1
|
Malaviya P, Kowluru RA. Homocysteine and mitochondrial quality control in diabetic retinopathy. EYE AND VISION (LONDON, ENGLAND) 2024; 11:5. [PMID: 38229140 PMCID: PMC10790378 DOI: 10.1186/s40662-023-00362-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/08/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND Diabetic retinopathy is a progressive disease, and one of the key metabolic abnormalities in the pathogenesis of diabetic retinopathy, mitochondrial damage, is also influenced by the duration of hyperglycemia. Mitochondrial quality control involves a coordination of mitochondrial dynamics, biogenesis and removal of the damaged mitochondria. In diabetes, these processes are impaired, and the damaged mitochondria continue to produce free radicals. Diabetic patients also have high homocysteine and reduced levels of hydrogen sulfide, and hyperhomocysteinemia is shown to exacerbate diabetes-induced mitochondrial damage and worsen their dynamics. This study aims to investigate the temporal relationship between hyperhomocysteinemia and retinal mitochondrial quality control in diabetic retinopathy. METHODS Human retinal endothelial cells incubated in 20 mM D-glucose for 24 to 96 h, in the absence or presence of 100 µM homocysteine, with/without a hydrogen sulfide donor GYY4137, were analyzed for mitochondrial ROS (MitoSox fluorescence), DNA damage (transcripts of mtDNA-encoded ND6 and CytB), copy numbers, oxygen consumption rate (Seahorse XF analyzer) and mitophagy (mitophagosomes immunofluorescence labeling and flow cytometry). Results were confirmed in the retina from mice genetically manipulated for hyperhomocysteinemia (cystathionine β-synthase deficient mice, Cbs+/-), streptozotocin-induced diabetic for 8 to 24 weeks. At 24 weeks of diabetes, vascular health was evaluated by counting acellular capillaries in the trypsin digested retinal vasculature and by fluorescein angiography. RESULTS Homocysteine, in high glucose medium, exacerbated mitochondrial ROS production, mtDNA damage and impaired mitochondrial respiration within 24 h, and slowed down/worsened mitochondrial biogenesis and mitophagy, as compared to 48 to 96 h in high glucose alone. GYY4137 supplementation ameliorated homocysteine + high glucose-induced mitochondrial damage and impairment in biogenesis and mitophagy. Similar results were obtained from Cbs+/- mice-mitochondrial ROS, mtDNA damage and decline in biogenesis and mitophagy were observed within eight weeks of diabetes vs. 16 to 24 weeks of diabetes in Cbs+/+ mice, and at 24 weeks of diabetes, Cbs+/- mice had significantly higher acellular capillaries and vascular leakage. CONCLUSIONS Hyperhomocysteinemia, in a hyperglycemic environment, overwhelms the mitochondria, accelerating and exacerbating their dysfunction, and also delays/worsens their removal, augmenting the development of diabetic retinopathy. Thus, our results strengthen the importance of maintaining homocysteine-hydrogen sulfide balance during the early stages of diabetes for a patient to prevent/retard vision loss.
Collapse
Affiliation(s)
- Pooja Malaviya
- Department of Ophthalmology, Visual Sciences and Anatomical Sciences, Wayne State University, 4717 St. Antoine, Detroit, MI, 48201, USA
| | - Renu A Kowluru
- Department of Ophthalmology, Visual Sciences and Anatomical Sciences, Wayne State University, 4717 St. Antoine, Detroit, MI, 48201, USA.
| |
Collapse
|
2
|
Zhao H, Gao H, Zhang Y, Lan T, Wang J, Zhao H, Zhang H, Xue M, Liang H. Folic Acid Protects against Ethanol-Induced Hepatic Mitophagy Imbalance by ROS Scavenging and Attenuating the Elevated Hcy Levels. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14276-14288. [PMID: 37738285 DOI: 10.1021/acs.jafc.3c01851] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
Ample evidence indicates that ethanol-induced oxidative stress and mitochondrial dysfunction are central to the pathogenesis of alcoholic liver disease (ALD). As an adaptive quality control mechanism, mitophagy removes dysfunctional mitochondria to avert hepatic lesions in ALD. Folic acid exhibits potential radical scavenging properties and has been proven to ameliorate mitochondrial disorder in oxidative stress-related diseases. In this study, we aimed to uncover the mitophagy regulatory effects of folic acid in a 10w alcohol C57BL/6J mice feeding model (56% v/v) and L02 cells model cultured with ethanol (2.5% v/v). The results showed that folic acid alleviates ethanol-induced liver injury, decreasing oxidative stress and restoring liver enzyme. Furthermore, folic acid improved the mitochondrial function and inhibited ethanol-activated mitophagy through decreasing PINK1-Parkin and Drp1 expression, which inhibited the release of mitochondrial cytochrome C to the cytoplasm, preventing hepatocyte apoptosis. Intriguingly, folic acid attenuates the elevated hepatic homocysteine (Hcy) level. Additionally, the pretreatment of L02 cells with folic acid also ameliorated Hcy-induced oxidative damage, mitochondrial fission, and mitophagy. In summary, these results suggest that folic acid has beneficial effects in mitophagy remodeling by ROS scavenging and facilitating Hcy metabolism and could be developed as a potential therapeutic agent against ALD.
Collapse
Affiliation(s)
- Huichao Zhao
- Qingdao Municipal Hospital, Qingdao University. Qingdao 266071, China
| | - Haiqi Gao
- Department of Biochemistry and Molecular Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Yabin Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao 266071, China
| | - Tongtong Lan
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao 266071, China
| | - Jingwen Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao 266071, China
| | - Haifeng Zhao
- Qingdao Institute of Food and Drug Inspection, Qingdao 266071, China
| | - Huaqi Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao 266071, China
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Hui Liang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao 266071, China
| |
Collapse
|
3
|
Jiménez-Loygorri JI, Benítez-Fernández R, Viedma-Poyatos Á, Zapata-Muñoz J, Villarejo-Zori B, Gómez-Sintes R, Boya P. Mitophagy in the retina: Viewing mitochondrial homeostasis through a new lens. Prog Retin Eye Res 2023; 96:101205. [PMID: 37454969 DOI: 10.1016/j.preteyeres.2023.101205] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
Mitochondrial function is key to support metabolism and homeostasis in the retina, an organ that has one of the highest metabolic rates body-wide and is constantly exposed to photooxidative damage and external stressors. Mitophagy is the selective autophagic degradation of mitochondria within lysosomes, and can be triggered by distinct stimuli such as mitochondrial damage or hypoxia. Here, we review the importance of mitophagy in retinal physiology and pathology. In the developing retina, mitophagy is essential for metabolic reprogramming and differentiation of retina ganglion cells (RGCs). In basal conditions, mitophagy acts as a quality control mechanism, maintaining a healthy mitochondrial pool to meet cellular demands. We summarize the different autophagy- and mitophagy-deficient mouse models described in the literature, and discuss the potential role of mitophagy dysregulation in retinal diseases such as glaucoma, diabetic retinopathy, retinitis pigmentosa, and age-related macular degeneration. Finally, we provide an overview of methods used to monitor mitophagy in vitro, ex vivo, and in vivo. This review highlights the important role of mitophagy in sustaining visual function, and its potential as a putative therapeutic target for retinal and other diseases.
Collapse
Affiliation(s)
- Juan Ignacio Jiménez-Loygorri
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain.
| | - Rocío Benítez-Fernández
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain; Departament of Neuroscience and Movement Science, Faculty of Science and Medicine, University of Fribourg, 1700, Fribourg, Switzerland
| | - Álvaro Viedma-Poyatos
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Juan Zapata-Muñoz
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Beatriz Villarejo-Zori
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Raquel Gómez-Sintes
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Patricia Boya
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain; Departament of Neuroscience and Movement Science, Faculty of Science and Medicine, University of Fribourg, 1700, Fribourg, Switzerland.
| |
Collapse
|
4
|
Washington J, Ritch R, Liu Y. Homocysteine and Glaucoma. Int J Mol Sci 2023; 24:10790. [PMID: 37445966 DOI: 10.3390/ijms241310790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/10/2023] [Accepted: 06/11/2023] [Indexed: 07/15/2023] Open
Abstract
Elevated levels of homocysteine (Hcy), a non-proteinogenic amino acid, may lead to a host of manifestations across the biological systems, particularly the nervous system. Defects in Hcy metabolism have been associated with many neurodegenerative diseases including glaucoma, i.e., the leading cause of blindness. However, the pathophysiology of elevated Hcy and its eligibility as a risk factor for glaucoma remain unclear. We aimed to provide a comprehensive review of the relationship between elevated Hcy levels and glaucoma. Through a systemic search of the PubMed and Google Scholar databases, we found that elevated Hcy might play an important role in the pathogenesis of glaucoma. Further research will be necessary to help clarify the specific contribution of elevated Hcy in the pathogenesis of glaucoma. A discovery and conceptual understanding of Hcy-associated glaucoma could be the keys to providing better therapeutic treatment, if not prophylactic treatment, for this disease.
Collapse
Affiliation(s)
- Joshua Washington
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Robert Ritch
- New York Eye & Ear Infirmary, New York, NY 10003, USA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- James & Jean Culver Vision Discovery Institute, 4 Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
5
|
Sompol P, Gollihue JL, Weiss BE, Lin RL, Case SL, Kraner SD, Weekman EM, Gant JC, Rogers CB, Niedowicz DM, Sudduth TL, Powell DK, Lin AL, Nelson PT, Thibault O, Wilcock DM, Norris CM. Targeting Astrocyte Signaling Alleviates Cerebrovascular and Synaptic Function Deficits in a Diet-Based Mouse Model of Small Cerebral Vessel Disease. J Neurosci 2023; 43:1797-1813. [PMID: 36746627 PMCID: PMC10010459 DOI: 10.1523/jneurosci.1333-22.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Despite the indispensable role that astrocytes play in the neurovascular unit, few studies have investigated the functional impact of astrocyte signaling in cognitive decline and dementia related to vascular pathology. Diet-mediated induction of hyperhomocysteinemia (HHcy) recapitulates numerous features of vascular contributions to cognitive impairment and dementia (VCID). Here, we used astrocyte targeting approaches to evaluate astrocyte Ca2+ dysregulation and the impact of aberrant astrocyte signaling on cerebrovascular dysfunction and synapse impairment in male and female HHcy diet mice. Two-photon imaging conducted in fully awake mice revealed activity-dependent Ca2+ dysregulation in barrel cortex astrocytes under HHcy. Stimulation of contralateral whiskers elicited larger Ca2+ transients in individual astrocytes of HHcy diet mice compared with control diet mice. However, evoked Ca2+ signaling across astrocyte networks was impaired in HHcy mice. HHcy also was associated with increased activation of the Ca2+/calcineurin-dependent transcription factor NFAT4, which has been linked previously to the reactive astrocyte phenotype and synapse dysfunction in amyloid and brain injury models. Targeting the NFAT inhibitor VIVIT to astrocytes, using adeno-associated virus vectors, led to reduced GFAP promoter activity in HHcy diet mice and improved functional hyperemia in arterioles and capillaries. VIVIT expression in astrocytes also preserved CA1 synaptic function and improved spontaneous alternation performance on the Y maze. Together, the results demonstrate that aberrant astrocyte signaling can impair the major functional properties of the neurovascular unit (i.e., cerebral vessel regulation and synaptic regulation) and may therefore represent a promising drug target for treating VCID and possibly Alzheimer's disease and other related dementias.SIGNIFICANCE STATEMENT The impact of reactive astrocytes in Alzheimer's disease and related dementias is poorly understood. Here, we evaluated Ca2+ responses and signaling in barrel cortex astrocytes of mice fed with a B-vitamin deficient diet that induces hyperhomocysteinemia (HHcy), cerebral vessel disease, and cognitive decline. Multiphoton imaging in awake mice with HHcy revealed augmented Ca2+ responses in individual astrocytes, but impaired signaling across astrocyte networks. Stimulation-evoked arteriole dilation and elevated red blood cell velocity in capillaries were also impaired in cortex of awake HHcy mice. Astrocyte-specific inhibition of the Ca2+-dependent transcription factor, NFAT, normalized cerebrovascular function in HHcy mice, improved synaptic properties in brain slices, and stabilized cognition. Results suggest that astrocytes are a mechanism and possible therapeutic target for vascular-related dementia.
Collapse
Affiliation(s)
- Pradoldej Sompol
- Sanders-Brown Center on Aging
- Departments of Pharmacology and Nutritional Sciences
| | | | - Blaine E Weiss
- Sanders-Brown Center on Aging
- Departments of Pharmacology and Nutritional Sciences
| | - Ruei-Lung Lin
- Departments of Pharmacology and Nutritional Sciences
| | - Sami L Case
- Departments of Pharmacology and Nutritional Sciences
| | | | | | - John C Gant
- Sanders-Brown Center on Aging
- Departments of Pharmacology and Nutritional Sciences
| | | | | | | | | | - Ai-Ling Lin
- Sanders-Brown Center on Aging
- Departments of Pharmacology and Nutritional Sciences
| | - Peter T Nelson
- Sanders-Brown Center on Aging
- Pathology, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Olivier Thibault
- Sanders-Brown Center on Aging
- Departments of Pharmacology and Nutritional Sciences
| | | | - Christopher M Norris
- Sanders-Brown Center on Aging
- Departments of Pharmacology and Nutritional Sciences
| |
Collapse
|
6
|
Samra YA, Zaidi Y, Rajpurohit P, Raghavan R, Cai L, Kaddour-Djebbar I, Tawfik A. Warburg Effect as a Novel Mechanism for Homocysteine-Induced Features of Age-Related Macular Degeneration. Int J Mol Sci 2023; 24:ijms24021071. [PMID: 36674587 PMCID: PMC9865636 DOI: 10.3390/ijms24021071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/09/2023] Open
Abstract
Age-related macular degeneration (AMD) is a major cause of blindness. Recent studies have reported impaired glycolysis in AMD patients with a high lactate/pyruvate ratio. Elevated homocysteine (Hcy) (Hyperhomocysteinemia, HHcy) was observed in several clinical studies, reporting an association between HHcy and AMD. We established the effect of HHcy on barrier function, retinal pigment epithelium (RPE) structure, and induced choroidal neovascularization (CNV) in mice. We hypothesize that HHcy contributes to AMD by inducing a metabolic switch in the mitochondria, in which cells predominantly produce energy by the high rate of glycolysis, or "Warburg", effect. Increased glycolysis results in an increased production of lactate, cellular acidity, activation of angiogenesis, RPE barrier dysfunction, and CNV. Evaluation of cellular energy production under HHcy was assessed by seahorse analysis, immunofluorescence, and western blot experiments. The seahorse analysis evaluated the extracellular acidification rate (ECAR) as indicative of glycolysis. HHcy showed a significant increase in ECAR both in vivo using (Cystathionine β-synthase) cbs+/- and cbs-/- mice retinas and in vitro (Hcy-treated ARPE-19) compared to wild-type mice and RPE cells. Moreover, HHcy up-regulated glycolytic enzyme (Glucose transporter-1 (GlUT-1), lactate dehydrogenase (LDH), and hexokinase 1 (HK1)) in Hcy-treated ARPE-19 and primary RPE cells isolated from cbs+/+, cbs+/-, and cbs-/- mice retinas. Inhibition of GLUT-1 or blocking of N-methyl-D-aspartate receptors (NMDAR) reduced glycolysis in Hcy-treated RPE and improved albumin leakage and CNV induction in Hcy-injected mice eyes. The current study suggests that HHcy causes a metabolic switch in the RPE cells from mitochondrial respiration to glycolysis during AMD and confirms the involvement of NMDAR in this process. Therefore, targeting Glycolysis or NMDAR could be a novel therapeutic target for AMD.
Collapse
Affiliation(s)
- Yara A. Samra
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yusra Zaidi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Pragya Rajpurohit
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Raju Raghavan
- Department of Pharmacology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Lun Cai
- Department of Pharmacology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Ismail Kaddour-Djebbar
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA
| | - Amany Tawfik
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
- Eye Research Institute, Oakland University, Rochester, MI 48309-4479, USA
- Eye Research Center (OUWB)/ERC, William Beaumont School of Medicine, Royal Oak, MI 48309-4479, USA
- Correspondence: ; Tel.: +1-248-370-2398; Fax: +1-248-370-4211
| |
Collapse
|
7
|
Zhang H, Shen H, Gong W, Sun X, Jiang X, Wang J, Jin L, Xu X, Luo D, Wang X. Plasma homocysteine and macular thickness in older adults-the Rugao Longevity and Aging Study. Eye (Lond) 2022; 36:1050-1060. [PMID: 33976397 PMCID: PMC9046221 DOI: 10.1038/s41433-021-01549-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/08/2021] [Accepted: 04/09/2021] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES To determine the association of plasma homocysteine levels with retinal layer thickness in a large community cohort of older adults. METHODS The Rugao Longevity and Ageing Study is an observational, prospective and community-based cohort study. A total of 989 older adults who underwent spectral-domain optical coherence tomography (SD-OCT) were included and analyzed. Foveal, macular retinal nerve fibre layer (mRNFL) and ganglion cell layer plus inner plexiform layer (GC-IPL) thicknesses were measured by SD-OCT. Plasma homocysteine levels were measured using chemiluminescence immunoassay. Linear regression analyses were performed to evaluate the relationship between plasma homocysteine and retinal layer thickness while controlling for confounding factors. RESULTS Of the 989 participants, 500 (50.56%) were men. The mean age was 78.26 (4.58) years, and the mean plasma homocysteine level was 16.38 (8.05) μmol/L. In multivariable analyses, each unit increase in plasma homocysteine was associated with an 8.84 × 10-2 (95% CI: -16.54 × 10-2 to -1.15 × 10-2, p = 0.032) μm decrease in the average inner thickness of the GC-IPL after controlling for confounding factors. The association remained significant even in participants without major cardiovascular disease or diabetes (β = -10.33 × 10-2, 95% CI: -18.49 × 10-2 to -2.18 × 10-2, p = 0.013). No significant associations of plasma homocysteine levels with macular thickness or mRNFL were found in primary and sensitivity analyses (p > 0.05). CONCLUSIONS Increased plasma homocysteine levels are associated with a thinner GC-IPL. Plasma homocysteine may be a risk factor for thinner retinas in older adults.
Collapse
Affiliation(s)
- Hui Zhang
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443Human Phenome Institute, Fudan University, Shanghai, China
| | - Hangqi Shen
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.412478.c0000 0004 1760 4628Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China ,Shanghai Engineering Center for Visual Science and Photomedicin, Shanghai, China ,grid.412478.c0000 0004 1760 4628Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China ,grid.412478.c0000 0004 1760 4628National Clinical Research Center for Eye Diseases, Shanghai, China
| | - Wei Gong
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.412478.c0000 0004 1760 4628Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China ,Shanghai Engineering Center for Visual Science and Photomedicin, Shanghai, China ,grid.412478.c0000 0004 1760 4628Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China ,grid.412478.c0000 0004 1760 4628National Clinical Research Center for Eye Diseases, Shanghai, China
| | - Xuehui Sun
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443Human Phenome Institute, Fudan University, Shanghai, China
| | - Xiaoyan Jiang
- grid.24516.340000000123704535Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
| | - Jiucun Wang
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443Human Phenome Institute, Fudan University, Shanghai, China
| | - Li Jin
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443Human Phenome Institute, Fudan University, Shanghai, China
| | - Xun Xu
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.412478.c0000 0004 1760 4628Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China ,Shanghai Engineering Center for Visual Science and Photomedicin, Shanghai, China ,grid.412478.c0000 0004 1760 4628Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China ,grid.412478.c0000 0004 1760 4628National Clinical Research Center for Eye Diseases, Shanghai, China
| | - Dawei Luo
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.412478.c0000 0004 1760 4628Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China ,Shanghai Engineering Center for Visual Science and Photomedicin, Shanghai, China ,grid.412478.c0000 0004 1760 4628Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China ,grid.412478.c0000 0004 1760 4628National Clinical Research Center for Eye Diseases, Shanghai, China
| | - Xiaofeng Wang
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443Human Phenome Institute, Fudan University, Shanghai, China ,grid.413597.d0000 0004 1757 8802Shanghai Key Laboratory of Clinical Geriatric Medicine and Huadong Hospital Clinical Research Center for Geriatric Medicine, Shanghai, China ,grid.8547.e0000 0001 0125 2443National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Mitochondrial Fragmentation in a High Homocysteine Environment in Diabetic Retinopathy. Antioxidants (Basel) 2022; 11:antiox11020365. [PMID: 35204246 PMCID: PMC8868328 DOI: 10.3390/antiox11020365] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Diabetic patients routinely have elevated homocysteine levels, and due to increase in oxidative stress, hyperhomocysteinemia is associated with increased mitochondrial damage. Mitochondrial homeostasis is directly related to the balance between their fission and fusion, and in diabetes this balance is disturbed. The aim of this study was to investigate the role of homocysteine in mitochondrial fission in diabetic retinopathy. Human retinal endothelial cells, either untransfected or transfected with siRNA of a fission protein (dynamin-related protein 1, Drp1) and incubated in the presence of 100 μM homocysteine, were analyzed for mitochondrial fragmentation by live-cell microscopy and GTPase activity of Drp1. Protective nucleoids and mtDNA damage were evaluated by SYBR DNA stain and by transcripts of mtDNA-encoded ND6 and cytochrome b. The role of nitrosylation of Drp1 in homocysteine-mediated exacerbation of mitochondrial fragmentation was determined by supplementing incubation medium with nitric-oxide inhibitor. Homocysteine exacerbated glucose-induced Drp1 activation and its nitrosylation, mitochondrial fragmentation and cell apoptosis, and further decreased nucleoids and mtDNA transcription. Drp1-siRNA or nitric-oxide inhibitor prevented glucose- and homocysteine-induced mitochondrial fission, damage and cell apoptosis. Thus, elevated homocysteine in a hyperglycemic environment increases Drp1 activity via increasing its nitrosylation, and this further fragments the mitochondria and increases apoptosis, ultimately leading to the development of diabetic retinopathy.
Collapse
|
9
|
Effect of long-term chronic hyperhomocysteinemia on retinal structure and function in the cystathionine-β-synthase mutant mouse. Exp Eye Res 2022; 214:108894. [PMID: 34906600 PMCID: PMC9251730 DOI: 10.1016/j.exer.2021.108894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/03/2023]
Abstract
Elevated levels of the excitatory amino acid homocysteine (Hcy) have been implicated in retinal diseases in humans including glaucoma and macular degeneration. It is not clear whether elevated Hcy levels are pathogenic. Models of hyperhomocysteinemia (Hhcy) have proven useful in addressing this including mice with deficiency in the enzyme cystathionine β-synthase (CBS). Cbs+/- mice have a ∼two-fold increase in plasma and retinal Hcy levels. Previous studies of visual function and structure in Cbs+/- mice during the first 10 months of life revealed mild ganglion cell loss, but minimal electrophysiological alterations. It is not clear whether extended, chronic exposure to moderate Hhcy elevation will lead to visual function loss and retinal pathology. The present study addressed this by performing comprehensive analyses of retinal function/structure in 20 month Cbs+/- and Cbs+/+ (WT) mice including IOP, SD-OCT, scotopic and photopic ERG, pattern ERG (pERG), and visual acuity. Eyes were harvested for histology and immunohistochemical analysis of Brn3a (ganglion cells), dihydroethidium (oxidative stress) and GFAP (gliosis). The analyses revealed no difference in IOP between groups for age/strain. Visual acuity measured ∼0.36c/d for mice at 20 months in Cbs+/- and WT mice; contrast sensitivity did not differ between groups at either age. Similarly SD-OCT, scotopic/photopic ERG and pERG revealed no differences between 20 month Cbs+/- and WT mice. There was minimal disruption in retinal structure when eyes were examined histologically. Morphometric analysis revealed no significant differences in retinal layers. Immunohistochemistry revealed ∼5 RGCs/100 μm retinal length in both Cbs+/- and WT mice at 20 months. While there was greater oxidative stress and gliosis in older (20 month) mice versus young (4 month) mice, there was no difference in these parameters between the 20 month Cbs+/- and WT mice. We conclude that chronic, moderate Hhcy (at least due to deficiency of Cbs) is not accompanied by retinal structural/functional changes that differ significantly from age-matched WT littermates. Despite considerable evidence that severe Hhcy is toxic to retina, moderate Hhcy appears tolerated by retina suggesting compensatory cellular survival mechanisms.
Collapse
|
10
|
Implication of N-Methyl-d-Aspartate Receptor in Homocysteine-Induced Age-Related Macular Degeneration. Int J Mol Sci 2021; 22:ijms22179356. [PMID: 34502266 PMCID: PMC8431693 DOI: 10.3390/ijms22179356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/12/2021] [Accepted: 08/26/2021] [Indexed: 02/03/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss. Elevated homocysteine (Hcy) (Hyperhomocysteinemia) (HHcy) has been reported in AMD. We previously reported that HHcy induces AMD-like features. This study suggests that N-Methyl-d-aspartate receptor (NMDAR) activation in the retinal pigment epithelium (RPE) is a mechanism for HHcy-induced AMD. Serum Hcy and cystathionine-β-synthase (CBS) were assessed by ELISA. The involvement of NMDAR in Hcy-induced AMD features was evaluated (1) in vitro using ARPE-19 cells, primary RPE isolated from HHcy mice (CBS), and mouse choroidal endothelial cells (MCEC); (2) in vivo using wild-type mice and mice deficient in RPE NMDAR (NMDARR−/−) with/without Hcy injection. Isolectin-B4, Ki67, HIF-1α, VEGF, NMDAR1, and albumin were assessed by immunofluorescence (IF), Western blot (WB), Optical coherence tomography (OCT), and fluorescein angiography (FA) to evaluate retinal structure, fluorescein leakage, and choroidal neovascularization (CNV). A neovascular AMD patient’s serum showed a significant increase in Hcy and a decrease in CBS. Hcy significantly increased HIF-1α, VEGF, and NMDAR in RPE cells, and Ki67 in MCEC. Hcy-injected WT mice showed disrupted retina and CNV. Knocking down RPE NMDAR improved retinal structure and CNV. Our findings underscore the role of RPE NMDAR in Hcy-induced AMD features; thus, NMDAR inhibition could serve as a promising therapeutic target for AMD.
Collapse
|
11
|
Icel E, Ucak T. The effects of vitamin B12 deficiency on retina and optic disk vascular density. Int Ophthalmol 2021; 41:3145-3151. [PMID: 34047909 DOI: 10.1007/s10792-021-01879-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE In this study, we aimed to determine the alterations in the retina and peripapillary vascular density in patients with vitamin B12 deficiency. MATERIAL AND METHOD The patients were divided into two groups regarding their vitamin B12 levels as the low vitamin B12 group (< 200 pg/ml) and the normal vitamin B12 group (≥ 200 pg/ml). The retinal nerve fiber layer thickness (RNFLT) and the central macular thickness (CMT) were measured through an SD-OCT analysis. The foveal avascular zone (FAZ), vessel density (VD) of the superficial capillary plexus (SCP), and deep capillary plexus (DCP) of the macula, and the VD of the radial peripapillary capillary plexus (RPCP) for the optic disk were determined by OCT-A. RESULTS Thirty-three patients were in the low vitamin B12 group and 54 were in the normal group. The mean RNFLT measurements were significantly lower in the low vitamin B12 group (p = 0.001). The RPCP values in all quadrants were significantly lower in the patients with vitamin B12 deficiency (p = 0.001); moreover, there was a significant increase in the FAZ value with a decrease in both superficial (p = 0.001) and deep (p = 0.001) VDs. There was a significant positive correlation between the serum vitamin B12 levels and the RPCP values and superficial and deep VDs, while the FAZ value was negatively correlated with the vitamin B12 levels. CONCLUSION Vitamin B12 deficiency has diverse effects on ocular structures and retinal vasculature. Decreased VDs and increased FAZ may be associated with severe ocular alterations in the long term, which should be further investigated.
Collapse
Affiliation(s)
- Erel Icel
- Department of Ophthalmology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Turgay Ucak
- Department of Ophthalmology, University of Health Sciences Turkey, Sisli Hamidiye Etfal Training and Research Hospital, İstanbul, Turkey.
| |
Collapse
|
12
|
Kaplan P, Tatarkova Z, Sivonova MK, Racay P, Lehotsky J. Homocysteine and Mitochondria in Cardiovascular and Cerebrovascular Systems. Int J Mol Sci 2020; 21:ijms21207698. [PMID: 33080955 PMCID: PMC7589705 DOI: 10.3390/ijms21207698] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022] Open
Abstract
Elevated concentration of homocysteine (Hcy) in the blood plasma, hyperhomocysteinemia (HHcy), has been implicated in various disorders, including cardiovascular and neurodegenerative diseases. Accumulating evidence indicates that pathophysiology of these diseases is linked with mitochondrial dysfunction. In this review, we discuss the current knowledge concerning the effects of HHcy on mitochondrial homeostasis, including energy metabolism, mitochondrial apoptotic pathway, and mitochondrial dynamics. The recent studies suggest that the interaction between Hcy and mitochondria is complex, and reactive oxygen species (ROS) are possible mediators of Hcy effects. We focus on mechanisms contributing to HHcy-associated oxidative stress, such as sources of ROS generation and alterations in antioxidant defense resulting from altered gene expression and post-translational modifications of proteins. Moreover, we discuss some recent findings suggesting that HHcy may have beneficial effects on mitochondrial ROS homeostasis and antioxidant defense. A better understanding of complex mechanisms through which Hcy affects mitochondrial functions could contribute to the development of more specific therapeutic strategies targeted at HHcy-associated disorders.
Collapse
|
13
|
Diabetic Retinopathy: Mitochondria Caught in a Muddle of Homocysteine. J Clin Med 2020; 9:jcm9093019. [PMID: 32961662 PMCID: PMC7564979 DOI: 10.3390/jcm9093019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/09/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022] Open
Abstract
Diabetic retinopathy is one of the most feared complications of diabetes. In addition to the severity of hyperglycemia, systemic factors also play an important role in its development. Another risk factor in the development of diabetic retinopathy is elevated levels of homocysteine, a non-protein amino acid, and hyperglycemia and homocysteine are shown to produce synergistic detrimental effects on the vasculature. Hyperhomocysteinemia is associated with increased oxidative stress, and in the pathogenesis of diabetic retinopathy, oxidative stress-mitochondrial dysfunction precedes the development of histopathology characteristic of diabetic retinopathy. Furthermore, homocysteine biosynthesis from methionine forms S-adenosyl methionine (SAM), and SAM is a co-substrate of DNA methylation. In diabetes, DNA methylation machinery is activated, and mitochondrial DNA (mtDNA) and several genes associated with mitochondrial homeostasis undergo epigenetic modifications. Consequently, high homocysteine, by further affecting methylation of mtDNA and that of genes associated with mtDNA damage and biogenesis, does not give any break to the already damaged mitochondria, and the vicious cycle of free radicals continues. Thus, supplementation of sensible glycemic control with therapies targeting hyperhomocysteinemia could be valuable for diabetic patients to prevent/slow down the development of this sight-threatening disease.
Collapse
|
14
|
De Miguel C, Hamrick WC, Sedaka R, Jagarlamudi S, Asico LD, Jose PA, Cuevas S. Uncoupling Protein 2 Increases Blood Pressure in DJ -1 Knockout Mice. J Am Heart Assoc 2020; 8:e011856. [PMID: 30995881 PMCID: PMC6512091 DOI: 10.1161/jaha.118.011856] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background The redox-sensitive chaperone DJ -1 and uncoupling protein 2 are protective against mitochondrial oxidative stress. We previously reported that renal-selective depletion and germline deletion of DJ -1 increases blood pressure in mice. This study aimed to determine the mechanisms involved in the oxidative stress-mediated hypertension in DJ -1 -/- mice. Methods and Results There were no differences in sodium excretion, renal renin expression, renal NADPH oxidase activity, and serum creatinine levels between DJ -1 -/- and wild-type mice. Renal expression of nitro-tyrosine, malondialdehyde, and urinary kidney injury marker-1 were increased in DJ -1 -/- mice relative to wild-type littermates. mRNA expression of mitochondrial heat shock protein 60 was also elevated in kidneys from DJ -1 -/- mice, indicating the presence of oxidative stress. Tempol-treated DJ -1 -/- mice presented higher serum nitrite/nitrate levels than vehicle-treated DJ -1 -/- mice, suggesting a role of the NO system in the high blood pressure of this model. Tempol treatment normalized renal kidney injury marker-1 and malondialdehyde expression as well as blood pressure in DJ -1 -/- mice, but had no effect in wild-type mice. The renal Ucp2 mRNA expression was increased in DJ -1 -/- mice versus wild-type and was also normalized by tempol. The renal-selective silencing of Ucp2 led to normalization of blood pressure and serum nitrite/nitrate ratio in DJ -1 -/- mice. Conclusions The deletion of DJ -1 leads to oxidative stress-induced hypertension associated with downregulation of NO function, and overexpression of Ucp2 in the kidney increases blood pressure in DJ -1 -/- mice. To our knowledge, this is the first report providing evidence of the role of uncoupling protein 2 in blood pressure regulation.
Collapse
Affiliation(s)
- Carmen De Miguel
- 1 Section of Cardio-Renal Physiology and Medicine Division of Nephrology Department of Medicine University of Alabama at Birmingham AL
| | - William C Hamrick
- 1 Section of Cardio-Renal Physiology and Medicine Division of Nephrology Department of Medicine University of Alabama at Birmingham AL
| | - Randee Sedaka
- 1 Section of Cardio-Renal Physiology and Medicine Division of Nephrology Department of Medicine University of Alabama at Birmingham AL
| | - Sudha Jagarlamudi
- 2 Division of Renal Diseases & Hypertension Department of Medicine The George Washington University School of Medicine and Health Sciences Washington DC
| | - Laureano D Asico
- 2 Division of Renal Diseases & Hypertension Department of Medicine The George Washington University School of Medicine and Health Sciences Washington DC
| | - Pedro A Jose
- 2 Division of Renal Diseases & Hypertension Department of Medicine The George Washington University School of Medicine and Health Sciences Washington DC
| | - Santiago Cuevas
- 3 Research Center for Genetic Medicine Children's National Health System Washington DC
| |
Collapse
|
15
|
Lee H, Smith SB, Sheu SS, Yoon Y. The short variant of optic atrophy 1 (OPA1) improves cell survival under oxidative stress. J Biol Chem 2020; 295:6543-6560. [PMID: 32245890 DOI: 10.1074/jbc.ra119.010983] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 03/31/2020] [Indexed: 01/23/2023] Open
Abstract
Optic atrophy 1 (OPA1) is a dynamin protein that mediates mitochondrial fusion at the inner membrane. OPA1 is also necessary for maintaining the cristae and thus essential for supporting cellular energetics. OPA1 exists as membrane-anchored long form (L-OPA1) and short form (S-OPA1) that lacks the transmembrane region and is generated by cleavage of L-OPA1. Mitochondrial dysfunction and cellular stresses activate the inner membrane-associated zinc metallopeptidase OMA1 that cleaves L-OPA1, causing S-OPA1 accumulation. The prevailing notion has been that L-OPA1 is the functional form, whereas S-OPA1 is an inactive cleavage product in mammals, and that stress-induced OPA1 cleavage causes mitochondrial fragmentation and sensitizes cells to death. However, S-OPA1 contains all functional domains of dynamin proteins, suggesting that it has a physiological role. Indeed, we recently demonstrated that S-OPA1 can maintain cristae and energetics through its GTPase activity, despite lacking fusion activity. Here, applying oxidant insult that induces OPA1 cleavage, we show that cells unable to generate S-OPA1 are more sensitive to this stress under obligatory respiratory conditions, leading to necrotic death. These findings indicate that L-OPA1 and S-OPA1 differ in maintaining mitochondrial function. Mechanistically, we found that cells that exclusively express L-OPA1 generate more superoxide and are more sensitive to Ca2+-induced mitochondrial permeability transition, suggesting that S-OPA1, and not L-OPA1, protects against cellular stress. Importantly, silencing of OMA1 expression increased oxidant-induced cell death, indicating that stress-induced OPA1 cleavage supports cell survival. Our findings suggest that S-OPA1 generation by OPA1 cleavage is a survival mechanism in stressed cells.
Collapse
Affiliation(s)
- Hakjoo Lee
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia 30912
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia 30912.,Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia 30912
| | - Shey-Shing Sheu
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia 30912
| |
Collapse
|
16
|
Behrouzi P, Grootswagers P, Keizer PLC, Smeets ETHC, Feskens EJM, de Groot LCPGM, van Eeuwijk FA. Dietary Intakes of Vegetable Protein, Folate, and Vitamins B-6 and B-12 Are Partially Correlated with Physical Functioning of Dutch Older Adults Using Copula Graphical Models. J Nutr 2020; 150:634-643. [PMID: 31858107 PMCID: PMC7056616 DOI: 10.1093/jn/nxz269] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/14/2019] [Accepted: 10/09/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND In nutritional epidemiology, dealing with confounding and complex internutrient relations are major challenges. An often-used approach is dietary pattern analyses, such as principal component analysis, to deal with internutrient correlations, and to more closely resemble the true way nutrients are consumed. However, despite these improvements, these approaches still require subjective decisions in the preselection of food groups. Moreover, they do not make efficient use of multivariate dietary data, because they detect only marginal associations. We propose the use of copula graphical models (CGMs) to model and make statistical inferences regarding complex associations among variables in multivariate data, where associations between all variables can be learned simultaneously. OBJECTIVE We aimed to reconstruct nutritional intake and physical functioning networks in Dutch older adults by applying a CGM. METHODS We addressed this issue by uncovering the pairwise associations between variables while correcting for the effect of remaining variables. More specifically, we used a CGM to infer the precision matrix, which contains all the conditional independence relations between nodes in the graph. The nonzero elements of the precision matrix indicate the presence of a direct association. We applied this method to reconstruct nutrient-physical functioning networks from the combined data of 4 studies (Nu-Age, ProMuscle, ProMO, and V-Fit, total n = 662, mean ± SD age = 75 ± 7 y). The method was implemented in the R package nutriNetwork which is freely available at https://cran.r-project.org/web/packages/nutriNetwork. RESULTS Greater intakes of vegetable protein and vitamin B-6 were partially correlated with higher scores on the total Short Physical Performance Battery (SPPB) and the chair rise test. Greater intakes of vitamin B-12 and folate were partially correlated with higher scores on the chair rise test and the total SPPB, respectively. CONCLUSIONS We determined that vegetable protein, vitamin B-6, folate, and vitamin B-12 intakes are partially correlated with improved functional outcome measurements in Dutch older adults.
Collapse
Affiliation(s)
- Pariya Behrouzi
- Biometris, Mathematical and Statistical Methods, Wageningen University and Research, Wageningen, Netherlands
| | - Pol Grootswagers
- Department of Human Nutrition, Wageningen University and Research, Wageningen, Netherlands
| | - Paul L C Keizer
- Biometris, Mathematical and Statistical Methods, Wageningen University and Research, Wageningen, Netherlands
| | - Ellen T H C Smeets
- Department of Human Nutrition, Wageningen University and Research, Wageningen, Netherlands
| | - Edith J M Feskens
- Department of Human Nutrition, Wageningen University and Research, Wageningen, Netherlands
| | | | - Fred A van Eeuwijk
- Biometris, Mathematical and Statistical Methods, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
17
|
Kowluru RA, Mohammad G, Sahajpal N. Faulty homocysteine recycling in diabetic retinopathy. EYE AND VISION 2020; 7:4. [PMID: 31938715 PMCID: PMC6953140 DOI: 10.1186/s40662-019-0167-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022]
Abstract
Background Although hyperglycemia is the main instigator in the development of diabetic retinopathy, elevated circulating levels of a non-protein amino acid, homocysteine, are also associated with an increased risk of retinopathy. Homocysteine is recycled back to methionine by methylenetetrahydrofolate reductase (MTHFR) and/or transsulfurated by cystathionine β-synthase (CBS) to form cysteine. CBS and other transsulfuration enzyme cystathionine-γ-lyase (CSE), through desulfuration, generates H2S. Methionine cycle also regulates DNA methylation, an epigenetic modification associated with the gene suppression. The aim of this study was to investigate homocysteine and its metabolism in diabetic retinopathy. Methods Homocysteine and H2S levels were analyzed in the retina, and CBS, CSE and MTHFR in the retinal microvasculature from human donors with established diabetic retinopathy. Mitochondrial damage was evaluated in retinal microvessels by quantifying enzymes responsible for maintaining mitochondrial dynamics (fission-fusion-mitophagy). DNA methylation status of CBS and MTHFR promoters was examined using methylated DNA immunoprecipitation technique. The direct effect of homocysteine on mitochondrial damage was confirmed in human retinal endothelial cells (HRECs) incubated with 100 μM L-homocysteine. Results Compared to age-matched nondiabetic control human donors, retina from donors with established diabetic retinopathy had ~ 3-fold higher homocysteine levels and ~ 50% lower H2S levels. The enzymes important for both transsulfuration and remethylation of homocysteine including CBS, CSE and MTHFR, were 40–60% lower in the retinal microvasculature from diabetic retinopathy donors. While the mitochondrial fission protein, dynamin related protein 1, and mitophagy markers optineurin and microtubule-associated protein 1A/1B-light chain 3 (LC3), were upregulated, the fusion protein mitofusin 2 was downregulated. In the same retinal microvessel preparations from donors with diabetic retinopathy, DNA at the promoters of CBS and MTHFR were hypermethylated. Incubation of HRECs with homocysteine increased reactive oxygen species and decreased transcripts of mtDNA-encoded CYTB. Conclusions Compromised transsulfuration and remethylation processes play an important role in the poor removal of retinal homocysteine in diabetic patients. Thus, regulation of their homocysteine levels should ameliorate retinal mitochondrial damage, and by regulating DNA methylation status of the enzymes responsible for homocysteine transsulfuration and remethylation, should prevent excess accumulation of homocysteine.
Collapse
Affiliation(s)
- Renu A Kowluru
- Department of Ophthalmology, Visual Sciences and Anatomical Sciences, Wayne State University, 4717 St. Antoine, Detroit, MI 48201 USA
| | - Ghulam Mohammad
- Department of Ophthalmology, Visual Sciences and Anatomical Sciences, Wayne State University, 4717 St. Antoine, Detroit, MI 48201 USA
| | - Nikhil Sahajpal
- Department of Ophthalmology, Visual Sciences and Anatomical Sciences, Wayne State University, 4717 St. Antoine, Detroit, MI 48201 USA
| |
Collapse
|
18
|
Navneet S, Zhao J, Wang J, Mysona B, Barwick S, Ammal Kaidery N, Saul A, Kaddour-Djebbar I, Bollag WB, Thomas B, Bollinger KE, Smith SB. Hyperhomocysteinemia-induced death of retinal ganglion cells: The role of Müller glial cells and NRF2. Redox Biol 2019; 24:101199. [PMID: 31026769 PMCID: PMC6482349 DOI: 10.1016/j.redox.2019.101199] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/05/2019] [Accepted: 04/10/2019] [Indexed: 12/23/2022] Open
Abstract
Hyperhomocysteinemia (Hhcy), or increased levels of the excitatory amino acid homocysteine (Hcy), is implicated in glaucoma, a disease characterized by increased oxidative stress and loss of retinal ganglion cells (RGCs). Whether Hhcy is causative or merely a biomarker for RGC loss in glaucoma is unknown. Here we analyzed the role of NRF2, a master regulator of the antioxidant response, in Hhcy-induced RGC death in vivo and in vitro. By crossing Nrf2−/− mice and two mouse models of chronic Hhcy (Cbs+/- and Mthfr+/- mice), we generated Cbs+/-Nrf2−/− and Mthfr+/-Nrf2−/− mice and performed systematic analysis of retinal architecture and visual acuity followed by assessment of retinal morphometry and gliosis. We observed significant reduction of inner retinal layer thickness and reduced visual acuity in Hhcy mice lacking NRF2. These functional deficits were accompanied by fewer RGCs and increased gliosis. Given the key role of Müller glial cells in maintaining RGCs, we established an ex-vivo indirect co-culture system using primary RGCs and Müller cells. Hhcy-exposure decreased RGC viability, which was abrogated when cells were indirectly cultured with wildtype (WT) Müller cells, but not with Nrf2−/− Müller cells. Exposure of WT Müller cells to Hhcy yielded a robust mitochondrial and glycolytic response, which was not observed in Nrf2−/− Müller cells. Taken together, the in vivo and in vitro data suggest that deleterious effects of Hhcy on RGCs are likely dependent upon the health of retinal glial cells and the availability of an intact retinal antioxidant response mechanism. Oxidative stress is linked to homocysteine (Hcy)-induced retinal ganglion cell death. NRF2's role in protecting ganglion cells from excess Hcy was studied in vitro/vivo. Hyper-Hcy mice were crossed with Nrf2−/− mice to study retinal function/structure. Ganglion cells co-cultured with primary WT Müller glial cells survived Hcy treatment. Nrf2−/− Müller cells did not afford neuroprotective advantage to Hcy-treated cells.
Collapse
Affiliation(s)
- Soumya Navneet
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Jing Zhao
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Barbara Mysona
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Shannon Barwick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Navneet Ammal Kaidery
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Alan Saul
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ismail Kaddour-Djebbar
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA
| | - Wendy B Bollag
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA
| | - Bobby Thomas
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Department of Drug Discovery, Medical University of South Carolina, Charleston, SC, USA
| | - Kathryn E Bollinger
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
19
|
Prendecki M, Florczak-Wyspianska J, Kowalska M, Ilkowski J, Grzelak T, Bialas K, Wiszniewska M, Kozubski W, Dorszewska J. Biothiols and oxidative stress markers and polymorphisms of TOMM40 and APOC1 genes in Alzheimer's disease patients. Oncotarget 2018; 9:35207-35225. [PMID: 30443289 PMCID: PMC6219666 DOI: 10.18632/oncotarget.26184] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/01/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive disease, with frequently observed improper biothiols turnover, homocysteine (Hcy) and glutathione (GSH). GSH protects cells from oxidative stress and may be determined by 8-oxo-2’-deoxyguanosine (8-oxo2dG) level and its repair enzyme 8-oxoguanine DNA glycosylase (OGG1). The presence of unfavorable alleles, e.g., in APOE cluster, TOMM40 or APOC1 is known to facilitate the dementia onset under oxidative stress. The aim of the study was to analyze rs1052452, rs2075650 TOMM40 polymorphisms, rs4420638 APOC1, and their correlation with Hcy, GSH, 8-oxo2dG, OGG1 levels in plasma of AD patients and controls. We recruited 230 individuals: 88 AD, 80 controls without (UC), 62 controls with (RC) positive family history of AD. The TOMM40 genotype was determined by HRM and capillary electrophoresis, while APOC1 by HRM. The concentrations of OGG1, 8-oxo2dG were determined by ELISA, whereas Hcy, GSH by HPLC/EC. We showed that over 60% of AD patients had increased Hcy levels (p<0.01 vs. UC, p<0.001 vs. RC), while GSH (p<0.01 vs. UC), 8-oxo2dG (p<0.01 vs. UC, p<0.001 vs. RC) were reduced. Minor variants: rs10524523-L, rs4420638-G, rs2075650-G were significantly overrepresented in AD. For rs4420638-G, rs2075650-G variants, the association remained significant in APOE E4 non-carriers. The misbalance of analyzed biothiols, and 8-oxo2dG, OGG1 were more pronounced in carriers of major variants: rs10524523-S/VL, rs4420638-A, rs2075650-A. We showed, for the first time, that APOC1 and TOMM40 rs2075650 polymorphisms may be independent risk factors of developing AD, whose major variants are accompanied by disruption of biothiols metabolism and inefficient removal of DNA oxidation.
Collapse
Affiliation(s)
- Michal Prendecki
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Marta Kowalska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jan Ilkowski
- Department of Emergency Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Teresa Grzelak
- Division of Biology of Civilization-Linked Diseases, Department of Chemistry and Clinical Biochemistry, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Bialas
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Malgorzata Wiszniewska
- Faculty of Health Care, Stanislaw Staszic University of Applied Sciences in Pila, Pila, Poland.,Department of Neurology, Specialistic Hospital in Pila, Pila, Poland
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
20
|
Sheng W, Lu Y, Mei F, Wang N, Liu ZZ, Han YY, Wang HT, Zou S, Xu H, Zhang X. Effect of Resveratrol on Sirtuins, OPA1, and Fis1 Expression in Adult Zebrafish Retina. ACTA ACUST UNITED AC 2018; 59:4542-4551. [PMID: 30208422 DOI: 10.1167/iovs.18-24539] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Weiwei Sheng
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
- Queen Mary School of Nanchang University, Nanchang, China
| | - Ye Lu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
| | - Feng Mei
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
| | - Ning Wang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
| | - Zhi-Zhi Liu
- Institute of Life Science of Nanchang University, Nanchang, China
- School of Life Sciences of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Ying-Ying Han
- Institute of Life Science of Nanchang University, Nanchang, China
- School of Life Sciences of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Han-Tsing Wang
- Institute of Life Science of Nanchang University, Nanchang, China
- School of Life Sciences of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Suqi Zou
- Institute of Life Science of Nanchang University, Nanchang, China
- School of Life Sciences of Nanchang University, Nanchang, China
| | - Hong Xu
- Institute of Life Science of Nanchang University, Nanchang, China
- School of Life Sciences of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| |
Collapse
|
21
|
Cueto R, Zhang L, Shan HM, Huang X, Li X, Li YF, Lopez J, Yang WY, Lavallee M, Yu C, Ji Y, Yang X, Wang H. Identification of homocysteine-suppressive mitochondrial ETC complex genes and tissue expression profile - Novel hypothesis establishment. Redox Biol 2018; 17:70-88. [PMID: 29679893 PMCID: PMC6006524 DOI: 10.1016/j.redox.2018.03.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 03/22/2018] [Indexed: 12/13/2022] Open
Abstract
Hyperhomocysteinemia (HHcy) is an independent risk factor for cardiovascular disease (CVD) which has been implicated in matochondrial (Mt) function impairment. In this study, we characterized Hcy metabolism in mouse tissues by using LC-ESI-MS/MS analysis, established tissue expression profiles for 84 nuclear-encoded Mt electron transport chain complex (nMt-ETC-Com) genes in 20 human and 19 mouse tissues by database mining, and modeled the effect of HHcy on Mt-ETC function. Hcy levels were high in mouse kidney/lung/spleen/liver (24-14 nmol/g tissue) but low in brain/heart (~5 nmol/g). S-adenosylhomocysteine (SAH) levels were high in the liver/kidney (59-33 nmol/g), moderate in lung/heart/brain (7-4 nmol/g) and low in spleen (1 nmol/g). S-adenosylmethionine (SAM) was comparable in all tissues (42-18 nmol/g). SAM/SAH ratio was as high as 25.6 in the spleen but much lower in the heart/lung/brain/kidney/liver (7-0.6). The nMt-ETC-Com genes were highly expressed in muscle/pituitary gland/heart/BM in humans and in lymph node/heart/pancreas/brain in mice. We identified 15 Hcy-suppressive nMt-ETC-Com genes whose mRNA levels were negatively correlated with tissue Hcy levels, including 11 complex-I, one complex-IV and two complex-V genes. Among the 11 Hcy-suppressive complex-I genes, 4 are complex-I core subunits. Based on the pattern of tissue expression of these genes, we classified tissues into three tiers (high/mid/low-Hcy responsive), and defined heart/eye/pancreas/brain/kidney/liver/testis/embryonic tissues as tier 1 (high-Hcy responsive) tissues in both human and mice. Furthermore, through extensive literature mining, we found that most of the Hcy-suppressive nMt-ETC-Com genes were suppressed in HHcy conditions and related with Mt complex assembly/activity impairment in human disease and experimental models. We hypothesize that HHcy inhibits Mt complex I gene expression leading to Mt dysfunction.
Collapse
Affiliation(s)
- Ramon Cueto
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Lixiao Zhang
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Hui Min Shan
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Xiao Huang
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Xinyuan Li
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Ya-Feng Li
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Jahaira Lopez
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - William Y Yang
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Muriel Lavallee
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Catherine Yu
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA; The Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | - Yong Ji
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China.
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA; Department of Pharmacology, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA; Thrombosis Research Center, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA; Cardiovascular Research Center, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA; Department of Pharmacology, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA; Thrombosis Research Center, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA; Cardiovascular Research Center, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Ablation of Toll-like receptor 4 mitigates central blood pressure response during hyperhomocysteinemia. J Hypertens 2018; 35:2226-2237. [PMID: 28665886 DOI: 10.1097/hjh.0000000000001460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The objective of this study was to define the mechanisms of homocysteine-induced effects on the aortic wall that promote vascular remodeling and hypertension as well as explore the role of Toll-like receptor 4 in homocysteine-induced effects. METHOD Five strains of mice were utilized in this study: C57BL/6J, C3H/HeOuJ, CBS+/-, C3H/HeJ and CBS+/-/C3H. Aorta, heart and blood were collected at the end of the experiments. Blood pressure (BP) was recorded using noninvasive tail cuff method. To determinate effects of vasoactive agent and endothelial-dependent vasodilator on aorta contractility, we performed vascular function measurements. In addition, the expression of mitochondrial fusion and fission proteins, antioxidant markers and collagen fragments were assessed. RESULTS BP measurements demonstrated a significant increase in SBP and DBPs in CBS+/- mice compared with other groups. CBS+/- mice aorta had lower response to phenylephrine and acetylcholine compared with other groups; however, CBS+/-/C3H mice response was improved. Dynamin-related protein 1 protein expression was significantly upregulated in CBS+/- mice, whereas C3H mice showed downregulation. In addition, CBS+/- mice showed increased oxidative stress, inflammation and decreased nitric oxide. These effects were normalized in CBS+/-/C3H mice. CONCLUSION Our findings demonstrate the dominance of endothelial cell mitochondrial fission over mitochondrial fusion in hyperhomocysteinemia and oxidative stress. This may explain the endothelial cell loss and dysfunction that follows collagen deposition, which contributes to inward aorta remodeling in hypertension.
Collapse
|
23
|
Navneet S, Cui X, Zhao J, Wang J, Kaidery NA, Thomas B, Bollinger KE, Yoon Y, Smith SB. Excess homocysteine upregulates the NRF2-antioxidant pathway in retinal Müller glial cells. Exp Eye Res 2018; 178:228-237. [PMID: 29608906 DOI: 10.1016/j.exer.2018.03.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/14/2018] [Accepted: 03/22/2018] [Indexed: 10/17/2022]
Abstract
This study evaluated the effects of elevated homocysteine (Hcy) on the oxidative stress response in retinal Müller glial cells. Elevated Hcy has been implicated in retinal diseases including glaucoma and optic neuropathy, which are characterized by retinal ganglion cell (RGC) loss. To understand the mechanisms of Hcy-induced RGC loss, in vitro and in vivo models have been utilized. In vitro isolated RGCs are quite sensitive to elevated Hcy levels, while in vivo murine models of hyperhomocysteinemia (HHcy) demonstrate a more modest RGC loss (∼20%) over a period of many months. This differential response to Hcy between isolated cells and the intact retina suggests that the retinal milieu invokes mechanisms that buffer excess Hcy. Oxidative stress has been implicated as a mechanism of Hcy-induced neuron loss and NRF2 is a transcription factor that plays a major role in regulating cytoprotective responses to oxidative stress. In the present study we investigated whether HHcy upregulates NRF2-mediated stress responses in Müller cells, the chief retinal glial cell responsible for providing trophic support to retinal neurons. Primary Müller cells were exposed to L-Hcy-thiolactone [50μM-10mM] and assessed for viability, reactive oxygen species (ROS), and glutathione (GSH) levels. Gene/protein levels of Nrf2 and levels of NRF2-regulated antioxidants (NQO1, CAT, SOD2, HMOX1, GPX1) were assessed in Hcy-exposed Müller cells. Unlike isolated RGCs, isolated Müller cells are viable over a wide range of Hcy concentrations [50 μM - 1 mM]. Moreover, when exposed to elevated Hcy, Müller cells demonstrate decreased oxidative stress and decreased ROS levels. GSH levels increased by ∼20% within 24 h exposure to Hcy. Molecular analyses revealed 2-fold increase in Nrf2 expression. Expression of antioxidant genes Nqo1, Cat, Sod2, Hmox1, Gpx1 increased significantly. The consequences of Hcy exposure were evaluated also in Müller cells harvested from Nrf2-/- mice. In contrast to WT Müller cells, in which oxidative stress decreased upon exposure to Hcy, the Nrf2-/- Müller cells showed a significant increase in oxidative stress. Our data suggest that at least during early stages of Hhcy, a cytoprotective response may be in place, mediated in part by NRF2 in Müller cells.
Collapse
Affiliation(s)
- Soumya Navneet
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Xuezhi Cui
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Jing Zhao
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Navneet Ammal Kaidery
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Bobby Thomas
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Kathryn E Bollinger
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, United States.
| |
Collapse
|
24
|
Ohia SE, Robinson J, Mitchell L, Ngele KK, Heruye S, Opere CA, Njie-Mbye YF. Regulation of Aqueous Humor Dynamics by Hydrogen Sulfide: Potential Role in Glaucoma Pharmacotherapy. J Ocul Pharmacol Ther 2017; 34:61-69. [PMID: 29215951 DOI: 10.1089/jop.2017.0077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hydrogen sulfide (H2S) is a gaseous transmitter with well-known biological actions in a wide variety of tissues and organs. The potential involvement of this gas in physiological and pathological processes in the eye has led to several in vitro, ex vivo, and in vivo studies to understand its pharmacological role in some mammalian species. Evidence from literature demonstrates that 4 enzymes responsible for the biosynthesis of this gas (cystathionine β-synthase, CBS; cystathionine γ-lyase, CSE; 3-mercaptopyruvate sulfurtransferase, 3MST; and d-amino acid oxidase) are present in the cornea, iris, ciliary body, lens, and retina. Studies of the pharmacological actions of H2S (using several compounds as fast- and slow-releasing gas donors) on anterior uveal tissues reveal an effect on sympathetic neurotransmission and the ability of the gas to relax precontracted iris and ocular vascular smooth muscles, responses that were blocked by inhibitors of CSE, CBS, and KATP channels. In the retina, there is evidence that H2S can inhibit excitatory amino acid neurotransmission and can also protect this tissue from a wide variety of insults. Furthermore, exogenous application of H2S-releasing compounds was reported to increase aqueous humor outflow facility in an ex vivo model of the porcine ocular anterior segment and lowered intraocular pressure (IOP) in both normotensive and glaucomatous rabbits. Taken together, the finding that H2S-releasing compounds can lower IOP and can serve a neuroprotective role in the retina suggests that H2S prodrugs could be used as tools or therapeutic agents in diseases such as glaucoma.
Collapse
Affiliation(s)
- Sunny E Ohia
- 1 Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University , Houston, Texas
| | - Jenaye Robinson
- 1 Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University , Houston, Texas
| | - Leah Mitchell
- 1 Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University , Houston, Texas
| | - Kalu K Ngele
- 2 Department of Biology/Microbiology/Biotechnology, Federal University Ndufu Alike Ikwo , Abakaliki, Nigeria
| | - Segewkal Heruye
- 3 Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University , Omaha, Nebraska
| | - Catherine A Opere
- 3 Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University , Omaha, Nebraska
| | - Ya Fatou Njie-Mbye
- 1 Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University , Houston, Texas
| |
Collapse
|
25
|
Jeremic N, Weber GJ, Tyagi SC. Ablation of toll-like receptor 4 mitigates cardiac mitochondrial dysfunction in hyperhomocysteinemia. Can J Physiol Pharmacol 2017; 95:1369-1375. [DOI: 10.1139/cjpp-2016-0744] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hyperhomocysteinemia (HHcy) is a risk factor for adverse cardiovascular events; however, the mechanism for development of this disease is still unknown. Toll-like receptor 4 (TRL4) is a molecule involved in the immune response pathway and is quickly becoming a receptor of interest in the field of hypertension. In this study, we hypothesized that ablation of TLR4 mitigates cardiac mitochondrial dysfunction in a model of HHcy. Five strains of mice (C57BL/6J, CBS+/−, C3H, CBS+/−/C3H, and C3H/HeOuJ) 10–12 weeks old were utilized. We found that HHcy causes heart hypertrophy and promotes oxidative stress while mice with HHcy and inactivated TLR4 showed significant improvement in examined parameters. A dominance of endothelial cell mitochondrial fission over mitochondrial fusion in HHcy and oxidative stress was observed, which may explain the endothelial cell loss and dysfunction that contributes to inward cardiac remodeling.
Collapse
Affiliation(s)
- Nevena Jeremic
- Health Sciences Centre, Department of Physiology, School of Medicine, University of Louisville, 500 South Preston Street, Louisville, KY 40202, USA
- Health Sciences Centre, Department of Physiology, School of Medicine, University of Louisville, 500 South Preston Street, Louisville, KY 40202, USA
| | - Gregory J. Weber
- Health Sciences Centre, Department of Physiology, School of Medicine, University of Louisville, 500 South Preston Street, Louisville, KY 40202, USA
- Health Sciences Centre, Department of Physiology, School of Medicine, University of Louisville, 500 South Preston Street, Louisville, KY 40202, USA
| | - Suresh C. Tyagi
- Health Sciences Centre, Department of Physiology, School of Medicine, University of Louisville, 500 South Preston Street, Louisville, KY 40202, USA
- Health Sciences Centre, Department of Physiology, School of Medicine, University of Louisville, 500 South Preston Street, Louisville, KY 40202, USA
| |
Collapse
|
26
|
Hussein J, El-Bana M, Refaat E, El-Naggar ME. Synthesis of carvacrol-based nanoemulsion for treating neurodegenerative disorders in experimental diabetes. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.08.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
27
|
Familtseva A, Jeremic N, Kunkel GH, Tyagi SC. Toll-like receptor 4 mediates vascular remodeling in hyperhomocysteinemia. Mol Cell Biochem 2017; 433:177-194. [PMID: 28386844 DOI: 10.1007/s11010-017-3026-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/01/2017] [Indexed: 01/12/2023]
Abstract
Although hyperhomocysteinemia (HHcy) is known to promote downstream pro-inflammatory cytokine elevation, the precise mechanism is still unknown. One of the possible receptors that could have significant attention in the field of hypertension is toll-like receptor 4 (TLR-4). TLR-4 is a cellular membrane protein that is ubiquitously expressed in all cell types of the vasculature. Its mutation can attenuate the effects of HHcy-mediated vascular inflammation and mitochondria- dependent cell death that suppresses hypertension. In this review, we observed that HHcy induces vascular remodeling through immunological adaptation, promoting inflammatory cytokine up-regulation (IL-1β, IL-6, TNF-α) and initiation of mitochondrial dysfunction leading to cell death and chronic vascular inflammation. The literature suggests that HHcy promotes TLR-4-driven chronic vascular inflammation and mitochondria-mediated cell death inducing peripheral vascular remodeling. In the previous studies, we have characterized the role of TLR-4 mutation in attenuating vascular remodeling in hyperhomocysteinemia. This review includes, but is not limited to, the physiological synergistic aspects of the downstream elevation of cytokines found within the vascular inflammatory cascade. These events subsequently induce mitochondrial dysfunction defined by excessive mitochondrial fission and mitochondrial apoptosis contributing to vascular remodeling followed by hypertension.
Collapse
Affiliation(s)
- Anastasia Familtseva
- Department of Physiology, School of Medicine, Health Sciences Centre, University of Louisville, A-1215, 500, South Preston Street, Louisville, KY, 40202, USA
| | - Nevena Jeremic
- Department of Physiology, School of Medicine, Health Sciences Centre, University of Louisville, A-1215, 500, South Preston Street, Louisville, KY, 40202, USA.
| | - George H Kunkel
- Department of Physiology, School of Medicine, Health Sciences Centre, University of Louisville, A-1215, 500, South Preston Street, Louisville, KY, 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, Health Sciences Centre, University of Louisville, A-1215, 500, South Preston Street, Louisville, KY, 40202, USA
| |
Collapse
|
28
|
Cui X, Navneet S, Wang J, Roon P, Chen W, Xian M, Smith SB. Analysis of MTHFR, CBS, Glutathione, Taurine, and Hydrogen Sulfide Levels in Retinas of Hyperhomocysteinemic Mice. Invest Ophthalmol Vis Sci 2017; 58:1954-1963. [PMID: 28384716 PMCID: PMC5381329 DOI: 10.1167/iovs.16-21247] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/02/2017] [Indexed: 02/03/2023] Open
Abstract
Purpose Hyperhomocysteinemia (Hhcy) is implicated in certain retinal neurovascular diseases, although whether it is causative remains uncertain. In isolated ganglion cells (GCs), mild Hhcy induces profound death, whereas retinal phenotypes in Hhcy mice caused by mutations in remethylation (methylene tetrahydrofolatereductase [Mthfr+/-]) or transsulfuration pathways (cystathionine β-synthase [Cbs+/-]) demonstrate mild GC loss and mild vasculopathy. The current work investigated compensation in vivo of one pathway for the other, and, because the transsulfuration pathway yields cysteine necessary for formation of glutathione (GSH), taurine, and hydrogen sulfide (H2S), they were analyzed also. Methods Retinas isolated from wild-type (WT), Mthfr+/-, and Cbs+/- mice (12 and 22 weeks) were analyzed for methylene tetrahydrofolate reductase (MTHFR), cystathionine-β-synthase (CBS), and cystathionase (CTH) RNA/protein levels. Retinas were evaluated for levels of reduced:oxidized GSH (GSH:GSSG), Slc7a11 (xCT), taurine, taurine transporter (TAUT), and H2S. Results Aside from decreased CBS RNA/protein levels in Cbs+/- retinas, there were minimal alterations in remethylation/transsulfuration pathways in the two mutant mice strains. Glutathione and taurine levels in Mthfr+/- and Cbs+/- retinas were similar to WT, which may be due to robust levels of xCT and TAUT in mutant retinas. Interestingly, levels of H2S were markedly increased in retinas of Mthfr+/- and Cbs+/- mice compared with WT. Conclusions Ganglion cell loss and vasculopathy observed in Mthfr+/- and Cbs+/- mouse retinas may be milder than expected, not because of compensatory increases of enzymes in remethylation/transsulfuration pathways, but because downstream transsulfuration pathway products GSH, taurine, and H2S are maintained at robust levels. Elevation of H2S is particularly intriguing owing to neuroprotective properties reported for this gasotransmitter.
Collapse
Affiliation(s)
- Xuezhi Cui
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Soumya Navneet
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Jing Wang
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Penny Roon
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Wei Chen
- Department of Chemistry, Washington State University, Pullman, Washington, United States
| | - Ming Xian
- Department of Chemistry, Washington State University, Pullman, Washington, United States
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
29
|
Srivastav K, Saxena S, Mahdi AA, Shukla RK, Meyer CH, Akduman L, Khanna VK. Increased serum level of homocysteine correlates with retinal nerve fiber layer thinning in diabetic retinopathy. Mol Vis 2016; 22:1352-1360. [PMID: 27994434 PMCID: PMC5135738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/30/2016] [Indexed: 11/16/2022] Open
Abstract
PURPOSE To study the correlation between serum levels of vitamin B12, folic acid, and homocysteine and the severity of diabetic retinopathy and the correlation with retinal nerve fiber layer (RNFL) thinning on spectral domain optical coherence tomography (SD-OCT). METHODS In a tertiary care center-based prospective cross-sectional study, 60 consecutive cases and 20 healthy controls in the age group of 40-65 years were included. The eyes of the cases were divided into three groups according to Early Treatment Diabetic Retinopathy Study (ETDRS) classification: diabetes mellitus without retinopathy (n = 20), non-proliferative diabetic retinopathy with macular edema (n = 20), and proliferative diabetic retinopathy with macular edema (n = 20). The serum levels of vitamin B12 and folic acid were measured using a standard protocol. The serum homocysteine assay was performed using an enzyme-linked immunosorbent assay (ELISA) kit. Average RNFL thickness was measured using SD-OCT. Statistical analysis was used to assess the correlations between the study variables. RESULTS Increased severity of diabetic retinopathy was found to correlate with an increase in the serum levels of homocysteine (F = 53.79; p<0.001). The mean serum levels of vitamin B12 and folic acid were found to be within the normal reference range. A positive correlation was found between retinal nerve fiber layer thinning and serum levels of homocysteine (p<0.001). CONCLUSIONS This study, for the first time, demonstrated a correlation between increased homocysteine with a decrease in RNFL thickness and increased severity of diabetic retinopathy.
Collapse
Affiliation(s)
- Khushboo Srivastav
- Retina Service, Department of Ophthalmology, King George’s Medical University, Lucknow, India
| | - Sandeep Saxena
- Retina Service, Department of Ophthalmology, King George’s Medical University, Lucknow, India
| | - Abbas A. Mahdi
- Department of Biochemistry, King George’s Medical University, Lucknow, India
| | | | | | - Levent Akduman
- Vitreoretinal and uveitis service, Department of Ophthalmology, Saint Louis University School of Medicine,St. Louis, MO
| | - Vinay K. Khanna
- CSIR - Indian Institute of Toxicology and Research, Lucknow, India
| |
Collapse
|
30
|
Brown CJ. Preservation of retinal structure and function after cilioretinal artery occlusion: a case report. Int Med Case Rep J 2016; 9:29-34. [PMID: 26929671 PMCID: PMC4755464 DOI: 10.2147/imcrj.s96858] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cilioretinal artery occlusion is a cause of sudden, often catastrophic loss of central vision. There are no established effective treatments. Recently, a patient presented 24 hours after a cilioretinal artery occlusion, following a cardiac catheterization prior to which her blood thinners had been discontinued. Lacking an effective way to address the severe retinal ischemic oxidative stress, she was offered, under compassionate use, a multivitamin complex designed to address retinal ischemia and oxidative stress. Significant components of this product are L-methylfolate and n-acetyl cysteine. The patient experienced a rapid unexpected improvement in vision and preservation of retinal structure, suggesting that marked improvement in retinal artery occlusions outcomes may be possible as late as 24 hours postocclusion. This is the third reported case of cilioretinal artery occlusion associated with cardiac catheterization.
Collapse
|
31
|
Brooks BP, Thompson AH, Sloan JL, Manoli I, Carrillo-Carrasco N, Zein WM, Venditti CP. Ophthalmic Manifestations and Long-Term Visual Outcomes in Patients with Cobalamin C Deficiency. Ophthalmology 2016; 123:571-82. [PMID: 26825575 DOI: 10.1016/j.ophtha.2015.10.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/25/2015] [Accepted: 10/25/2015] [Indexed: 01/07/2023] Open
Abstract
PURPOSE To explore the ocular manifestations of cobalamin C (cblC) deficiency, an inborn error of intracellular vitamin B12 metabolism. DESIGN Retrospective, observational case series. PARTICIPANTS Twenty-five cblC patients underwent clinical and ophthalmic examination at the National Institutes of Health between August 2004 and September 2012. Patient ages ranged from 2 to 27 years at last ophthalmic visit, and follow-up ranged from 0 to 83 months (median, 37 months; range, 13-83 months) over a total of 69 visits. METHODS Best-corrected visual acuity, slit-lamp biomicroscopy, dilated fundus examination, wide-field photography, fundus autofluorescence imaging, sedated electroretinography, optical coherence tomography, genetics and metabolite assessment. MAIN OUTCOME MEASURES Visual acuity and presence and degree of retinal degeneration and optic nerve pallor. RESULTS Nystagmus (64%), strabismus (52%), macular degeneration (72%), optic nerve pallor (68%), and vascular changes (64%) were present. c.271dupA (p.R91KfsX14) homozygous patients (n = 14) showed early and extensive macular degeneration. Electroretinography showed that scotopic and photopic responses were reduced and delayed, but were preserved remarkably in some patients despite severe degeneration. Optical coherence tomography images through the central macular lesion of a patient with severe retinal degeneration showed extreme thinning, some preservation of retinal lamination, and nearly complete loss of the outer nuclear layer. Despite hyperhomocysteinemia, no patients exhibited lens dislocation. CONCLUSIONS This longitudinal study reports ocular outcomes in the largest group of patients with cblC deficiency systematically examined at a single center over an extended period. Differences in progression and severity of macular degeneration, optic nerve pallor, and vascular attenuation between homozygous c.271dupA (p.R91KfsX14) patients and compound heterozygotes were noted. The pace and chronicity of ophthalmic manifestations lacked strict correlation to metabolic status as measured during visits. Prenatal or early treatment, or both, may have mitigated ocular disease, leading to better functional acuity, but patients still progressed to severe macular degeneration. The effects of prenatal or early treatment, or both, in siblings; the manifestation of severe disease in infancy; the presence of comorbid developmental abnormalities; and the possible laminar structural defect noted in many patients are findings showing that cblC deficiency displays a developmental as well as a degenerative ocular phenotype.
Collapse
Affiliation(s)
- Brian P Brooks
- National Eye Institute, Ophthalmic Genetics and Visual Function Branch, National Institutes of Health, Bethesda, Maryland; National Human Genome Research Institute, Genetics and Molecular Biology Branch, National Institutes of Health, Bethesda, Maryland.
| | - Amy H Thompson
- National Eye Institute, Ophthalmic Genetics and Visual Function Branch, National Institutes of Health, Bethesda, Maryland
| | - Jennifer L Sloan
- National Human Genome Research Institute, Genetics and Molecular Biology Branch, National Institutes of Health, Bethesda, Maryland
| | - Irini Manoli
- National Human Genome Research Institute, Genetics and Molecular Biology Branch, National Institutes of Health, Bethesda, Maryland
| | - Nuria Carrillo-Carrasco
- National Center for Advancing Translational Sciences, Therapeutics for Rare and Neglected Diseases, National Institutes of Health, Bethesda, Maryland
| | - Wadih M Zein
- National Eye Institute, Ophthalmic Genetics and Visual Function Branch, National Institutes of Health, Bethesda, Maryland
| | - Charles P Venditti
- National Human Genome Research Institute, Genetics and Molecular Biology Branch, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
32
|
Ola MS, Alhomida AS. Neurodegeneration in diabetic retina and its potential drug targets. Curr Neuropharmacol 2014; 12:380-6. [PMID: 25342945 PMCID: PMC4207077 DOI: 10.2174/1570159x12666140619205024] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/27/2014] [Accepted: 06/19/2014] [Indexed: 12/04/2022] Open
Abstract
Diabetic retinopathy (DR) is one of the major complications of diabetes causing vision loss and blindness worldwide. DR is widely recognized as a neurodegenerative disease as evidenced from early changes at cellular and molecular levels in the neuronal component of the diabetic retina, which is further supported by various retinal functional tests indicating functional deficits in the retina soon after diabetes progression. Diabetes alters the level of a number of neurodegenerative metabolites, which increases influx through several metabolic pathways which in turn induce an increase in oxidative stress and a decrease in neurotrophic factors, thereby damage retinal neurons. Loss of neurons may implicate in vascular pathology, a clinical signs of DR observed at later stages of the disease. Here, we discuss diabetes-induced potential metabolites known to be detrimental to neuronal damage and their mechanism of action. In addition, we highlight important neurotrophic factors, whose level have been found to be dysregulated in diabetic retina and may damage neurons. Furthermore, we discuss potential drugs and strategies based on targeting diabetes-induced metabolites, metabolic pathways, oxidative stress, and neurotrophins to protect retinal neurons, which may ameliorate vision loss and vascular damage in DR.
Collapse
Affiliation(s)
- Mohammad Shamsul Ola
- Department of Biochemistry, College of Science, King Saud University, Riyadh, KSA, Saudi Arabia
| | - Abdullah S Alhomida
- Department of Biochemistry, College of Science, King Saud University, Riyadh, KSA, Saudi Arabia
| |
Collapse
|
33
|
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: 3.9] [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
|
34
|
Tawfik A, Smith SB. Increased ER stress as a mechanism of retinal neurovasculopathy in mice with severe hyperhomocysteinemia. AUSTIN JOURNAL OF CLINICAL OPHTHALMOLOGY 2014; 1:1023. [PMID: 25580465 PMCID: PMC4287205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Hyperhomocysteinemia is implicated in retinal neurovascular diseases including arterial occlusive disease, venous occlusive disease and pseudoexfoliation glaucoma. The mechanism for these diseases is not known. Here we used hyperhomocysteinemic mice lacking the gene encoding cystathionine-beta-synthase (cbs-/-) to examine whether ER stress could be a mechanism for the retinal neurovasculopathy reported in these mice. Retinas of cbs+/+ and cbs-/- mice (age: 3-5 wks) were used to investigate the expression of ER stress genes (BiP/GRP78, Perk, Atf6, Atf4, Ire1α, Chop) and the proteins they encode. The levels of poly(ADP-ribose) polymerase (PARP) and cleaved cysteine-aspartic proteases-3 (caspase-3), proteins known to be involved in apoptosis, were also examined. Quantitative reverse transcription polymerase chain reaction and western blotting revealed an increase in BiP/GRP78 and PERK in retinas of cbs-/- mice compared with cbs+/+ mice. There was an elevation of CCAAT-enhancer-binding protein homologous protein (CHOP) in retinal cryosections of cbs-/- mice indicating apoptosis, which was confirmed by increased levels of PARP and cleaved caspase-3. The data suggest that the genes and proteins that are major players in the ER stress pathway, particularly the PERK pathway, are upregulated in retinas of cbs-/- mice. The data support a role for ER stress in the pathophysiology associated with the hyperhomocysteinemia-linked retinal disease.
Collapse
Affiliation(s)
- Amany Tawfik
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, GA
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, GA
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, GA
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, GA
- Department of Ophthalmology, Medical College of Georgia, Georgia Regents University, Augusta, GA
| |
Collapse
|
35
|
Familtseva A, Kalani A, Chaturvedi P, Tyagi N, Metreveli N, Tyagi SC. Mitochondrial mitophagy in mesenteric artery remodeling in hyperhomocysteinemia. Physiol Rep 2014; 2:e00283. [PMID: 24771691 PMCID: PMC4001876 DOI: 10.14814/phy2.283] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Although high levels of homocysteine also termed as hyperhomocysteinemia (HHcy) has been associated with inflammatory bowel disease and mesenteric artery occlusion, the mitochondrial mechanisms behind endothelial dysfunction that lead to mesenteric artery remodeling are largely unknown. We hypothesize that in HHcy there is increased mitochondrial fission due to altered Mfn‐2/Drp‐1 ratio, which leads to endothelial dysfunction and collagen deposition in the mesenteric artery inducing vascular remodeling. To test this hypothesis, we used four groups of mice: (i) WT (C57BL/6J); (ii) mice with HHcy (CBS+/−); (iii) oxidative stress resistant mice (C3H) and (iv) mice with HHcy and oxidative stress resistance (CBS+/−/C3H). For mitochondrial dynamics, we studied the expression of Mfn‐2 which is a mitochondrial fusion protein and Drp‐1 which is a mitochondrial fission protein by western blots, real‐time PCR and immunohistochemistry. We also examined oxidative stress markers, endothelial cell, and gap junction proteins that play an important role in endothelial dysfunction. Our data showed increase in oxidative stress, mitochondrial fission (Drp‐1), and collagen deposition in CBS+/− compared to WT and C3H mice. We also observed significant down regulation of Mfn‐2 (mitochondrial fusion marker), CD31, eNOS and connexin 40 (gap junction protein) in CBS+/− mice as compared to WT and C3H mice. In conclusion, our data suggested that HHcy increased mitochondrial fission (i.e., decreased Mfn‐2/Drp‐1 ratio, causing mitophagy) that leads to endothelial cell damage and collagen deposition in the mesenteric artery. This is a novel report on the role of mitochondrial dynamics alteration defining mesenteric artery remodeling. e00283 This article is a novel report on the role of mitochondrial dynamics in mesenteric artery remodeling during hyperhomocysteinemia. The study can contribute significantly toward understanding the mesenteric mitochondrial mechanisms underpinning inflammatory bowel disease – a major clinical concern.
Collapse
Affiliation(s)
- Anastasia Familtseva
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, 40202, Kentucky
| | | | | | | | | | | |
Collapse
|
36
|
Alaimo A, Gorojod RM, Beauquis J, Muñoz MJ, Saravia F, Kotler ML. Deregulation of mitochondria-shaping proteins Opa-1 and Drp-1 in manganese-induced apoptosis. PLoS One 2014; 9:e91848. [PMID: 24632637 PMCID: PMC3954806 DOI: 10.1371/journal.pone.0091848] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 02/17/2014] [Indexed: 01/15/2023] Open
Abstract
Mitochondria are dynamic organelles that undergo fusion and fission processes. These events are regulated by mitochondria-shaping proteins. Changes in the expression and/or localization of these proteins lead to a mitochondrial dynamics impairment and may promote apoptosis. Increasing evidence correlates the mitochondrial dynamics disruption with the occurrence of neurodegenerative diseases. Therefore, we focused on this topic in Manganese (Mn)-induced Parkinsonism, a disorder associated with Mn accumulation preferentially in the basal ganglia where mitochondria from astrocytes represent an early target. Using MitoTracker Red staining we observed increased mitochondrial network fission in Mn-exposed rat astrocytoma C6 cells. Moreover, Mn induced a marked decrease in fusion protein Opa-1 levels as well as a dramatic increase in the expression of fission protein Drp-1. Additionally, Mn provoked a significant release of high MW Opa-1 isoforms from the mitochondria to the cytosol as well as an increased Drp-1 translocation to the mitochondria. Both Mdivi-1, a pharmacological Drp-1 inhibitor, and rat Drp-1 siRNA reduced the number of apoptotic nuclei, preserved the mitochondrial network integrity and prevented cell death. CsA, an MPTP opening inhibitor, prevented mitochondrial Δψm disruption, Opa-1 processing and Drp-1 translocation to the mitochondria therefore protecting Mn-exposed cells from mitochondrial disruption and apoptosis. The histological analysis and Hoechst 33258 staining of brain sections of Mn-injected rats in the striatum showed a decrease in cellular mass paralleled with an increase in the occurrence of apoptotic nuclei. Opa-1 and Drp-1 expression levels were also changed by Mn-treatment. Our results demonstrate for the first time that abnormal mitochondrial dynamics is implicated in both in vitro and in vivo Mn toxicity. In addition we show that the imbalance in fusion/fission equilibrium might be involved in Mn-induced apoptosis. This knowledge may provide new therapeutic tools for the treatment of Manganism and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Agustina Alaimo
- Laboratorio de Apoptosis en el Sistema Nervioso y Nano-Oncología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Química Biológica, Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Roxana M. Gorojod
- Laboratorio de Apoptosis en el Sistema Nervioso y Nano-Oncología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Química Biológica, Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Juan Beauquis
- Laboratorio de Neurobiología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Manuel J. Muñoz
- Departamento de Fisiología, Biología Molecular y Celular and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Saravia
- Laboratorio de Neurobiología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Mónica L. Kotler
- Laboratorio de Apoptosis en el Sistema Nervioso y Nano-Oncología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Química Biológica, Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
37
|
Markand S, Tawfik A, Ha Y, Gnana-Prakasam J, Sonne S, Ganapathy V, Sen N, Xian M, Smith SB. Cystathionine beta synthase expression in mouse retina. Curr Eye Res 2013; 38:597-604. [PMID: 23470016 DOI: 10.3109/02713683.2013.774024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
UNLABELLED Abstract Purpose: Cystathionine β-synthase (CBS), a key enzyme in the transsulfuration metabolic pathway, converts homocysteine to cystathionine, which is converted to cysteine required for the synthesis of major retinal antioxidant glutathione (GSH). Enzyme activity assays suggest that CBS is present in human and pig retina, however recent studies reported that CBS is not expressed in mouse retina. We found this species difference puzzling. Given the plethora of studies using mouse retina as a model system, coupled with the importance of GSH in retina, we investigated CBS expression in mouse retina at the molecular and cell biological level. METHODS Wildtype (WT) mice or mice lacking the gene encoding CBS (cbs(-)(/)(-)) were used in these studies. RNA and protein were isolated from retinas and liver (positive control) for the analysis of cbs gene expression by RT-PCR and CBS protein expression by Western blotting, respectively. CBS was analyzed by immunofluorescence in retinal cryosections and primary retinal cells (ganglion, Müller, retinal pigment epithelial). CBS enzyme activity was measured in primary Müller cells. RESULTS RT-PCR revealed robust cbs expression in WT liver, brain and retina. Western blotting detected CBS in retina, brain and liver of WT mice, but not in cbs(-)(/)(-) mice liver. In immunohistochemical studies, CBS was present abundantly in the ganglion cell layer of retina; it was detected also in primary isolations of Müller, RPE and ganglion cells. CBS activity was detected in Müller cells by fluorescent detection of H2S. CONCLUSIONS We have compelling molecular evidence that CBS is expressed in mouse retina at the gene and protein level. Our immunofluorescence data suggest that it is present in several retinal cell types and the data from the enzyme activity assay suggest activity in Müller cells. These findings set the stage to investigate the role of CBS and the transsulfuration pathway in the generation of GSH in mouse retina.
Collapse
Affiliation(s)
- Shanu Markand
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA 30912-2000, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
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: 2.9] [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
|
39
|
Ola MS, Nawaz MI, Khan HA, Alhomida AS. Neurodegeneration and neuroprotection in diabetic retinopathy. Int J Mol Sci 2013; 14:2559-72. [PMID: 23358247 PMCID: PMC3588002 DOI: 10.3390/ijms14022559] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 01/12/2013] [Accepted: 01/17/2013] [Indexed: 02/06/2023] Open
Abstract
Diabetic retinopathy is widely considered to be a neurovascular disease. This is in contrast to its previous identity as solely a vascular disease. Early in the disease progression of diabetes, the major cells in the neuronal component of the retina consist of retinal ganglion cells and glial cells, both of which have been found to be compromised. A number of retinal function tests also indicated a functional deficit in diabetic retina, which further supports dysfunction of neuronal cells. As an endocrinological disorder, diabetes alters metabolism both systemically and locally in several body organs, including the retina. A growing body of evidences indicates increased levels of excitotoxic metabolites, including glutamate, branched chain amino acids and homocysteine in cases of diabetic retinopathy. Also present, early in the disease, are decreased levels of folic acid and vitamin-B12, which are potential metabolites capable of damaging neurons. These altered levels of metabolites are found to activate several metabolic pathways, leading to increases in oxidative stress and decreases in the level of neurotrophic factors. As a consequence, they may damage retinal neurons in diabetic patients. In this review, we have discussed those potential excitotoxic metabolites and their implications in neuronal damage. Possible therapeutic targets to protect neurons are also discussed. However, further research is needed to understand the exact molecular mechanism of neurodegeneration so that effective neuroprotection strategies can be developed. By protecting retinal neurons early in diabetic retinopathy cases, damage of retinal vessels can be protected, thereby helping to ameliorate the progression of diabetic retinopathy, a leading cause of blindness worldwide.
Collapse
Affiliation(s)
- Mohammad Shamsul Ola
- Department of Biochemistry, Faculty of Science, King Saud University, Riyadh 11415, Saudi Arabia.
| | | | | | | |
Collapse
|
40
|
Poddar R, Paul S. Novel crosstalk between ERK MAPK and p38 MAPK leads to homocysteine-NMDA receptor-mediated neuronal cell death. J Neurochem 2012; 124:558-70. [PMID: 23176034 DOI: 10.1111/jnc.12102] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 11/20/2012] [Accepted: 11/21/2012] [Indexed: 11/29/2022]
Abstract
Hyperhomocysteinemia is an independent risk factor for both acute and chronic neurological disorders, but little is known about the underlying mechanisms by which elevated homocysteine can promote neuronal cell death. We recently established a role for NMDA receptor-mediated activation of extracellular signal-regulated kinase (ERK)-MAPK in homocysteine-induced neuronal cell death. In this study, we examined the involvement of the stress-induced MAPK, p38 in homocysteine-induced neuronal cell death, and further explored the relationship between the two MAPKs, ERK and p38, in triggering cell death. Homocysteine-mediated NMDA receptor stimulation and subsequent Ca(2+) influx led to a biphasic activation of p38 MAPK characterized by an initial rapid, but transient activation followed by a delayed and more prolonged response. Selective inhibition of the delayed p38 MAPK activity was sufficient to attenuate homocysteine-induced neuronal cell death. Using pharmacological and RNAi approaches, we further demonstrated that both the initial and delayed activation of p38 MAPK is downstream of, and dependent on activation of ERK MAPK. Our findings highlight a novel interplay between ERK and p38 MAPK in homocysteine-NMDA receptor-induced neuronal cell death.
Collapse
Affiliation(s)
- Ranjana Poddar
- University of New Mexico Health Sciences Center, Department of Neurology, 1, University of New Mexico, Albuquerque, NM 87131, USA.
| | | |
Collapse
|
41
|
Age-related changes in visual function in cystathionine-beta-synthase mutant mice, a model of hyperhomocysteinemia. Exp Eye Res 2011; 96:124-31. [PMID: 22197750 DOI: 10.1016/j.exer.2011.12.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 12/06/2011] [Accepted: 12/09/2011] [Indexed: 11/24/2022]
Abstract
Homocysteine is an amino acid required for the metabolism of methionine. Excess homocysteine is implicated in cardiovascular and neurological disease and new data suggest a role in various retinopathies. Mice lacking cystathionine-beta-synthase (cbs(-/-)) have an excess of retinal homocysteine and develop anatomical abnormalities in multiple retinal layers, including photoreceptors and ganglion cells; heterozygous (cbs(+/-)) mice demonstrate ganglion cell loss and mitochondrial abnormalities in the optic nerve. The purpose of the present study was to determine whether elevated homocysteine, due to absent or diminished cbs, alters visual function. We examined cbs(-/-) (3 weeks) and cbs(+/-) mice (5, 10, 15, 30 weeks) and results were compared to those obtained from wild type (WT) littermates. Conventional dark- and light-adapted ERGs were recorded, along with dc-ERG to assess retinal pigment epithelial (RPE) function. The visual evoked potential (VEP) was used to assess transmission to the visual cortex. The amplitudes of the major ERG components were reduced in cbs(-/-) mice at age 3 weeks and VEPs were delayed markedly. These findings are consistent with the early retinal disruption observed anatomically in these mice. In comparison, at 3 weeks of age, responses of cbs(+/-) mice did not differ significantly from those of WT mice. Functional abnormalities were not observed in cbs(+/-) mice until 15 weeks of age, at which time amplitude reductions were noted for the ERG a- and b-wave and the light peak component, but not for other components generated by the RPE. VEP implicit times were delayed in cbs(+/-) mice at 15 and 30 weeks, while VEP amplitudes were unaffected. The later onset of functional defects in cbs(+/-) mice is consistent with a slow loss of ganglion cells reported previously in the heterozygous mutant. Light peak abnormalities indicate that RPE function is also compromised in older cbs(+/-) mice. The data suggest that severe elevations of homocysteine are associated with marked alterations of retinal function while modest homocysteine elevation is reflected in milder and delayed alterations of retinal function. The work lays the foundation to explore the role of homocysteine in retinal diseases such as glaucoma and optic neuropathy.
Collapse
|