy and Neurobiology, School of Medicine, Biomedical Science Institution, Kyung Hee University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Abstract
Hydrogen sulfide (H2S) is an endogenously produced gaseous signaling molecule that elicits a number of cytoprotective effects in mammalian species. H2S was originally considered toxic at elevated levels, but 15 years ago the labile molecule was discovered in mammalian tissue and termed a gasotransmitter, thus opening the door for research aimed towards understanding its physiologic nature. Since then, novel findings have depicted the beneficial aspects of H2S therapy, such as vasodilation, antioxidant upregulation, inflammation inhibition, and activation of anti-apoptotic pathways. These cytoprotective alterations effectively treat multiple forms of cardiac injury at the preclinical level of research. The field has progressed towards instituting novel H2S donors that prove more effective at activating the subsequent cardioprotective enhancements over longer time periods. As more findings explore the efficacy of H2S, research focused on detection of sulfhydrated targets is on the rise. Understanding the molecular mechanisms that stem from H2S treatment may lead the field towards powerful therapeutics in the clinical setting. This review will discuss the cytoprotective and cardioprotective effects of H2S therapy, provide analysis on the molecular alterations that lead to these enhancements, and explore recently developed therapeutics that may bring this gasotransmitter into the clinic in the near future.
Collapse
Affiliation(s)
- Larry A Barr
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine
| | | |
Collapse
|
70
|
Alterations of retinal vasculature in cystathionine-β-synthase heterozygous mice: a model of mild to moderate hyperhomocysteinemia. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2573-85. [PMID: 25016930 DOI: 10.1016/j.ajpath.2014.05.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/16/2014] [Accepted: 05/15/2014] [Indexed: 11/21/2022]
Abstract
Mild to moderate hyperhomocysteinemia is prevalent in humans and is implicated in neurovascular diseases, including recently in certain retinal diseases. Herein, we used hyperhomocysteinemic mice deficient in the Cbs gene encoding cystathionine-β-synthase (Cbs(+/-)) to evaluate retinal vascular integrity. The Cbs(+/+) (wild type) and Cbs(+/-) (heterozygous) mice (aged 16 to 52 weeks) were subjected to fluorescein angiography and optical coherence tomography to assess vasculature in vivo. Retinas harvested for cryosectioning or flat mount preparations were subjected to immunofluorescence microscopy to detect blood vessels (isolectin-B4), angiogenesis [anti-vascular endothelial growth factor (VEGF) and anti-CD105], gliosis [anti-glial fibrillary acidic protein (GFAP)], pericytes (anti-neural/glial antigen 2), blood-retinal barrier [anti-zonula occludens protein 1 (ZO-1) and anti-occludin], and hypoxia [anti-pimonidazole hydrochloride (Hypoxyprobe-1)]. Levels of VEGF, GFAP, ZO-1, and occludin were determined by immunoblotting. Results of these analyses showed a mild vascular phenotype in young mice, which progressed with age. Fluorescein angiography revealed progressive neovascularization and vascular leakage in Cbs(+/-) mice; optical coherence tomography confirmed new vessels in the vitreous by 1 year. Immunofluorescence microscopy demonstrated vascular patterns consistent with ischemia, including a capillary-free zone centrally and new vessels with capillary tufts midperipherally in older mice. This was associated with increased VEGF, CD105, and GFAP and decreased ZO-1/occludin levels in the Cbs(+/-) retinas. Retinal vein occlusion was observed in some Cbs(+/-) mouse retinas. We conclude that mild to moderate elevation of homocysteine in Cbs(+/-) mice is accompanied by progressive alterations in retinal vasculature characterized by ischemia, neovascularization, incompetent blood-retinal barrier, and vascular occlusion.
Collapse
|
71
|
Kalani A, Kamat PK, Familtseva A, Chaturvedi P, Muradashvili N, Narayanan N, Tyagi SC, Tyagi N. Role of microRNA29b in blood-brain barrier dysfunction during hyperhomocysteinemia: an epigenetic mechanism. J Cereb Blood Flow Metab 2014; 34:1212-22. [PMID: 24802332 PMCID: PMC4083388 DOI: 10.1038/jcbfm.2014.74] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 03/04/2014] [Accepted: 03/25/2014] [Indexed: 12/22/2022]
Abstract
Although blood-brain barrier (BBB) integrity is maintained by the cross-talk of endothelial cells, junction proteins, and neurogliovascular network, the epigenetic mechanisms behind BBB permeability are largely unknown. We are reporting for the first time miR29b-mediated regulation of BBB, which is a novel mechanism underlying BBB integrity. We hypothesize that miR29b regulates BBB dysfunction by regulating DNMT3b, which consequently regulates the levels of metalloproteinases, that can eat up the membrane and junction proteins leading to leaky vasculature. In addition, 5'-azacytidine (5'-aza) was used to test its efficacy on BBB permeability. Blood-brain barrier disruption model was created by using homocysteine, and in the models miR29b was identified to be most affected, by using microRNA RT(2)-qPCR array. MiR29b mimics and inhibitors also confirmed that miR29b regulates the levels DNMT3b and MMP9. In hyperhomocysteinemic cystathionine-β-synthase deficient (CBS(+/-)) mice with high brain vessel permeability, miR29b levels were also high as compared with wild-type (WT) mice. Interestingly, 5'-aza improved BBB permeability by decreasing the expression of miR29b. In conclusion, our data suggested miR29b-mediated regulation of BBB dysfunction through DNMT3b and MMP9. It also potentiates the use of microRNAs as candidates for future epigenetic therapies in the improvement of BBB integrity.
Collapse
Affiliation(s)
- Anuradha Kalani
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Pradip K Kamat
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Anastasia Familtseva
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Pankaj Chaturvedi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Nino Muradashvili
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Nithya Narayanan
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Neetu Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
72
|
Homocysteine in renovascular complications: hydrogen sulfide is a modulator and plausible anaerobic ATP generator. Nitric Oxide 2014; 41:27-37. [PMID: 24963795 DOI: 10.1016/j.niox.2014.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 11/23/2022]
Abstract
Homocysteine (Hcy) is a non-protein amino acid derived from dietary methionine. High levels of Hcy, known as hyperhomocysteinemia (HHcy) is known to cause vascular complications. In the mammalian tissue, Hcy is metabolized by transsulfuration enzymes to produce hydrogen sulfide (H2S). H2S, a pungent smelling gas was previously known for its toxic effects in the central nervous system, recent studies however has revealed protective effects in a variety of diseases including hypertension, diabetes, inflammation, atherosclerosis, and renal disease progression and failure. Interestingly, under stress conditions including hypoxia, H2S can reduce metabolic demand and also act as a substrate for ATP production. This review highlights some of the recent advances in H2S research as a potential therapeutic agent targeting renovascular diseases associated with HHcy.
Collapse
|
73
|
Fiorito G, Guarrera S, Valle C, Ricceri F, Russo A, Grioni S, Mattiello A, Di Gaetano C, Rosa F, Modica F, Iacoviello L, Frasca G, Tumino R, Krogh V, Panico S, Vineis P, Sacerdote C, Matullo G. B-vitamins intake, DNA-methylation of One Carbon Metabolism and homocysteine pathway genes and myocardial infarction risk: the EPICOR study. Nutr Metab Cardiovasc Dis 2014; 24:483-488. [PMID: 24418380 DOI: 10.1016/j.numecd.2013.10.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Several epidemiological studies highlighted the association between folate and B-vitamins low intake and cardiovascular diseases (CVD) risk. Contrasting results were reported on the relationship between folate intake and DNA-methylation. Folate and B-vitamins may modulate DNA-methylation of specific enzymes which are included in the One-Carbon Metabolism (OCM) and in the homocysteine (Hcy) pathways. The aim of the study was to evaluate whether DNA-methylation profiles of OCM and Hcy genes could modulate the myocardial infarction (MI) risk conferred by a low B-vitamins intake. METHODS AND RESULTS Study sample (206 MI cases and 206 matched controls) is a case-control study nested in the prospective EPIC cohort. Methylation levels of 33 candidate genes where extracted by the whole epigenome analysis (Illumina-HumanMethylation450K-BeadChip). We identified three differentially methylated regions in males (TCN2 promoter, CBS 5'UTR, AMT gene-body) and two in females (PON1 gene-body, CBS 5'UTR), each of them characterized by an increased methylation in cases. Functional in silico analysis suggested a decreased expression in cases. A Recursively Partitioned Mixture Model cluster algorithm identified distinct methylation profiles associated to different MI risk: high-risk vs. low-risk methylation profile groups, OR = 3.49, p = 1.87 × 10(-)(4) and OR = 3.94, p = 0.0317 in males and females respectively (multivariate logistic regression adjusted for classical CVD risk factors). Moreover, a general inverse relationship between B-vitamins intake and DNA-methylation of the candidate genes was observed. CONCLUSIONS Our findings support the hypothesis that DNA-methylation patterns in specific regions of OCM and Hcy pathways genes may modulate the CVD risk conferred by folate and B-vitamins low intake.
Collapse
Affiliation(s)
- G Fiorito
- Human Genetics Foundation, Torino, Italy; Department of Medical Sciences, University of Torino, Italy
| | - S Guarrera
- Human Genetics Foundation, Torino, Italy
| | - C Valle
- Human Genetics Foundation, Torino, Italy
| | - F Ricceri
- Human Genetics Foundation, Torino, Italy; Department of Medical Sciences, University of Torino, Italy
| | - A Russo
- Human Genetics Foundation, Torino, Italy; Department of Medical Sciences, University of Torino, Italy
| | - S Grioni
- Department of Preventive and Predictive Medicine, Epidemiology and Prevention Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, Milano, Italy
| | - A Mattiello
- Department of Clinical and Experimental Medicine, Federico II University, Napoli, Italy
| | - C Di Gaetano
- Human Genetics Foundation, Torino, Italy; Department of Medical Sciences, University of Torino, Italy
| | - F Rosa
- Human Genetics Foundation, Torino, Italy
| | - F Modica
- Human Genetics Foundation, Torino, Italy
| | - L Iacoviello
- Department of Epidemiology and Prevention, IRCCS Istituto Neurologico Mediterraneo Neuromed, Italy
| | - G Frasca
- Cancer Registry and Histopathology Unit, "Civile-M.P. Arezzo" Hospital, Ragusa, Italy
| | - R Tumino
- Cancer Registry and Histopathology Unit, "Civile-M.P. Arezzo" Hospital, Ragusa, Italy
| | - V Krogh
- Department of Preventive and Predictive Medicine, Epidemiology and Prevention Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, Milano, Italy
| | - S Panico
- Department of Clinical and Experimental Medicine, Federico II University, Napoli, Italy
| | - P Vineis
- Human Genetics Foundation, Torino, Italy; Epidemiology and Public Health, Imperial College London, UK
| | - C Sacerdote
- Human Genetics Foundation, Torino, Italy; Cancer Epidemiology, CPO-Piemonte, Torino, Italy
| | - G Matullo
- Human Genetics Foundation, Torino, Italy; Department of Medical Sciences, University of Torino, Italy.
| |
Collapse
|
74
|
Veeranki S, Givvimani S, Pushpakumar S, Tyagi SC. Hyperhomocysteinemia attenuates angiogenesis through reduction of HIF-1α and PGC-1α levels in muscle fibers during hindlimb ischemia. Am J Physiol Heart Circ Physiol 2014; 306:H1116-27. [PMID: 24585779 DOI: 10.1152/ajpheart.00003.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hyperhomocysteinemia (HHcy) is associated with elderly frailty, skeletal muscle injury and malfunction, reduced vascular integrity and function, and mortality. Although HHcy has been implicated in the impairment of angiogenesis after hindlimb ischemia in murine models, the underlying mechanisms are still unclear. We hypothesized that HHcy compromises skeletal muscle perfusion, collateral formation, and arteriogenesis by diminishing postischemic vasculogenic responses in muscle fibers. To test this hypothesis, we created femoral artery ligation in wild-type and heterozygous cystathionine β-synthase (CBS(+/-)) mice (a model for HHcy) and assessed tissue perfusion, collateral vessel formation, and skeletal muscle function using laser-Doppler perfusion imaging, barium angiography, and fatigue tests. In addition, we assessed postischemic levels of VEGF and levels of its muscle-specific regulators: hypoxia-inducible factor (HIF)-1α and peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α. The observations indicated dysregulation of VEGF, HIF-1α, and PGC-1α levels in ischemic skeletal muscles of CBS(+/-) mice. Concomitant with the reduced ischemic angiogenic responses, we also observed diminished leptin expression and attenuated Akt signaling in ischemic muscle fibers of CBS(+/-) mice. Moreover, there was enhanced atrogene, ubiquitin ligases that conjugate proteins for degradation during muscle atrophy, transcription, and reduced muscle function after ischemia in CBS(+/-) mice. These results suggest that HHcy adversely affects muscle-specific ischemic responses and contributes to muscle frailty.
Collapse
Affiliation(s)
- Sudhakar Veeranki
- Department of Physiology and Biophysics, University of Louisville, Louisville, Kentucky
| | | | | | | |
Collapse
|
75
|
Dharuri H, Henneman P, Demirkan A, van Klinken JB, Mook-Kanamori DO, Wang-Sattler R, Gieger C, Adamski J, Hettne K, Roos M, Suhre K, Van Duijn CM, van Dijk KW, 't Hoen PAC. Automated workflow-based exploitation of pathway databases provides new insights into genetic associations of metabolite profiles. BMC Genomics 2013; 14:865. [PMID: 24320595 PMCID: PMC3879060 DOI: 10.1186/1471-2164-14-865] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/02/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified many common single nucleotide polymorphisms (SNPs) that associate with clinical phenotypes, but these SNPs usually explain just a small part of the heritability and have relatively modest effect sizes. In contrast, SNPs that associate with metabolite levels generally explain a higher percentage of the genetic variation and demonstrate larger effect sizes. Still, the discovery of SNPs associated with metabolite levels is challenging since testing all metabolites measured in typical metabolomics studies with all SNPs comes with a severe multiple testing penalty. We have developed an automated workflow approach that utilizes prior knowledge of biochemical pathways present in databases like KEGG and BioCyc to generate a smaller SNP set relevant to the metabolite. This paper explores the opportunities and challenges in the analysis of GWAS of metabolomic phenotypes and provides novel insights into the genetic basis of metabolic variation through the re-analysis of published GWAS datasets. RESULTS Re-analysis of the published GWAS dataset from Illig et al. (Nature Genetics, 2010) using a pathway-based workflow (http://www.myexperiment.org/packs/319.html), confirmed previously identified hits and identified a new locus of human metabolic individuality, associating Aldehyde dehydrogenase family1 L1 (ALDH1L1) with serine/glycine ratios in blood. Replication in an independent GWAS dataset of phospholipids (Demirkan et al., PLoS Genetics, 2012) identified two novel loci supported by additional literature evidence: GPAM (Glycerol-3 phosphate acyltransferase) and CBS (Cystathionine beta-synthase). In addition, the workflow approach provided novel insight into the affected pathways and relevance of some of these gene-metabolite pairs in disease development and progression. CONCLUSIONS We demonstrate the utility of automated exploitation of background knowledge present in pathway databases for the analysis of GWAS datasets of metabolomic phenotypes. We report novel loci and potential biochemical mechanisms that contribute to our understanding of the genetic basis of metabolic variation and its relationship to disease development and progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Peter A C 't Hoen
- Center for Human and Clinical Genetics, Leiden University Medical Center, S4-P, PO Box 9600, 2300, RC Leiden, Netherlands.
| |
Collapse
|
76
|
Bhattacharyya S, Saha S, Giri K, Lanza IR, Nair KS, Jennings NB, Rodriguez-Aguayo C, Lopez-Berestein G, Basal E, Weaver AL, Visscher DW, Cliby W, Sood AK, Bhattacharya R, Mukherjee P. Cystathionine beta-synthase (CBS) contributes to advanced ovarian cancer progression and drug resistance. PLoS One 2013; 8:e79167. [PMID: 24236104 PMCID: PMC3827285 DOI: 10.1371/journal.pone.0079167] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 09/18/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Epithelial ovarian cancer is the leading cause of gynecologic cancer deaths. Most patients respond initially to platinum-based chemotherapy after surgical debulking, however relapse is very common and ultimately platinum resistance emerges. Understanding the mechanism of tumor growth, metastasis and drug resistant relapse will profoundly impact the therapeutic management of ovarian cancer. METHODS/PRINCIPAL FINDINGS Using patient tissue microarray (TMA), in vitro and in vivo studies we report a role of of cystathionine-beta-synthase (CBS), a sulfur metabolism enzyme in ovarian carcinoma. We report here that the expression of cystathionine-beta-synthase (CBS), a sulfur metabolism enzyme, is common in primary serous ovarian carcinoma. The in vitro effects of CBS silencing can be reversed by exogenous supplementation with the GSH and H2S producing chemical Na2S. Silencing CBS in a cisplatin resistant orthotopic model in vivo by nanoliposomal delivery of CBS siRNA inhibits tumor growth, reduces nodule formation and sensitizes ovarian cancer cells to cisplatin. The effects were further corroborated by immunohistochemistry that demonstrates a reduction of H&E, Ki-67 and CD31 positive cells in si-RNA treated as compared to scrambled-RNA treated animals. Furthermore, CBS also regulates bioenergetics of ovarian cancer cells by regulating mitochondrial ROS production, oxygen consumption and ATP generation. This study reports an important role of CBS in promoting ovarian tumor growth and maintaining drug resistant phenotype by controlling cellular redox behavior and regulating mitochondrial bioenergetics. CONCLUSION The present investigation highlights CBS as a potential therapeutic target in relapsed and platinum resistant ovarian cancer.
Collapse
Affiliation(s)
- Sanjib Bhattacharyya
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic Rochester, Minnesota, United States of America
| | - Sounik Saha
- Department of Pathology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, United States of America
| | - Karuna Giri
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic Rochester, Minnesota, United States of America
| | - Ian R. Lanza
- Division of Endocrinology, College of Medicine, Mayo Clinic Rochester, Minnesota, United States of America
| | - K. Sreekumar Nair
- Division of Endocrinology, College of Medicine, Mayo Clinic Rochester, Minnesota, United States of America
| | - Nicholas B. Jennings
- Department of Gynecologic Oncology, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Cristian Rodriguez-Aguayo
- Department of Cancer Biology, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Gabriel Lopez-Berestein
- Department of Cancer Biology, M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Eati Basal
- Department of Obstetrics and Gynecology, College of Medicine, Mayo Clinic Rochester, Minnesota, United States of America
| | - Amy L. Weaver
- Department of Biostatistics and Bioinformatics, College of Medicine, Mayo Clinic Rochester, Minnesota, United States of America
| | - Daniel W. Visscher
- Department of Pathology, College of Medicine, Mayo Clinic Rochester, Minnesota, United States of America
| | - William Cliby
- Department of Obstetrics and Gynecology, College of Medicine, Mayo Clinic Rochester, Minnesota, United States of America
| | - Anil K. Sood
- Department of Gynecologic Oncology, M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Cancer Biology, M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, United States of America
| | - Priyabrata Mukherjee
- Department of Pathology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
77
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
78
|
Hydrogen sulphide pathway contributes to the enhanced human platelet aggregation in hyperhomocysteinemia. Proc Natl Acad Sci U S A 2013; 110:15812-7. [PMID: 24019484 DOI: 10.1073/pnas.1309049110] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Homocysteine is metabolized to methionine by the action of 5,10 methylenetetrahydrofolate reductase (MTHFR). Alternatively, by the transulfuration pathway, homocysteine is transformed to hydrogen sulphide (H2S), through multiple steps involving cystathionine β-synthase and cystathionine γ-lyase. Here we have evaluated the involvement of H2S in the thrombotic events associated with hyperhomocysteinemia. To this purpose we have used platelets harvested from healthy volunteers or patients newly diagnosed with hyperhomocysteinemia with a C677T polymorphism of the MTHFR gene (MTHFR++). NaHS (0.1-100 µM) or l-cysteine (0.1-100 µM) significantly increased platelet aggregation harvested from healthy volunteers induced by thrombin receptor activator peptide-6 amide (2 µM) in a concentration-dependent manner. This increase was significantly potentiated in platelets harvested from MTHFR++ carriers, and it was reversed by the inhibition of either cystathionine β-synthase or cystathionine γ-lyase. Similarly, in MTHFR++ carriers, the content of H2S was significantly higher in either platelets or plasma compared with healthy volunteers. Interestingly, thromboxane A2 production was markedly increased in response to both NaHS or l-cysteine in platelets of healthy volunteers. The inhibition of phospholipase A2, cyclooxygenase, or blockade of the thromboxane receptor markedly reduced the effects of H2S. Finally, phosphorylated-phospholipase A2 expression was significantly higher in MTHFR++ carriers compared with healthy volunteers. In conclusion, the H2S pathway is involved in the prothrombotic events occurring in hyperhomocysteinemic patients.
Collapse
|
79
|
Serrano Díaz N, Páez MC, Bautista Niño PK, Díaz Martínez LA, Guío Mahecha E. Variables pre-analíticas que afectan las concentraciones de homocisteína: aplicación para biobancos con fines de investigación. MEDUNAB 2013. [DOI: 10.29375/01237047.1843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Objetivo: El presente estudio evaluó el impacto de las variables pre-analíticas sobre las concentraciones séricas de la Homocisteína, y su posible aplicación en biobancos con fines de investigación.
Metodología: En diez adultos voluntarios auto declarados sanos, se tomaron muestras de sangre periférica bajo diferentes condiciones de ayuno, posición de toma de la muestra (supino versus sentada) y diferentes intervalos de tiempo entre la toma y la separación definitiva de componentes. Todas las alícuotas fueron almacenadas a -800C en el biobanco hasta el momento de ser procesadas. La medición de homocisteína se hizo por duplicado en Immulite® 2000. Se realizó análisis de concordancia por medio de coeficiente de Lin (σ) y MANOVA.
Resultados: La medición de homocisteína es altamente reproducible (σ=0.908, IC95% 0.861 a 0.955), sin que el ayuno o el tiempo de centrifugación de la muestra afecte su concentración. Sin embrago, la posición al momento de la toma de muestra, implica una reducción media de 14.2% (IC95% 8.4% a 20.0%) en la concentración de Homocisteína en poción decúbito supino versus la toma en posición sentado.
Conclusión: La homocisteína es un biomarcador estable, sin que su valor se vea alterado por variables pre-analíticas como los tiempos entre toma de muestra, centrifugación y separación de componentes (almacenamiento temporal a 4°C). Sin embargo la postura del participante al momento de la toma de muestra produce una variabilidad significativa. Estos hallazgos reiteran el papel de un biobanco en la estandarización de los procesos de toma, manipulación, almacenamiento y gestión con criterios de excelencia.
Collapse
|
80
|
Rhodehouse BC, Mayo JN, Beard RS, Chen CH, Bearden SE. Opening of the blood-brain barrier before cerebral pathology in mild hyperhomocysteinemia. PLoS One 2013; 8:e63951. [PMID: 23696861 PMCID: PMC3655957 DOI: 10.1371/journal.pone.0063951] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 04/08/2013] [Indexed: 12/11/2022] Open
Abstract
Hyperhomocysteinemia (HHcy) is a risk factor for cognitive impairment. The purpose of this study was to determine the temporal pattern of cerebral pathology in a mouse model of mild HHcy, because understanding this time course provides the basis for understanding the mechanisms involved. C57Bl/6 mice with heterozygous deletion cystathionine β-synthase (cbs+/−; Het) were used as a model of mild HHcy along with their wild-type littermates (cbs+/+; WT). Mice were ‘young’ (5.3±0.2 months of age) and ‘old’ (16.6±0.9 months of age). Blood-brain barrier (BBB) permeability was quantified from Evans blue and sodium fluorescein extravasation. Microvascular architecture was assessed by z-stack confocal microscopy. Leukoaraiosis was measured from Luxol fast blue stained slides of paraffin brain sections. Inflammation was quantified using standard antibody-based immunohistochemical techniques. Cognitive function was assessed using the Morris water maze. BBB permeability was significantly greater in Het vs. WT mice at all ages (p<0.05). There were no differences in microvascular architecture among the groups. Compared with all other groups, old Het mice had significantly greater leukoaraiosis, inflammation in the fornix, and cognitive impairment (p<0.05). In mild HHcy, increased permeability of the BBB precedes the onset of cerebral pathology. This new paradigm may play a role in the progression of disease in HHcy.
Collapse
Affiliation(s)
- Bryce C. Rhodehouse
- Department of Biological Sciences, Idaho State University, Pocatello, Idaho, United States of America
| | - Jamie N. Mayo
- Department of Biological Sciences, Idaho State University, Pocatello, Idaho, United States of America
| | - Richard S. Beard
- Department of Biological Sciences, Idaho State University, Pocatello, Idaho, United States of America
| | - Cheng-Hung Chen
- Department of Biological Sciences, Idaho State University, Pocatello, Idaho, United States of America
| | - Shawn E. Bearden
- Department of Biological Sciences, Idaho State University, Pocatello, Idaho, United States of America
- ISU Biomedical Research Institute, Idaho State University, Pocatello, Idaho, United States of America
- * E-mail:
| |
Collapse
|
81
|
Ma S, Zhang H, Sun W, Gong H, Wang Y, Ma C, Wang J, Cao C, Yang X, Tian J, Jiang Y. Hyperhomocysteinemia induces cardiac injury by up-regulation of p53-dependent Noxa and Bax expression through the p53 DNA methylation in ApoE(-/-) mice. Acta Biochim Biophys Sin (Shanghai) 2013; 45:391-400. [PMID: 23619570 DOI: 10.1093/abbs/gmt030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hyperhomocysteinemia (HHcy) is a risk factor for cardiovascular disease and has a strong correlation with heart failure. However, the effects of HHcy on cardiac tissue remain less well understood. To elucidate the role of p53-dependent apoptosis in HHcy-induced cardiac injury, we fed ApoE(-/-) mice with high methionine diet to establish HHcy model. Serum Hcy, cardiac enzymes, and lipids were measured. The protein levels of Noxa, DNMT1, caspases-3/9, and p53 were determined by enzyme-linked immunosorbent assay. Bcl-2 and Bax proteins were detected by immunohistochemistry staining. S-adenosyl methionine and S-adenosyl homocysteine concentrations were determined by high-performance liquid chromatography. The mRNA levels of p53 and DNMT1 were analyzed by real-time polymerase chain reaction (PCR) and the methylation levels of p53 were analyzed by nested methylation-specific-PCR. Our data showed that the concentrations of serum Hcy and lipids were increased in Meth group compared with the N-control group, which indicated that the model was established successfully. The expression levels of p53 and Noxa were increased in Meth group, while the methylation status of p53 was hypomethylation. The activities of caspase-3/9 were increased in Meth group compared with the N-control group. In addition, immunohistochemistry staining showed that the expression of Bax was significantly increased in Meth and Meth-F group compared with the N-control group. In summary, HHcy induces cardiac injury by up-regulation of p53-dependent pro-apoptotic related genes Noxa and Bax, while p53 DNA hypomethylation is a key molecular mechanism in pathological process induced by HHcy.
Collapse
Affiliation(s)
- Shengchao Ma
- Department of Laboratory Medical, Ningxia Medical University, Yinchuan 750004, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Mangoni AA, Zinellu A, Carru C, Attia JR, McEvoy M. Serum thiols and cardiovascular risk scores: a combined assessment of transsulfuration pathway components and substrate/product ratios. J Transl Med 2013; 11:99. [PMID: 23587204 PMCID: PMC3637611 DOI: 10.1186/1479-5876-11-99] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 04/09/2013] [Indexed: 12/22/2022] Open
Abstract
Background Serum thiols have shown associations with surrogate markers of cardiovascular disease. However, little information is available on their combined association with validated cardiovascular risk scores for primary prevention at population level. We sought to determine whether individual serum thiol concentrations and substrate/product ratios within the transsulfuration pathway are independently associated with such scores. Methods Data on clinical and demographic characteristics, serum thiols (homocysteine, cysteine, taurine, glutamylcysteine, total glutathione and cysteinylglycine) and high-sensitivity C-reactive protein (CRP) were collected from a sample of the Hunter Community Study without previous cardiovascular events [n=350, median age (IQR) = 62 (59–66) years]. Five-year absolute cardiovascular risk score for each subject was calculated using the Framingham Risk Equation. Results Median risk score was 7% (IQR 4–10). After adjusting for body mass index, estimated glomerular filtration rate and physical activity regression analysis showed independent associations between cardiovascular risk scores and a) higher serum homocysteine (B 0.066, 95% CI 0.040 to 0.091, P<0.001) and lower cysteine (B −0.003, 95% CI −0.005 to −0.001, P=0.003) and glutathione (B −0.029, 95% CI −0.056 to −0.003, P=0.03) concentrations; and b) higher homocysteine/cysteine (B 0.114, 95% CI 0.066 to 0.161, P<0.001) and lower glutathione/cysteinylglycine (B −1.145, 95% CI −2.030 to −0.260, P=0.011) ratios. No significant associations were observed between cardiovascular risk scores, taurine and CRP. Conclusions Serum homocysteine, cysteine and glutathione are independently associated with cardiovascular risk scores at population level. Enzymatic pathways involved in reduced bioconversion of homocysteine into cysteine and increased glutathione degradation might play an important role in such associations.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Division of Applied Medicine, Section of Translational Medical Sciences, School of Medicine and Dentistry, University of Aberdeen, Polwarth Building, Foresterhill, Aberdeen AB25 2ZD, UK.
| | | | | | | | | |
Collapse
|
83
|
Tawfik A, Al-Shabrawey M, Roon P, Sonne S, Covar JA, Matragoon S, Ganapathy PS, Atherton SS, El-Remessy A, Ganapathy V, Smith SB. Alterations of retinal vasculature in cystathionine-Beta-synthase mutant mice, a model of hyperhomocysteinemia. Invest Ophthalmol Vis Sci 2013; 54:939-49. [PMID: 23307965 DOI: 10.1167/iovs.12-10536] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Mice with moderate/severe hyperhomocysteinemia due to deficiency or absence of the cbs gene encoding cystathionine-beta-synthase (CBS) have marked retinal disruption, ganglion cell loss, optic nerve mitochondrial dysfunction, and ERG defects; those with mild hyperhomocysteinemia have delayed retinal morphological/functional phenotype. Excess homocysteine is a risk factor for cardiovascular diseases; however, it is not known whether excess homocysteine alters retinal vasculature. METHODS Cbs(+/+), cbs(+/-), and cbs(-/-) mice (age ∼3 weeks) were subjected to angiography; retinas were harvested for cryosections, flat-mount preparations, or trypsin digestion and subjected to immunofluorescence microscopy to visualize vessels using isolectin-B4, to detect angiogenesis using anti-VEGF and anti-endoglin (anti-CD105) and activated glial cells (anti-glial fibrillary acidic protein [anti-GFAP]) and to investigate the blood-retinal barrier using the tight junction markers zonula occludens-1 (ZO-1) and occludin. Expression of vegf was determined by quantitative RT-PCR (qRT-PCR) and immunoblotting. Human retinal endothelial cells (HRECs) were treated with excess homocysteine to analyze permeability. RESULTS Angiography revealed vascular leakage in cbs(-/-) mice; immunohistochemical analysis demonstrated vascular patterns consistent with ischemia; isolectin-B4 labeling revealed a capillary-free zone centrally and new vessels with capillary tufts midperipherally. This was associated with increased vegf mRNA and protein, CD105, and GFAP in cbs(-/-) retinas concomitant with a marked decrease in ZO-1 and occludin. Homocysteine-treated HRECs showed increased permeability. CONCLUSIONS Severe elevation of homocysteine in cbs(-/-) mutant mice is accompanied by alterations in retinal vasculature (ischemia, neovascularization, and incompetent blood-retinal barrier). The marked disruption of retinal structure and decreased visual function reported in cbs(-/-) mice may reflect vasculopathy as well as neuropathy.
Collapse
Affiliation(s)
- Amany Tawfik
- Departments of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Lin VS, Chang CJ. Fluorescent probes for sensing and imaging biological hydrogen sulfide. Curr Opin Chem Biol 2012; 16:595-601. [PMID: 22921406 DOI: 10.1016/j.cbpa.2012.07.014] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 07/21/2012] [Accepted: 07/26/2012] [Indexed: 10/28/2022]
Abstract
Hydrogen sulfide (H(2)S) has long been recognized as a toxic molecule in biological systems. However, emerging studies now link controlled fluxes of this reactive sulfur species to cellular regulation and signaling events akin to other small molecule messengers, such as nitric oxide, hydrogen peroxide, and carbon monoxide. Progress in the development of fluorescent small-molecule indicators with high selectivity for hydrogen sulfide offers a promising approach for studying its production, trafficking, and downstream physiological and/or pathological effects.
Collapse
Affiliation(s)
- Vivian S Lin
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
85
|
Jain SK. L-cysteine supplementation as an adjuvant therapy for type-2 diabetes. Can J Physiol Pharmacol 2012; 90:1061-4. [PMID: 22783875 DOI: 10.1139/y2012-087] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diabetes remains a major public health issue. According to the American Diabetes Association, 23.5 million, or 10.7% of people in the USA aged 20 years and older, have diabetes. Type-2 diabetes is treated both by controlling the diet and with oral hypoglycemic drugs. However, for many patients, achieving a tight control of glucose is difficult with current regimens. This chapter discusses a relatively low-cost dietary supplement that could be used as an adjuvant therapy for type-2 diabetes. A review of the literature indicates that cysteine-rich whey protein improves glucose metabolism in diabetic animals and type-2 diabetic patients. Similarly, in animal studies, improvement in glucose metabolism is observed after supplementation with L-cysteine, or molecules containing a cysteine moiety. This chapter discusses the biochemical mechanisms by which L-cysteine can upregulate the insulin-dependent signaling cascades of glucose metabolism. Further studies are needed to examine whether clinical interventions using L-cysteine as an adjuvant therapy indeed help to control glycemia and vascular inflammation in the diabetic patient population.
Collapse
Affiliation(s)
- Sushil K Jain
- Department of Pediatrics, LSU Health Sciences Center, Shreveport, LA 71130, USA.
| |
Collapse
|
86
|
Abstract
Hydrogen sulfide (H(2)S) has recently emerged as a mammalian gaseous messenger molecule, akin to nitric oxide and carbon monoxide. H(2)S is predominantly formed from Cys or its derivatives by the enzymes cystathionine β-synthase and cystathionine γ-lyase. One of the mechanisms by which H(2)S signals is by sulfhydration of reactive Cys residues in target proteins. Although analogous to protein nitrosylation, sulfhydration is substantially more prevalent and usually increases the catalytic activity of targeted proteins. Physiological actions of sulfhydration include the regulation of inflammation and endoplasmic reticulum stress signalling as well as of vascular tension.
Collapse
Affiliation(s)
- Bindu D Paul
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
87
|
Mayo JN, Beard RS, Price TO, Chen CH, Erickson MA, Ercal N, Banks WA, Bearden SE. Nitrative stress in cerebral endothelium is mediated by mGluR5 in hyperhomocysteinemia. J Cereb Blood Flow Metab 2012; 32:825-34. [PMID: 22186670 PMCID: PMC3345916 DOI: 10.1038/jcbfm.2011.185] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hyperhomocysteinemia (HHcy) disrupts nitric oxide (NO) signaling and increases nitrative stress in cerebral microvascular endothelial cells (CMVECs). This is mediated, in part, by protein nitrotyrosinylation (3-nitrotyrosine; 3-NT) though the mechanisms by which extracellular homocysteine (Hcy) generates intracellular 3-NT are unknown. Using a murine model of mild HHcy (cbs(+/-) mouse), we show that 3-NT is significantly elevated in cerebral microvessels with concomitant reductions in serum NO bioavailability as compared with wild-type littermate controls (cbs(+/+)). Directed pharmacology identified a receptor-dependent mechanism for 3-NT formation in CMVECs. Homocysteine increased expression of inducible NO synthase (iNOS) and formation of 3-NT, both of which were blocked by inhibition of metabotropic glutamate receptor-5 (mGluR5) with the specific antagonist 2-methyl-6-(phenylethynyl) pyridine hydrochloride. Activation of mGluR5 is both sufficient and necessary to drive the nitrative stress because direct activation using the mGluR5-specific agonist (RS)-2-chloro-5-hydroxyphenylglycine also increased iNOS expression and 3-NT formation while knockdown of mGluR5 receptor expression by short hairpin RNA (shRNA) blocked their increase in response to Hcy. Nitric oxide derived from iNOS was required for Hcy-mediated formation of 3-NT because the effect was blocked by 1400W. These results provide the first evidence for a receptor-dependent process that explains how plasma Hcy levels control intracellular nitrative stress in cerebral microvascular endothelium.
Collapse
Affiliation(s)
- Jamie N Mayo
- Department of Biological Sciences, Idaho State University, Pocatello, ID 83209, USA
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Sen U, Sathnur PB, Kundu S, Givvimani S, Coley DM, Mishra PK, Qipshidze N, Tyagi N, Metreveli N, Tyagi SC. Increased endogenous H2S generation by CBS, CSE, and 3MST gene therapy improves ex vivo renovascular relaxation in hyperhomocysteinemia. Am J Physiol Cell Physiol 2012; 303:C41-51. [PMID: 22517358 DOI: 10.1152/ajpcell.00398.2011] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Hydrogen sulfide (H(2)S) has recently been identified as a regulator of various physiological events, including vasodilation, angiogenesis, antiapoptotic, and cellular signaling. Endogenously, H(2)S is produced as a metabolite of homocysteine (Hcy) by cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (3MST). Although Hcy is recognized as vascular risk factor at an elevated level [hyperhomocysteinemia (HHcy)] and contributes to vascular injury leading to renovascular dysfunction, the exact mechanism is unclear. The goal of the current study was to investigate whether conversion of Hcy to H(2)S improves renovascular function. Ex vivo renal artery culture with CBS, CSE, and 3MST triple gene therapy generated more H(2)S in the presence of Hcy, and these arteries were more responsive to endothelial-dependent vasodilation compared with nontransfected arteries treated with high Hcy. Cross section of triple gene-delivered renal arteries immunostaining suggested increased expression of CD31 and VEGF and diminished expression of the antiangiogenic factor endostatin. In vitro endothelial cell culture demonstrated increased mitophagy during high levels of Hcy and was mitigated by triple gene delivery. Also, dephosphorylated Akt and phosphorylated FoxO3 in HHcy were reversed by H(2)S or triple gene delivery. Upregulated matrix metalloproteinases-13 and downregulated tissue inhibitor of metalloproteinase-1 in HHcy were normalized by overexpression of triple genes. Together, these results suggest that H(2)S plays a key role in renovasculopathy during HHcy and is mediated through Akt/FoxO3 pathways. We conclude that conversion of Hcy to H(2)S by CBS, CSE, or 3MST triple gene therapy improves renovascular function in HHcy.
Collapse
Affiliation(s)
- Utpal Sen
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Kentucky, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Tetrahydrocurcumin ameliorates homocysteinylated cytochrome-c mediated autophagy in hyperhomocysteinemia mice after cerebral ischemia. J Mol Neurosci 2012; 47:128-38. [PMID: 22212488 DOI: 10.1007/s12031-011-9695-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 12/08/2011] [Indexed: 12/21/2022]
Abstract
High levels of homocysteine (Hcy) known as hyperhomocysteinemia (HHcy), contribute to autophagy and ischemia/reperfusion injury (I/R). Previous studies have shown that I/R injury and HHcy cause increased cerebrovascular permeability; however, the associated mechanism remains obscure. Interestingly, during HHcy, cytochome-c becomes homocysteinylated (Hcy-cyto-c). Cytochrome-c (cyto-c) transports electrons and facilitates bioenergetics in the system. However, its role in autophagy during ischemia/reperfusion injury is unclear. Tetrahydrocurcumin (THC) is a major herbal antioxidant and anti-inflammatory agent. Therefore, the objective of this study was to determine whether THC ameliorates autophagy during ischemia/reperfusion injury by reducing homocysteinylation of cyto-c in hyperhomocysteinemia pathological condition. To test this hypothesis, we employed 8-10-week-old male cystathionine-beta-synthase heterozygote knockout (CBS⁺/⁻) mice (genetically hyperhomocystemic mice). Experimental group was: CBS⁺/⁻, CBS⁺/⁻ + THC (25 mg/kg in 0.1% DMSO dose); CBS ⁺/⁻/I/R, and CBS⁺/⁻/I/R + THC (25 mg/kg in 0.1% DMSO dose). Ischemia was performed for 30 min and reperfusion for 72 h. THC was injected intra-peritoneally (I.P.) once daily for a period of 3 days after 30 min of ischemia. The infarct area was measured using 2,3,5-triphenyltetrazolium chloride staining. Permeability was determined by brain edema and Evans Blue extravasation. The brain tissues were analyzed for oxidative stress, matrix metalloproteinase-9 (MMP-9), damage-regulated autophagy modulator (DRAM), and microtubule-associated protein 1 light chain 3 (LC3) by Western blot. The mRNA levels of S-adenosyl-L-homocysteine hydrolases (SAHH) and methylenetetrahydrofolate reductase (MTHFR) genes were measured by quantitative real-time polymerase chain reaction. Co-immunoprecipitation was used to determine the homocysteinylation of cyto-c. We found that brain edema and Evans Blue leakage were reduced in I/R + THC-treated groups as compared to sham-operated groups along with reduced brain infarct size. THC also decreased oxidative damage and ameliorated the homocysteinylation of cyto-c in-part by MMP-9 activation which leads to autophagy in I/R groups as compared to sham-operated groups. This study suggests a potential therapeutic role of dietary THC in cerebral ischemia.
Collapse
|
90
|
Basu P, Qipshidze N, Tyagi SC, Sen U. Remodeling in vein expresses arterial phenotype in hyperhomocysteinemia. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2011; 3:266-279. [PMID: 22162783 PMCID: PMC3230260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 10/26/2011] [Indexed: 05/31/2023]
Abstract
Accumulating evidences suggest that homocysteine, a non-protein amino acid, is involved in vessel remodeling and blood flow at elevated level, although the exact mechanism is unclear. Here we hypothesized that homocysteine affects vein in such a way that vein develops arterial phenotype. We tested our hypothesis employing wild type (WT, C57BL/6J) and CBS+/- (cystathionine β-synthase heterozygote, a genetic model of hyperhomocysteinemia) supplemented with or without folic acid (FA, a homocysteine lowering agent). Vena cava blood flow was measured by ultrasound transonic flow probe. Tissue collagen and elastin were detected by histochemistry. Super oxide was detected by dihydroethidium (DHE) staining. Expressions of MMP-2, -9, -12, TIMP -2,-4, were measured by Western blot. MMP-13, TIMP-1, -3, and vein and aortic markers, EphB4 and EphrinB2, respectively were measured by RT-PCR. The results indicated relatively low blood flow and significant increase of collagen/elastin ratio in the CBS+/- mice compared to WT. Although FA treatment did not alter blood flow in CBS+/- mice, the collagen/elastin ratio was normalized. A relatively increased content of super oxide and gelatinase activity was observed in CBS+/- vena cava vs WT and normalized by FA treatment. Western blot analyses showed significant increase in MMP-9,-12 and decrease in TIMP-2, -4 expressions. Expressions of MMP-13, TIMP-1 and -3, Ephrin B2 were increased, whereas EphB4 was decreased with reverse change in FA treatment, with no change in MMP-13 and TIMP-1. We conclude that chronic HHcy causes vascular remodeling that expresses arterial phenotype in vein.
Collapse
Affiliation(s)
- Poulami Basu
- Department of Physiology and Biophysics, University of Louisville School of Medicine Louisville, KY, USA
| | | | | | | |
Collapse
|
91
|
Truss NJ, Warner TD. Gasotransmitters and platelets. Pharmacol Ther 2011; 132:196-203. [DOI: 10.1016/j.pharmthera.2011.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 06/15/2011] [Indexed: 10/18/2022]
|
92
|
Hyperhomocysteinemia increases permeability of the blood-brain barrier by NMDA receptor-dependent regulation of adherens and tight junctions. Blood 2011; 118:2007-14. [PMID: 21705496 DOI: 10.1182/blood-2011-02-338269] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hyperhomocysteinemia (HHcy) increases permeability of the blood-brain barrier, but the mechanisms are undetermined. Homocysteine (Hcy) is an agonist of the neuronal N-methyl-D-aspartate receptor (NMDAr). We tested the hypothesis that HHcy disrupts the blood-brain barrier by an NMDAr-dependent mechanism in endothelium. In brain microvascular endothelial cells, there was no change in expression of the adherens junction protein VE-cadherin with Hcy treatment, but there was a significant decrease in the amount of β-catenin at the membrane. Moreover, Hcy caused nuclear translocation of β-catenin and attachment to the promoter for the tight junction protein claudin-5, with concomitant reduction in claudin-5 expression. Using a murine model of HHcy (cbs(+/-)), treatment for 2 weeks with an NMDAr antagonist (memantine) rescued cerebrovascular expression of claudin-5 and blood-brain barrier permeability to both exogenous sodium fluorescein and endogenous IgG. Memantine had no effect on these parameters in wild-type littermates. The same results were obtained using an in vitro model with brain microvascular endothelial cells. These data provide the first evidence that the NMDAr is required for Hcy-mediated increases in blood-brain barrier permeability. Modulating cerebral microvascular NMDAr activity may present a novel therapeutic target in diseases associated with opening of the blood-brain barrier in HHcy, such as stroke and dementia.
Collapse
|
93
|
Abstract
Hydrogen sulfide (H(2)S) has been known as a highly toxic gas for several centuries. There have been considerable advances made in the H(2)S field regarding its physiological role; however, there is much more work that needs to be done. The biosynthesis of H(2)S has been attributed to three endogenous enzymes: cystathionine β-synthase (CBS), cystathionine γ-lyase (CGL or CSE) and 3-mercaptopyruvate sulfurtransferase (3-MST). These enzymes require further investigation to more fully elucidate the cellular expression profile, regulation and precise role of these critical enzymes in the production of H(2)S. In recent years, H(2)S has been demonstrated to have cytoprotective effects in multiple organ systems. In particular, it has been demonstrated that the administration of H(2)S either prior to ischaemia or at reperfusion significantly ameliorates myocardial and hepatic ischaemia-reperfusion injury. Therefore, this review focuses on the cardioprotective and hepatoprotective role of H(2)S. In addition, the review provides a summary of several known molecular targets of H(2)S protection.
Collapse
Affiliation(s)
- Adrienne L King
- Department of Surgery, Division of Cardiothoracic Surgery, The Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA 30033, USA
| | | |
Collapse
|