1
|
Huang A, Ma J, Zhu H, Qi Y, Jin Y, Zhang M, Yin L, Luo M, Chen S, Xie C, Huang H. Blood metabolites mediate causal inference studies on the effect of gut microbiota on the risk of vascular calcification. J Adv Res 2025:S2090-1232(25)00198-5. [PMID: 40139524 DOI: 10.1016/j.jare.2025.03.038] [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: 01/17/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Emerging evidence indicates a notable connection between gut microbiota and Vascular Calcification (VC). Gut microbiota influences various disease processes through host metabolic pathways; however, the causative link between gut microbiota and VC, along with the potential mediating role of metabolites, is still not well understood. METHODS We leveraged data from the largest Genome-Wide Association Studies (GWAS) concerning gut microbiota, blood metabolites, and VC. To explore the causal relationships among these variables, we conducted two-sample bidirectional Mendelian Randomization (MR) analyses. Furthermore, mediation analyses were conducted to determine if metabolites act as an intermediary in the impact of gut microbiota on VC. In addition, we recruited CKD patients for mass spectrometry and CT examination, and performed a correlation analysis between the expression of blood metabolites and VC score. Finally, we experimentally validated the effects of intermediate metabolites on VC. RESULTS We identified 19 positive gut microbiota species and 52 positive blood metabolites with causal effects on VC. Additionally, the onset of VC was found to induce changes in the abundance of 24 gut microbiota species and 56 metabolites. Further analyses revealed that up to 13 positive gut microbiota species regulate the expression of 20 positive metabolites. Mediation analysis suggests that the gut microbiota g_KLE1615 promotes VC by downregulating the methionine-to-phosphate ratio. Mass spectrometry results indicate that over half of the metabolites identified through MR analysis show altered expression during CKD progression. Among them, 7 metabolites were significantly associated with the progression of VC. Further in vitro experiments confirmed the inhibitory effect of the intermediate metabolite methionine on VC. CONCLUSION Gut microbiota and blood metabolites are causally linked to VC. These findings provide a theoretical basis for microbiome- and metabolome-based therapeutic strategies for targeting VC and enhances our comprehension of the gut-vascular axis.
Collapse
Affiliation(s)
- Aoran Huang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Jianshuai Ma
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Huijin Zhu
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Yanli Qi
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Yang Jin
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Mingxuan Zhang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Li Yin
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Minhong Luo
- Department of Nephrology, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Sifan Chen
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510000, China
| | - Chen Xie
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China.
| | - Hui Huang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China.
| |
Collapse
|
2
|
Karuntu JS, Almushattat H, Nguyen XTA, Plomp AS, Wanders RJA, Hoyng CB, van Schooneveld MJ, Schalij-Delfos NE, Brands MM, Leroy BP, van Karnebeek CDM, Bergen AA, van Genderen MM, Boon CJF. Syndromic retinitis pigmentosa. Prog Retin Eye Res 2024; 107:101324. [PMID: 39733931 DOI: 10.1016/j.preteyeres.2024.101324] [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: 07/17/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024]
Abstract
Retinitis pigmentosa (RP) is a progressive inherited retinal dystrophy, characterized by the degeneration of photoreceptors, presenting as a rod-cone dystrophy. Approximately 20-30% of patients with RP also exhibit extra-ocular manifestations in the context of a syndrome. This manuscript discusses the broad spectrum of syndromes associated with RP, pathogenic mechanisms, clinical manifestations, differential diagnoses, clinical management approaches, and future perspectives. Given the diverse clinical and genetic landscape of syndromic RP, the diagnosis may be challenging. However, an accurate and timely diagnosis is essential for optimal clinical management, prognostication, and potential treatment. Broadly, the syndromes associated with RP can be categorized into ciliopathies, inherited metabolic disorders, mitochondrial disorders, and miscellaneous syndromes. Among the ciliopathies associated with RP, Usher syndrome and Bardet-Biedl syndrome are the most well-known. Less common ciliopathies include Cohen syndrome, Joubert syndrome, cranioectodermal dysplasia, asphyxiating thoracic dystrophy, Mainzer-Saldino syndrome, and RHYNS syndrome. Several inherited metabolic disorders can present with RP, including Zellweger spectrum disorders, adult Refsum disease, α-methylacyl-CoA racemase deficiency, certain mucopolysaccharidoses, ataxia with vitamin E deficiency, abetalipoproteinemia, several neuronal ceroid lipofuscinoses, mevalonic aciduria, PKAN/HARP syndrome, PHARC syndrome, and methylmalonic acidaemia with homocystinuria type cobalamin (cbl) C disease. Due to the mitochondria's essential role in supplying continuous energy to the retina, disruption of mitochondrial function can lead to RP, as seen in Kearns-Sayre syndrome, NARP syndrome, primary coenzyme Q10 deficiency, SSBP1-associated disease, and long chain 3-hydroxyacyl-CoA dehydrogenase deficiency. Lastly, Cockayne syndrome and PERCHING syndrome can present with RP, but they do not fit the abovementioned hierarchy and are thus categorized as miscellaneous. Several first-in-human clinical trials are underway or in preparation for some of these syndromic forms of RP.
Collapse
Affiliation(s)
- Jessica S Karuntu
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hind Almushattat
- Department of Ophthalmology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Xuan-Thanh-An Nguyen
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - Astrid S Plomp
- Department of Human Genetics, Amsterdam Reproduction & Development, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Ronald J A Wanders
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam, the Netherlands; Department of Laboratory Medicine, Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam, the Netherlands
| | - Carel B Hoyng
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mary J van Schooneveld
- Bartiméus Diagnostic Center for Complex Visual Disorders, Zeist, the Netherlands; Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Marion M Brands
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Bart P Leroy
- Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium; Department of Head & Skin, Ghent University, Ghent, Belgium; Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; Division of Ophthalmology and Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Clara D M van Karnebeek
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Arthur A Bergen
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands; Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Department of Human Genetics, Section Ophthalmogenetics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Maria M van Genderen
- Bartiméus Diagnostic Center for Complex Visual Disorders, Zeist, the Netherlands; Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Camiel J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Correa-Navarro VA, Romo-Morales GDC, Sánchez-Palafox JE, Rodríguez-Ríos D, Molina-Torres J, Ramírez-Chávez E, Zaina S, Lund G. A Survey of Fatty Acid Content of the Male Reproductive System in Mice Supplemented With Arachidonic Acid. J Lipids 2024; 2024:3351340. [PMID: 39734583 PMCID: PMC11671656 DOI: 10.1155/jl/3351340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/15/2024] [Indexed: 12/31/2024] Open
Abstract
Paternal exposure to high-fat diets or individual fatty acids (FAs) including arachidonic acid (AA) modifies progeny traits by poorly understood mechanisms. Specific male reproductive system FAs may be involved in paternal inheritance, as they can modify a range of cellular components, including the epigenome. Our objective was to determine FAs in compartments of the male reproductive system that potentially affect ejaculate composition-right and left testicular interstitial fluid (TIF), vesicular gland fluid (VGF), and epididymal adipose tissue (EAT)-in mice exposed to AA or vehicle daily for 10 days (n = 9-10/group). Whole blood (WB) and interscapular brown adipose tissue (IBAT) FA profiles were used as reference. AA significantly affected only VGF FAs relative to vehicle, that is, increased and decreased levels of arachidic and docosahexaenoic acid, respectively, versus vehicle (0.28% ± 0.01% and 0.23% ± 0.03%, respectively, p = 0.049, and 2.42% ± 0.47% and 3.00% ± 0.58%, respectively, p = 0.041). AA affected distinct FAs in WB. Additionally, we uncovered AA-dependent and AA-independent FA laterality. Myristic acid was higher in AA-exposed left versus right TIF (0.68% ± 0.35% and 0.60% ± 0.11%, respectively, p = 0.004). Right TIF contained higher oleic and linoleic acid and lower stearic acid than left TIF (29.01% ± 3.07% and 24.00% ± 2.18%, respectively, p = 0.005; 9.14% ± 1.88% and 7.05% ± 1.36%, respectively, p = 0.005; and 21.90% ± 2.92% and 26.01% ± 2.46%, respectively, p = 0.036), irrespective of exposure to AA. The TIF oleic/stearic acid ratio suggested higher Stearoyl-CoA Desaturase 1 activity in the right versus the left testis (1.35 ± 0.32 and 1.00 ± 0.17, respectively, p = 1.0 × 10-4). Multitissue comparisons revealed that TIF and VGF FA profiles were distinct from WB, EAT, or IBAT counterparts, suggesting tissue-specific FA fingerprints. In conclusion, AA modulated selected VGF long-chain FAs that may impact on uterine inflammation and subsequent embryonic development. AA altered local FA synthesis or selective uptake, rather than eliciting passive uptake from WB. Additionally, we uncover a significant laterality of testis FAs that may result in asymmetric sperm cell phenotypes.
Collapse
Affiliation(s)
- Viridiana Abigail Correa-Navarro
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Gloria del Carmen Romo-Morales
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Jaime Eduardo Sánchez-Palafox
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Dalia Rodríguez-Ríos
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| | - Jorge Molina-Torres
- Department of Biotechnology and Biochemistry, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| | - Enrique Ramírez-Chávez
- Department of Biotechnology and Biochemistry, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| | - Silvio Zaina
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Gertrud Lund
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| |
Collapse
|
4
|
Zhang R, Zhang Y, Shao Z. Dietary Intake of Branched-Chain Fatty Acids, Metabolic Parameters, High-Sensitivity C-Reactive Protein Levels, and Anthropometric Features Among Elite and Subelite Soccer Players. Int J Sport Nutr Exerc Metab 2024; 34:372-377. [PMID: 39142643 DOI: 10.1123/ijsnem.2024-0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Several studies have revealed the positive healthy impacts of branched-chain fatty acids (BCFAs). However, most of these studies evaluated the serum BCFAs in humans, or treatment with exogenous BCFAs in animal or in-vitro models and the health impacts of dietary BCFAs have not yet been studied. Due to positive effects of BCFAs in sport, in the current study, we aimed to investigate the association between dietary BCFAs and metabolic and inflammatory parameters among elite and subelite soccer players. METHODS A cross-sectional study was carried out among 335 elite and subelite soccer players (196 male), aged between 20 and 45 years old. Soccer players were enrolled from 32 teams under the directive of The Chinese Football Association. Demographic, anthropometric, and dietary assessments were performed and laboratory measurement including serum lipids, glycemic markers, and high-sensitivity C-reactive protein was measured. RESULTS Those with the highest dietary BCFAs consumption had higher appetite (p = .009). Also, high consumption of dietary BCFAs was associated with lower diastolic blood pressure (odds ratio: 0.958; confidence interval: 0.918-0.999; p = .046) and low high-sensitivity C-reactive protein concentrations in the third tertile of dietary BCFAs (odds ratio: 0.431; confidence interval: 0.300-0.618; p < .001). No other association between biochemical variables and dietary BCFAs was found. CONCLUSION As shown in the current study, higher dietary BCFAs consumption was associated with lower diastolic blood pressure and inflammation. Due to very limited number of studies, further studies are needed to have a better perspective of these associations and their underlying mechanisms.
Collapse
Affiliation(s)
- Rui Zhang
- College of Sciences, North China University of Technology, Bei Jing, China
| | - Yuyao Zhang
- Faculty of Education, University of Macau, Macau, China
| | - Zhe Shao
- Sports-Department, Beijing Technology and Business University, Bei Jing, China
| |
Collapse
|
5
|
Torequl Islam M, Shimul Bhuia M, Paulo Martins de Lima J, Paulo Araujo Maia F, Beatriz Herminia Ducati A, Douglas Melo Coutinho H. Phytanic acid, an inconclusive phytol metabolite: A review. Curr Res Toxicol 2023; 5:100120. [PMID: 37744206 PMCID: PMC10515296 DOI: 10.1016/j.crtox.2023.100120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/28/2023] [Accepted: 08/30/2023] [Indexed: 09/26/2023] Open
Abstract
Phytanic acid (PA: 3,7,11,15-tetramethylhexadecanoic acid) is an important biometabolite of the chlorophyll-derived diterpenoid phytol. Its biological sources (occurrence) and ADME (absorption, distribution, metabolism, and elimination) profile are well-discussed in the literature. Cumulative literature suggests that PA has beneficial as well as harmful biological roles in humans and other animals. This study aimed to sketch a brief summary of PA's beneficial and harmful pharmacological effects in test systems on the basis of existing literature reports. Literature findings propose that PA has anti-inflammatory and immunomodulatory, antidiabetic, anti-obesity, anticancer, and oocyte maturation effects. Although a high plasma PA-level mediated SLS remains controversial, it is evident to link it with Refsum's disease and other peroxisomal enzyme deficiency diseases in humans, including RCDP and LD; ZHDA and Alzheimer's disease; progressive ataxia and dysarthria; and an increased risk of some lymphomas such as LBL, FL, and NHL. PA exerts toxic effects on different kinds of cells, including neuronal, cardiac, and renal cells, through diverse pathways such as oxidative stress, mitochondrial disturbance, apoptosis, disruption of Na+/K+-ATPase activity, Ca2+ homeostasis, alteration of AChE and MAO activities, etc. PA is considered a cardiac biomarker in humans. In conclusion, PA may be one of the most important biometabolites in humans.
Collapse
Affiliation(s)
- Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Md. Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | | | | | | | | |
Collapse
|
6
|
Slanina AM, Coman AE, Anton-Păduraru DT, Popa E, Barbacariu CL, Novac O, Petroaie AD, Bacușcă AI, Manole M, Cosmescu A. PEX6 Mutation in a Child with Infantile Refsum Disease—A Case Report and Literature Review. CHILDREN 2023; 10:children10030530. [PMID: 36980088 PMCID: PMC10047879 DOI: 10.3390/children10030530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
The aim of this paper is to describe the temporal progression and clinical picture of a 2-year-old child with infantile Refsum disease, as well as the diagnostic procedures performed; this case presented multiple hematologic, metabolic, and developmental complications and progressive disabilities. Genetic testing revealed a mutation of the PEX6 (Peroxisomal Biogenesis Factor 6) gene, and the metabolic profile was consistent with the diagnosis. Particularly, the child also presented altered coagulation factors and developed a spontaneous brain hemorrhage. The clinical picture includes several neurological, ophthalmological, digestive, cutaneous, and endocrine disorders as a result of the very long chain fatty acid accumulation as well as secondary oxidative anomalies. The study of metabolic disorders occurring because of genetic mutations is a subject of core importance in the pathology of children today. The PEX mutations, difficult to identify antepartum, are linked to an array of cell anomalies with severe consequences on the patient’s status, afflicting multiple organs and systems. This is the reason for which our case history may be relevant, including a vast number of symptoms, as well as modified biological parameters.
Collapse
Affiliation(s)
- Ana-Maria Slanina
- Department of Family Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iași 700115, Romania
| | - Adorata-Elena Coman
- Department of Family Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iași 700115, Romania
| | - Dana-Teodora Anton-Păduraru
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iași 700115, Romania
- Correspondence:
| | - Elena Popa
- Department of Family Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iași 700115, Romania
| | - Carmen-Liliana Barbacariu
- Department of Family Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iași 700115, Romania
| | - Otilia Novac
- Department of Family Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iași 700115, Romania
| | - Antoneta Dacia Petroaie
- Department of Family Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iași 700115, Romania
| | - Agnes-Iacinta Bacușcă
- Department of Family Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iași 700115, Romania
| | - Mihaela Manole
- Department of Family Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iași 700115, Romania
| | - Adriana Cosmescu
- Department of Family Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iași 700115, Romania
| |
Collapse
|
7
|
Tanti MJ, Maguire MJ, Warren DJ, Bamford J. Late onset AMACR deficiency with metabolic stroke-like episodes and seizures. BMJ Case Rep 2022; 15:e247964. [PMID: 35428665 PMCID: PMC9014048 DOI: 10.1136/bcr-2021-247964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2022] [Indexed: 11/04/2022] Open
Abstract
Alpha-methylacyl-CoA racemase (AMACR) deficiency is a rare peroxisomal disorder causing pristanic acid accumulation. Only 16 cases have been described so far. A female in her seventh decade presented with episodes of dysphasia, headache and sensory disturbance inconsistent with migraine, epilepsy or transient ischaemic attack. An MRI demonstrated unusual changes in the pons, red nuclei, thalami and white matter. Mitochondrial disease was suspected but detailed testing was negative. After eight years of symptoms, she developed a febrile encephalopathy with hemispheric dysfunction, focal convulsive seizures and coma. Her condition stabilised after one month. Lacosamide was continued for seizure prevention. The diagnosis remained elusive until whole genome sequencing revealed AMACR deficiency. Pristanic acid levels were highly elevated and dietary modification was recommended. Genetic peroxisomal disorders can present in older age; our patient is the oldest in the AMACR deficiency literature. Novel features in our case include central apnoea, dystonia and rapid eye movement behaviour disorder.
Collapse
Affiliation(s)
| | | | | | - John Bamford
- Department of Neurology, Leeds General Infirmary, Leeds, UK
| |
Collapse
|
8
|
Branched-Chain Fatty Acids as Mediators of the Activation of Hepatic Peroxisome Proliferator-Activated Receptor Alpha by a Fungal Lipid Extract. Biomolecules 2020; 10:biom10091259. [PMID: 32878262 PMCID: PMC7565516 DOI: 10.3390/biom10091259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022] Open
Abstract
The study aimed to test the hypothesis that monomethyl branched-chain fatty acids (BCFAs) and a lipid extract of Conidiobolus heterosporus (CHLE), rich in monomethyl BCFAs, are able to activate the nuclear transcription factor peroxisome proliferator-activated receptor alpha (PPARalpha). Rat Fao cells were incubated with the monomethyl BCFAs 12-methyltridecanoic acid (MTriA), 12-methyltetradecanoic acid (MTA), isopalmitic acid (IPA) and 14-methylhexadecanoic acid (MHD), and the direct activation of PPARalpha was evaluated by reporter gene assay using a PPARalpha responsive reporter gene. Furthermore, Fao cells were incubated with different concentrations of the CHLE and PPARalpha activation was also evaluated by using the reporter gene assay, and by determining the mRNA concentrations of selected PPARalpha target genes by real-time RT-PCR. The reporter gene assay revealed that IPA and the CHLE, but not MTriA, MHD and MTA, activate the PPARalpha responsive reporter gene. CHLE dose-dependently increased mRNA concentrations of the PPARalpha target genes acyl-CoA oxidase (ACOX1), cytochrome P450 4A1 (CYP4A1), carnitine palmitoyltransferase 1A (CPT1A) and solute carrier family 22 (organic cation/carnitine transporter), member 5 (SLC22A5). In conclusion, the monomethyl BCFA IPA is a potent PPARalpha activator. CHLE activates PPARalpha-dependent gene expression in Fao cells, an effect that is possibly mediated by IPA.
Collapse
|
9
|
Kim JT, Won SY, Kang K, Kim SH, Park MS, Choi KH, Nam TS, Denis SW, Ferdinandusse S, Lee JE, Choi SY, Kim MK. ACOX3 Dysfunction as a Potential Cause of Recurrent Spontaneous Vasospasm of Internal Carotid Artery. Transl Stroke Res 2020; 11:1041-1051. [PMID: 31975215 DOI: 10.1007/s12975-020-00779-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/30/2019] [Accepted: 01/02/2020] [Indexed: 11/24/2022]
Abstract
Recurrent spontaneous vasospasm of the extracranial internal carotid artery (RSV-eICA) is a rarely recognized cause of ischemic stroke in young adults. However, its pathophysiology remains largely unknown. Through whole-exome sequencing of the ACOX3 gene of two dizygotic Korean twin brothers affected by RSV-eICA, we identified two compound heterozygous missense variants c.235 T > G (p.F79 V) and c.665G > A (p.G222E). In silico analysis indicated that both variants were classified as pathogenic. In vitro ACOX3 enzyme assay indicated practically no enzyme activity in both F79 V and G222E mutants. To determine the effect of the mutants on vasospasm, we used a collagen contraction assay on human aortic smooth muscle cells (HASMC). Carbachol, a cholinergic agonist, induces contraction of HASMC. Knockdown of ACOX3 in HASMC, using siRNA, significantly repressed HASMC contraction triggered by carbachol. The carbachol-induced HASMC contraction was restored by transfection with plasmids encoding siRNA-resistant wild-type ACOX3, but not by transfection with ACOX3 G222E or by co-transfection with ACOX3 F79 V and ACOX3 G222E, indicating that the two ACOX3 mutants suppress carbachol-induced HASMC contraction. We propose that an ACOX3 dysfunction elicits a prolonged loss of the basal aortic myogenic tone. As a result, smooth muscles of the ICA's intermediate segment, in which the sympathetic innervation is especially rich, becomes hypersensitive to sympathomimetic stimuli (e.g., heavy exercise) leading to a recurrent vasospasm. Therefore, ACOX3 dysfunction would be a causal mechanism of RSV-eICA. For the first time, we report the possible involvement of ACOX3 in maintaining the basal myogenic tone of human arterial smooth muscle.
Collapse
Affiliation(s)
- Joon-Tae Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - So Yeon Won
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, South Korea
| | - KyungWook Kang
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Sang-Hoon Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Man-Seok Park
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Kang-Ho Choi
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Tai-Seung Nam
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Simone W Denis
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Sacha Ferdinandusse
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Ji Eun Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, South Korea.
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, 61469, South Korea.
| | - Myeong-Kyu Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea.
| |
Collapse
|
10
|
Phytol and its metabolites phytanic and pristanic acids for risk of cancer: current evidence and future directions. Eur J Cancer Prev 2019; 29:191-200. [PMID: 31436750 PMCID: PMC7012361 DOI: 10.1097/cej.0000000000000534] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This review summarizes the current evidence on the potential role of phytol, a microbial metabolite of chlorophyl A, and its metabolites, phytanic and pristanic acids, in carcinogenesis. Primary food sources in Western diets are the nut skin for phytol and lipids in dairy, beef and fish for its metabolites. Phytol and its metabolites gained interest as dietary compounds for cancer prevention because, as natural ligands of peroxisome proliferator-activated receptor-α and -γ and retinoid X receptor, phytol and its metabolites have provided some evidence in cell culture studies and limited evidence in animal models of anti-carcinogenic, anti-inflammatory and anti-metabolic-syndrome properties at physiological concentrations. However, there may be a narrow range of efficacy, because phytol and its metabolites at supra-physiological concentrations can cause in vitro cytotoxicity in non-cancer cells and can cause morbidity and mortality in animal models. In human studies, evidence for a role of phytol and its metabolites in cancer prevention is currently limited and inconclusive. In short, phytol and its metabolites are potential dietary compounds for cancer prevention, assuming the challenges in preventing cytotoxicity in non-cancer cells and animal models and understanding phytol metabolism can be mitigated.
Collapse
|
11
|
Schmölz L, Wallert M, Rozzino N, Cignarella A, Galli F, Glei M, Werz O, Koeberle A, Birringer M, Lorkowski S. Structure–Function Relationship Studies In Vitro Reveal Distinct and Specific Effects of Long‐Chain Metabolites of Vitamin E. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201700562] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/10/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Lisa Schmölz
- Department of Nutritional Biochemistry and PhysiologyInstitute of NutritionFriedrich Schiller University Jena Jena Germany
- Competence Center for Nutrition and Cardiovascular Health (nutriCARD)Halle‐Jena‐Leipzig
| | - Maria Wallert
- Department of Nutritional Biochemistry and PhysiologyInstitute of NutritionFriedrich Schiller University Jena Jena Germany
- Competence Center for Nutrition and Cardiovascular Health (nutriCARD)Halle‐Jena‐Leipzig
- Baker Heart and Diabetes Institute Melbourne Australia
| | - Nicolò Rozzino
- Department of Pharmaceutical and Pharmacological SciencesUniversity of Padova Padova Italy
| | | | - Francesco Galli
- Department of Pharmaceutical SciencesLaboratory of Nutrition and Clinical BiochemistryUniversity of Perugia Perugia Italy
| | - Michael Glei
- Competence Center for Nutrition and Cardiovascular Health (nutriCARD)Halle‐Jena‐Leipzig
- Department of Nutritional ToxicologyInstitute of NutritionFriedrich Schiller University Jena Jena Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich Schiller University Jena Jena Germany
| | - Andreas Koeberle
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich Schiller University Jena Jena Germany
| | - Marc Birringer
- Department of NutritionalFood and Consumer ScienceUniversity of Applied Sciences Fulda Germany
| | - Stefan Lorkowski
- Department of Nutritional Biochemistry and PhysiologyInstitute of NutritionFriedrich Schiller University Jena Jena Germany
- Competence Center for Nutrition and Cardiovascular Health (nutriCARD)Halle‐Jena‐Leipzig
| |
Collapse
|
12
|
Chaudhary S, Parvez S. Phytanic acid induced neurological alterations in rat brain synaptosomes and its attenuation by melatonin. Biomed Pharmacother 2017; 95:37-46. [PMID: 28826095 DOI: 10.1016/j.biopha.2017.07.156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/21/2017] [Accepted: 07/30/2017] [Indexed: 12/17/2022] Open
Abstract
Phytanic acid (3,7,11,15-tetramethylhexadecanoic acid) (Phyt) is a saturated branched chain fatty acid which originates after the breakdown of chlorophyll molecule, phytol. It plays an important role in a variety of metabolic disorders with peroxisomal impairments. The aim of our investigation was to evaluate the adverse effects of Phyt on synaptic functions by using synaptosomal preparation of rat brain as an in vitro model and the possible protective role of melatonin against Phyt-induced neurotoxicity. Melatonin is an antioxidant, secreted by the pineal gland. Melatonin and its metabolites have neuroprotective effects on cellular stress, by reducing reactive oxygen species (ROS) and reactive nitrogen species (RNS). In the present investigation, synaptosomes prepared from rat brain were co-treated with melatonin (10μM) and Phyt (50μM) for 2h. Co-treatment of Phyt with melatonin significantly restored the altered levels of protein carbonyl (PC) contents and lipid peroxidation (LPO). It also replenished the Phyt-induced alterations on the levels of non-enzymatic antioxidant defence reduced glutathione (GSH), enzymatic antioxidants such as catalase (CAT) and superoxide dismutase (SOD) and synaptosomal integral enzymes such as AChE, Na+, K+-ATPase and MAO. We observed that Phyt induced oxidative stress in synaptosomes as indicated by an elevation in the generation of ROS and melatonin was able to inhibit the elevated ROS generation. Moreover, the neurotoxic effects elicited by Phyt on NO level and membrane potential were totally prevented by the treatment of melatonin. The results of our investigation emphasize the potential use of melatonin as a nutraceutical and mitigatory agent against Phyt-induced oxidative stress.
Collapse
Affiliation(s)
- Shaista Chaudhary
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India.
| |
Collapse
|
13
|
Roca-Saavedra P, Mariño-Lorenzo P, Miranda J, Porto-Arias J, Lamas A, Vazquez B, Franco C, Cepeda A. Phytanic acid consumption and human health, risks, benefits and future trends: A review. Food Chem 2017; 221:237-247. [DOI: 10.1016/j.foodchem.2016.10.074] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/29/2016] [Accepted: 10/18/2016] [Indexed: 12/18/2022]
|
14
|
Schönfeld P, Reiser G. Brain energy metabolism spurns fatty acids as fuel due to their inherent mitotoxicity and potential capacity to unleash neurodegeneration. Neurochem Int 2017; 109:68-77. [PMID: 28366720 DOI: 10.1016/j.neuint.2017.03.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 03/20/2017] [Accepted: 03/24/2017] [Indexed: 12/21/2022]
Abstract
The brain uses long-chain fatty acids (LCFAs) to a negligible extent as fuel for the mitochondrial energy generation, in contrast to other tissues that also demand high energy. Besides this generally accepted view, some studies using cultured neural cells or whole brain indicate a moderately active mitochondrial β-oxidation. Here, we corroborate the conclusion that brain mitochondria are unable to oxidize fatty acids. In contrast, the combustion of liver-derived ketone bodies by neural cells is long-known. Furthermore, new insights indicate the use of odd-numbered medium-chain fatty acids as valuable source for maintaining the level of intermediates of the citric acid cycle in brain mitochondria. Non-esterified LCFAs or their activated forms exert a large variety of harmful side-effects on mitochondria, such as enhancing the mitochondrial ROS generation in distinct steps of the β-oxidation and therefore potentially increasing oxidative stress. Hence, the question arises: Why do in brain energy metabolism mitochondria selectively spurn LCFAs as energy source? The most likely answer are the relatively higher content of peroxidation-sensitive polyunsaturated fatty acids and the low antioxidative defense in brain tissue. There are two remarkable peroxisomal defects, one relating to α-oxidation of phytanic acid and the other to uptake of very long-chain fatty acids (VLCFAs) which lead to pathologically high tissue levels of such fatty acids. Both, the accumulation of phytanic acid and that of VLCFAs give an enlightening insight into harmful activities of fatty acids on neural cells, which possibly explain why evolution has prevented brain mitochondria from the equipment with significant β-oxidation enzymatic capacity.
Collapse
Affiliation(s)
- Peter Schönfeld
- Institut für Biochemie und Zellbiologie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany
| | - Georg Reiser
- Institut für Inflammation und Neurodegeneration (Neurobiochemie), Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany.
| |
Collapse
|
15
|
Phytanic acid attenuates insulin-like growth factor-1 activity via nitric oxide-mediated γ-secretase activation in rat aortic smooth muscle cells: possible implications for pathogenesis of infantile Refsum disease. Pediatr Res 2017; 81:531-536. [PMID: 27886192 DOI: 10.1038/pr.2016.258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 10/09/2016] [Indexed: 01/26/2023]
Abstract
BACKGROUND Infantile Refsum disease (IRD), a peroxisomal disease with defective phytanic acid oxidation, causes neurological impairment and development delay. Insulin-like growth factor-1 (IGF-1) regulates child development and to understand molecular mechanism(s) of IRD, we examined the effect of phytanic acid (PA) on IGF-1 activity. METHODS Bromodeoxyuridine (BrdU) incorporation was measured in rat aortic smooth muscle cell (SMC) cultures following treatment with fetal bovine serum (FBS), basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF) or IGF-1 in the absence or presence of PA. Gene expression and protein contents of IGF-1 receptor (IGF-1R) and PDGF receptor (PDGFR) were examined using quantitative PCR and western blotting. RESULTS PA inhibited mitogenic activities of FBS, PDGF and IGF-1 with more pronounced effect on IGF-1-induced bromodeoxyuridine (BrdU) incorporation. Palmitic acid or lignoceric acids did not inhibit IGF-1 activity. PA had no effect on PDGFR mRNA/protein levels but markedly increased IGF-1R mRNA levels. PA and nitric oxide (NO) markedly decreased IGF-1R protein. L-NAME, a NO synthase inhibitor and DAPT, a γ-secretase inhibitor, alleviated PA-induced decrease in IGF-1R protein. Both PA and NO donor increased γ-secretase activity which was alleviated by L-NAME. CONCLUSION This study demonstrates that PA attenuates IGF-1 activity possibly through IGF-1R impairment and NO-mediated modulation of γ-secretase activity.
Collapse
|
16
|
Phytanic Acid-Induced Neurotoxicological Manifestations and Apoptosis Ameliorated by Mitochondria-Mediated Actions of Melatonin. Mol Neurobiol 2016; 54:6960-6969. [PMID: 27785753 DOI: 10.1007/s12035-016-0209-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/11/2016] [Indexed: 12/27/2022]
Abstract
Phytanic acid, a saturated branched chain fatty acid and a major constituent of human diet, is predominantly found in dairy products, meat, and fish. It is a degradation product from the phytol side chain of chlorophyll. Degradation of PA is known to occur mainly in peroxisomes via α-oxidation and in mitochondria via β-oxidation. Due to its β-methyl group present at the 3-position of the carbon atoms, PA cannot be β-oxidized. Although alteration in the metabolism of PA may play an important role in neurodegeneration, the exact mechanism behind it remains to be evaluated. In this study, we have described the potential of PA to induce neurotoxicity as an in vitro model (neuronal cell line, SH-SY5Y cells). Cells were pretreated with melatonin (10 μM) for 1 h followed by with and without PA (100 μM) for 24 h. In the present study, our data has confirmed that PA markedly increased both intracellular reactive oxygen species and reactive nitrogen species levels. Our results have shown that PA treatment did not induce cell death by cleavage of caspase-3/PARP-1 mediated by mitochondria through intrinsic pathways; however, PA induced nitric oxide-dependent apoptosis in SH-SY5Y cells. Additionally, melatonin pretreatment reduced the cell death in SH-SY5Y cells. Melatonin also effectively exerted an antiapoptotic and anti-inflammatory action by regulating Bax, Bcl-2, p-NFκB, and iNOS expressions in SH-SY5Y cells. These results suggested that melatonin acted as an antioxidative and antiapoptotic agent by modulating ROS, apoptotic proteins, and inflammatory responses under BCFA-induced neurotoxic conditions. The protective effects of melatonin depend on direct scavenging activity of free radicals and indirect antioxidant effects. Further deciphering of the cellular and molecular mechanism associated with neuroprotection by melatonin is warranted in BCFA-induced neurotoxicity.
Collapse
|
17
|
Dhaunsi GS, Alsaeid M, Akhtar S. Phytanic acid activates NADPH oxidase through transactivation of epidermal growth factor receptor in vascular smooth muscle cells. Lipids Health Dis 2016; 15:105. [PMID: 27287039 PMCID: PMC4902935 DOI: 10.1186/s12944-016-0273-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/07/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Phytanic acid (PA) has been implicated in development of cancer and its defective metabolism is known to cause life-threatening conditions, such as Refsum disease, in children. To explore molecular mechanisms of phytanic acid-induced cellular pathology, we investigated its effect on NADPH oxidase (NOX) and epidermal growth factor receptor (EGFR) in rat aortic smooth muscle cells (RASMC). METHODS Smooth muscle cells were isolated from rat aortae using enzymic digestion with collagenase and elastase. Cultured RASMC were treated with varying concentrations (0.5-10 μg/ml) of phytanic acid in the presence/absence of fetal bovine serum (FBS) and/or EGFR inhibitor, AG1478. Following treatment with experimental agents, NOX activity was assayed in RASMC cultures by luminescence method. Gene expression of NOX-1 and p47phox was assessed using RT-PCR. NOX-1, p47phox and, total EGFR protein and its phosphorylated form were measured by Western blotting. RESULTS Treatment of RASMC with supraphysiological concentrations (>2.5 μg/ml) of PA significantly (p < 0.01) increased the NOX activity. PA also significantly increased gene/protein expression of NOX-1 and p47phox whereas p22phox and p67phox remained unaffected. Interestingly, PA (2.5-10 μg/ml) markedly (2-3 folds) increased the total and phosphorylated EGFR. Treatment of cells with EGFR inhibitor, AG1478, significantly blocked the PA-induced enhancement of NOX activity. CONCLUSIONS Our findings that PA transactivates EGFR and induces NOX activity in vascular smooth muscle cells provide new insights into molecular mechanisms of PA's role in cancer and Refsum disease.
Collapse
Affiliation(s)
- Gursev S Dhaunsi
- Departments of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait.
| | - Mayra Alsaeid
- Departments of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Saghir Akhtar
- Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
18
|
Nassi A, Malorgio F, Tedesco S, Cignarella A, Gaion RM. Upregulation of inducible NO synthase by exogenous adenosine in vascular smooth muscle cells activated by inflammatory stimuli in experimental diabetes. Cardiovasc Diabetol 2016; 15:32. [PMID: 26879172 PMCID: PMC4754884 DOI: 10.1186/s12933-016-0349-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/02/2016] [Indexed: 01/19/2023] Open
Abstract
Background Adenosine has been shown to induce nitric oxide (NO) production via inducible NO synthase (iNOS) activation in vascular smooth muscle cells (VSMCs). Although this is interpreted as a beneficial vasodilating pathway in vaso-occlusive disorders, iNOS is also involved in diabetic vascular dysfunction. Because the turnover of and the potential to modulate iNOS by adenosine in experimental diabetes have not been explored, we hypothesized that both the adenosine system and control of iNOS function are impaired in VSMCs from streptozotocin-diabetic rats. Methods Male Sprague–Dawley rats were injected with streptozotocin once to induce diabetes. Aortic VSMCs from diabetic and nondiabetic rats were isolated, cultured and exposed to lipopolysaccharide (LPS) plus a cytokine mix for 24 h in the presence or absence of (1) exogenous adenosine and related compounds, and/or (2) pharmacological agents affecting adenosine turnover. iNOS functional expression was determined by immunoblotting and NO metabolite assays. Concentrations of adenosine, related compounds and metabolites thereof were assayed by HPLC. Vasomotor responses to adenosine were determined in endothelium-deprived aortic rings. Results Treatment with adenosine-degrading enzymes or receptor antagonists increased iNOS formation in activated VSMCs from nondiabetic and diabetic rats. Following treatment with the adenosine transport inhibitor NBTI, iNOS levels increased in nondiabetic but decreased in diabetic VSMCs. The amount of secreted NO metabolites was uncoupled from iNOS levels in diabetic VSMCs. Addition of high concentrations of adenosine and its precursors or analogues enhanced iNOS formation solely in diabetic VSMCs. Exogenous adenosine and AMP were completely removed from the culture medium and converted into metabolites. A tendency towards elevated inosine generation was observed in diabetic VSMCs, which were also less sensitive to CD73 inhibition, but inosine supplementation did not affect iNOS levels. Pharmacological inhibition of NOS abolished adenosine-induced vasorelaxation in aortic tissues from diabetic but not nondiabetic animals. Conclusions Endogenous adenosine prevented cytokine- and LPS-induced iNOS activation in VSMCs. By contrast, supplementation with adenosine and its precursors or analogues enhanced iNOS levels in diabetic VSMCs. This effect was associated with alterations in exogenous adenosine turnover. Thus, overactivation of the adenosine system may foster iNOS-mediated diabetic vascular dysfunction.
Collapse
Affiliation(s)
- Alberto Nassi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy. .,Transplant Immunology Unit, Padua University Hospital, Padova, Italy.
| | - Francesca Malorgio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.
| | - Serena Tedesco
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.
| | | | - Rosa Maria Gaion
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.
| |
Collapse
|
19
|
Borges CG, Canani CR, Fernandes CG, Zanatta Â, Seminotti B, Ribeiro CAJ, Leipnitz G, Vargas CR, Wajner M. Reactive nitrogen species mediate oxidative stress and astrogliosis provoked by in vivo administration of phytanic acid in cerebellum of adolescent rats: A potential contributing pathomechanism of cerebellar injury in peroxisomal disorders. Neuroscience 2015; 304:122-32. [PMID: 26188285 DOI: 10.1016/j.neuroscience.2015.07.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/25/2015] [Accepted: 07/09/2015] [Indexed: 01/10/2023]
Abstract
Phytanic acid (Phyt) accumulates in various peroxisomal diseases including Refsum disease (RD) and Zellweger syndrome (ZS). Since the pathogenesis of the neurological symptoms and especially the cerebellar abnormalities in these disorders are poorly known, we investigated the effects of in vivo intracerebral administration of Phyt on a large spectrum of redox homeostasis parameters in the cerebellum of young rats. Malondialdehyde (MDA) levels, sulfhydryl oxidation, carbonyl content, nitrite and nitrate concentrations, 2',7'-dichlorofluorescein (DCFH) oxidation, total (tGS) and reduced glutathione (GSH) levels and the activities of important antioxidant enzymes were determined at different periods after Phyt administration. Immunohistochemical analysis was also carried out in the cerebellum. Phyt significantly increased MDA and nitric oxide (NO) production and decreased GSH levels, without altering tGS, DCFH oxidation, sulfhydryl oxidation, carbonyl content and the activities of glutathione peroxidase (GPx), superoxide dismutase (SOD), catalase (CAT), glutathione reductase (GR) and glucose-6-phosphate dehydrogenase (G6PD). Furthermore, immunohistochemical analysis revealed that Phyt caused astrogliosis and protein nitrosative damage in the cerebellum. It was also observed that the NO synthase inhibitor Nω-Nitro-L-arginine methyl ester (L-NAME) prevented the increase of MDA and NO production as well as the decrease of GSH and the immunohistochemical alterations caused by Phyt, strongly suggesting that reactive nitrogen species (RNS) were involved in these effects. The present data provide in vivo solid evidence that Phyt disrupts redox homeostasis and causes astrogliosis in rat cerebellum probably mediated by RNS production. It is therefore presumed that disequilibrium of redox status may contribute at least in part to the cerebellum alterations characteristic of patients affected by RD and other disorders with Phyt accumulation.
Collapse
Affiliation(s)
- C G Borges
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - C R Canani
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - C G Fernandes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Â Zanatta
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - B Seminotti
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - C A J Ribeiro
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - G Leipnitz
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - C R Vargas
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Departamento de Análises, Faculdade de Farmácia, UFRGS, Porto Alegre, RS, Brazil
| | - M Wajner
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| |
Collapse
|
20
|
Stewart MW, Vavra MW, Whaley NR. FUNDUS FINDINGS IN A PATIENT WITH α-METHLYACYL-COA RACEMASE DEFICIENCY. Retin Cases Brief Rep 2014; 5:262-6. [PMID: 25390181 DOI: 10.1097/icb.0b013e3181f047dd] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE To describe the ocular findings in a patient with α-methylacyl-CoA racemase deficiency. METHODS Case report. RESULTS A 45-year-old white man presented with seizures, hemiparesis, and altered mental status. As a part of an extensive investigation, an ophthalmic evaluation was performed. Funduscopic examination of both eyes showed a bull's-eye pattern pigmentary maculopathy. Fluorescein angiography demonstrated a prominent choroidal circulation without dye leakage. Optical coherence tomography showed a significant thinning of the macula with moderate nerve fiber layer loss. Serum pristanic levels were significantly elevated, and α-methylacyl-CoA racemase deficiency was confirmed by urine and bile analysis. The patient's clinical condition improved after plasma exchange and dietary restriction of fatty acids. CONCLUSION α-Methylacyl-CoA racemase deficiency is a rare peroxisomal enzyme disorder previously described in only six patients, two of whom had pigmentary retinopathies. This article describes the third patient with retinopathy and the first with published fundus findings.
Collapse
Affiliation(s)
- Michael W Stewart
- From the Departments of *Ophthalmology and †Neurology, Mayo School of Medicine, Jacksonville, Florida
| | | | | |
Collapse
|
21
|
Disruption of oxidative phosphorylation and synaptic Na(+), K(+)-ATPase activity by pristanic acid in cerebellum of young rats. Life Sci 2014; 94:67-73. [PMID: 24211616 DOI: 10.1016/j.lfs.2013.10.032] [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: 07/17/2013] [Revised: 10/04/2013] [Accepted: 10/28/2013] [Indexed: 11/24/2022]
Abstract
AIMS Peroxisomal biogenesis disorders (PBD) are inherited disorders clinically manifested by neurological symptoms and brain abnormalities, in which the cerebellum is usually involved. Biochemically, patients affected by these neurodegenerative diseases accumulate branched-chain fatty acids, including pristanic acid (Prist) in the brain and other tissues. MAIN METHODS In the present investigation we studied the in vitro influence of Prist, at doses found in PBD, on oxidative phosphorylation, by measuring the activities of the respiratory chain complexes I-IV and ATP production, as well as on creatine kinase and synaptic Na(+), K(+)-ATPase activities in rat cerebellum. KEY FINDINGS Prist significantly decreased complexes I-III (65%), II (40%) and especially II-III (90%) activities, without altering the activities of complex IV of the respiratory chain and creatine kinase. Furthermore, ATP formation and synaptic Na(+), K(+)-ATPase activity were markedly inhibited (80-90%) by Prist. We also observed that this fatty acid altered mitochondrial and synaptic membrane fluidity that may have contributed to its inhibitory effects on the activities of the respiratory chain complexes and Na(+), K(+)-ATPase. SIGNIFICANCE Considering the importance of oxidative phosphorylation for mitochondrial homeostasis and of Na(+), K(+)-ATPase for the maintenance of cell membrane potential, the present data indicate that Prist compromises brain bioenergetics and neurotransmission in cerebellum. We postulate that these pathomechanisms may contribute to the cerebellar alterations observed in patients affected by PBD in which Prist is accumulated.
Collapse
|
22
|
Disruption of mitochondrial homeostasis by phytanic acid in cerebellum of young rats. THE CEREBELLUM 2013; 12:362-9. [PMID: 23081695 DOI: 10.1007/s12311-012-0426-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Phytanic acid (Phyt) brain concentrations are highly increased in Refsum disease, a peroxisomal disorder clinically characterized by neurological features, cardiac abnormalities, and retinitis pigmentosa. Considering that the pathogenesis of cerebellar ataxia, a common finding in this disease, is still unknown, in the present work we investigated the in vitro effects of Phyt at concentrations similar to those found in affected patients on important parameters of mitochondrial homeostasis in cerebellum from young rats. The respiratory parameters states 3 and 4 and respiratory control ratio (RCR) determined by oxygen consumption, membrane potential (∆Ψm), NAD(P)H pool content, and swelling were evaluated in mitochondrial preparations from this cerebral structure. Phyt markedly increased state 4 respiration, whereas state 3 respiration, the RCR, the mitochondrial matrix NAD(P)H content, and ∆Ψm were decreased by this fatty acid, being the latter effect partially prevented by N-acetylcysteine. These data indicate that Phyt behaves as an uncoupler of oxidative phosphorylation and as a metabolic inhibitor disrupting mitochondrial homeostasis in cerebellum. It is proposed that these pathomechanisms may contribute at least in part to the cerebellar alterations found in Refsum disease.
Collapse
|
23
|
Toniolo A, Warden EA, Nassi A, Cignarella A, Bolego C. Regulation of SIRT1 in vascular smooth muscle cells from streptozotocin-diabetic rats. PLoS One 2013; 8:e65666. [PMID: 23734259 PMCID: PMC3667130 DOI: 10.1371/journal.pone.0065666] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 05/02/2013] [Indexed: 12/15/2022] Open
Abstract
Sirtuins enzymes are a conserved family of nicotinamide adenine dinucleotide (NAD)-dependent deacetylases and ADP-ribosyltransferases that mediate responses to oxidative stress, fasting and dietary restriction in mammals. Vascular smooth muscle cells (VSMCs) are involved in many mechanisms that regulate vascular biology in vivo but the role of SIRT1 has not been explored in much detail. Therefore, we investigated the regulation of SIRT1 in cultured VSMCs under various stress conditions including diabetes. Sprague-Dawley rats were made diabetic by injecting a single dose of streptozotocin (65 mg/Kg), and aortic VSMCs were isolated after 4 weeks. Immunocytochemistry showed that SIRT1 was localized predominantly in the nucleus, with lower staining in VSMCs from STZ-diabetic as compared with normoglycemic rats. Previous diabetes induction in vivo and high glucose concentrations in vitro significantly downregulated SIRT1 amounts as detected in Western blot assays, whereas TNF-α (30 ng/ml) stimulation failed to induce significant changes. Because estrogen signaling affects several pathways of oxidative stress control, we also investigated SIRT1 modulation by 17β-estradiol. Treatment with the hormone (10 nM) or a selective estrogen receptor-α agonist decreased SIRT1 levels in VSMCs from normoglycemic but not in those from STZ-diabetic animals. 17β-estradiol treatment also enhanced activation of AMP-dependent kinase, which partners with SIRT1 in a signaling axis. SIRT1 downregulation by 17β-estradiol could be observed as well in human peripheral blood mononuclear cells, a cell type in which SIRT1 downregulation is associated with insulin resistance and subclinical atherosclerosis. These data suggest that SIRT1 protein levels are regulated by diverse cellular stressors to a variable extent in VSMCs from diabetic and normoglycemic rats, warranting further investigation on SIRT1 as a modulator of VSMC activity in settings of vascular inflammation.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Nucleus/enzymology
- Cells, Cultured
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/pathology
- Down-Regulation/drug effects
- Estradiol/pharmacology
- Estrogens/pharmacology
- Glucose/pharmacology
- Humans
- Immunohistochemistry
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/enzymology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Rats
- Rats, Sprague-Dawley
- Sirtuin 1/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Alice Toniolo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Erica Alessia Warden
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Alberto Nassi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Andrea Cignarella
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- * E-mail:
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
24
|
Busanello ENB, Zanatta Â, Tonin AM, Viegas CM, Vargas CR, Leipnitz G, Ribeiro CAJ, Wajner M. Marked inhibition of Na+, K(+)- ATPase activity and the respiratory chain by phytanic acid in cerebellum from young rats: possible underlying mechanisms of cerebellar ataxia in Refsum disease. J Bioenerg Biomembr 2012; 45:137-44. [PMID: 23151916 DOI: 10.1007/s10863-012-9491-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 11/05/2012] [Indexed: 11/28/2022]
Abstract
Refsum disease is an autosomal recessive disorder of peroxisomal metabolism biochemically characterized by highly elevated concentrations of phytanic acid (Phyt) in a variety of tissues including the cerebellum. Reduction of plasma Phyt levels by dietary restriction intake ameliorates ataxia, a common clinical manifestation of this disorder, suggesting a neurotoxic role for this branched-chain fatty acid. Therefore, considering that the underlying mechanisms of cerebellum damage in Refsum disease are poorly known, in the present study we tested the effects of Phyt on important parameters of bioenergetics, such as the activities of the respiratory chain complexes I to IV, creatine kinase and Na(+), K(+)- ATPase in cerebellum preparations from young rats. The activities of complexes I, II, I-III and II-III and Na(+), K(+)- ATPase were markedly inhibited (65-85%) in a dose-dependent manner by Phyt. In contrast, creatine kinase and complex IV activities were not altered by this fatty acid. Therefore, it is presumed that impairment of the electron flow through the respiratory chain and inhibition of Na(+), K(+)- ATPase that is crucial for synaptic function may be involved in the pathophysiology of the cerebellar abnormalities manifested as ataxia in Refsum disease and in other peroxisomal disorders in which brain Phyt accumulates.
Collapse
Affiliation(s)
- Estela Natacha Brandt Busanello
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600 - Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Lin T, Yin X, Cai Q, Fan X, Xu K, Huang L, Luo J, Zheng J, Huang J. 13-Methyltetradecanoic acid induces mitochondrial-mediated apoptosis in human bladder cancer cells. Urol Oncol 2012; 30:339-45. [PMID: 20843711 DOI: 10.1016/j.urolonc.2010.04.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 04/27/2010] [Accepted: 04/27/2010] [Indexed: 10/19/2022]
Abstract
OBJECTIVE 13-Methyltetradecanoic acid (13-MTD), a saturated branched-chain fatty acid purified from soy fermentation products, is known to induce apoptosis in many types of human cancer cells. This study was designed to investigate the molecular mechanisms involved in 13-MTD-induced apoptosis in human bladder cancer cells. METHODS AND MATERIALS MTT assay was used to investigate the potential effects of 13-MTD on the growth and viability of human bladder cancer cells. To find out whether anti-proliferation and cell death were associated with apoptosis, we used flow cytometry to quantify the extent of apoptosis and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay to measures DNA degradation of apoptotic cells. The proteins involved in the 13-MTD induced apoptosis were examined using Western blot. RESULTS We show that 13-MTD inhibits cellular proliferation and viability in human bladder cancer cells, which has been attributed to apoptosis. 13-MTD down-regulates Bcl-2 and up-regulates Bax. This promotes mitochondrial dysfunction, leading to the release of cytochrome c from the mitochondria to the cytoplasm, as well as the proteolytic activation of caspases. Moreover, 13-MTD down-regulates AKT phosphorylation and activates phosphorylation of p38 and c-Jun N-terminal kinase (JNK). Up-regulating AKT phosphorylation and down-regulating JNK and P38 phosphorylation could attenuate the13-MTD-induced apoptosis. CONCLUSION Taken together, these data indicate that 13-MTD induces mitochondrial-mediated apoptosis through regulation of the AKT and MAPK pathways, suggesting 13-MTD is a potential candidate for treatment of human bladder cancer.
Collapse
Affiliation(s)
- Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Busanello ENB, Amaral AU, Tonin AM, Grings M, Moura AP, Eichler P, Vargas CR, Wajner M. Experimental evidence that pristanic acid disrupts mitochondrial homeostasis in brain of young rats. J Neurosci Res 2011; 90:597-605. [DOI: 10.1002/jnr.22802] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 08/17/2011] [Accepted: 08/25/2011] [Indexed: 11/10/2022]
|
27
|
Kruska N, Reiser G. Phytanic acid and pristanic acid, branched-chain fatty acids associated with Refsum disease and other inherited peroxisomal disorders, mediate intracellular Ca2+ signaling through activation of free fatty acid receptor GPR40. Neurobiol Dis 2011; 43:465-72. [PMID: 21570468 DOI: 10.1016/j.nbd.2011.04.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 04/18/2011] [Accepted: 04/22/2011] [Indexed: 01/28/2023] Open
Abstract
The accumulation of the two branched-chain fatty acids phytanic acid and pristanic acid is known to play an important role in several diseases with peroxisomal impairment, like Refsum disease, Zellweger syndrome and α-methylacyl-CoA racemase deficiency. Recent studies elucidated that the toxic activity of phytanic acid and pristanic acid is mediated by multiple mitochondrial dysfunctions, generation of reactive oxygen species and Ca2+ deregulation via the InsP3-Ca2+ signaling pathway in glial cells. However, the exact signaling mechanism through which both fatty acids mediate toxicity is still under debate. Here, we studied the ability of phytanic acid and pristanic acid to activate the free fatty acid receptor GPR40, a G-protein-coupled receptor, which was described to be involved in the Ca2+ signaling of fatty acids. We treated HEK 293 cells expressing the GPR40 receptor with phytanic acid or pristanic acid. This resulted in a significant increase in the intracellular Ca2+ level, similar to the effect seen after treatment with the synthetic GPR40 agonist GW9508. Furthermore, we demonstrate that the GPR40 activation might be due to an interaction of the carboxylate moiety of fatty acids with the receptor. Our findings indicate that the phytanic acid- and pristanic acid-mediated Ca2+ deregulation can involve the activation of GPR40. Therefore, we suppose that activation of GPR40 might be part of the signaling cascade of the toxicity of phytanic and pristanic acids.
Collapse
Affiliation(s)
- Nicol Kruska
- Institut für Neurobiochemie, Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | | |
Collapse
|
28
|
Busanello ENB, Viegas CM, Tonin AM, Grings M, Moura AP, de Oliveira AB, Eichler P, Wajner M. Neurochemical Evidence that Pristanic Acid Impairs Energy Production and Inhibits Synaptic Na+, K+-ATPase Activity in Brain of Young Rats. Neurochem Res 2011; 36:1101-7. [DOI: 10.1007/s11064-011-0453-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2011] [Indexed: 11/30/2022]
|
29
|
Perera NJ, Lewis B, Tran H, Fietz M, Sullivan DR. Refsum's Disease-Use of the Intestinal Lipase Inhibitor, Orlistat, as a Novel Therapeutic Approach to a Complex Disorder. J Obes 2011; 2011:482021. [PMID: 20871815 PMCID: PMC2943115 DOI: 10.1155/2011/482021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 07/12/2010] [Indexed: 11/23/2022] Open
Abstract
Refsum's Disease is an inherited metabolic disorder in which a metabolite of branched chain fatty acids accumulates due to lack of appropriate oxidative enzymes. Patients have elevated plasma phytanic acid levels and high concentrations of phytanic acid in a variety of tissues leading to progressive tissue damage. Besides retinal degeneration or retinal dystrophy associated with adult onset retinitis pigmentosa, additional symptoms include chronic polyneuropathy, cerebellar ataxia, sensorineural hearing loss, anosmia, ichthyosis, as well as skeletal, cardiac, hepatic, and renal abnormalities. Current management includes avoidance of dietary sources of branched chain fatty acids and regular plasmapheresis to prevent accumulation of these compounds to ameliorate progressive neurological deficits. Two brothers with Refsum's disease who experienced progressive symptoms despite optimal diet and plasmapheresis were commenced on a novel therapy. We report the effect of the intestinal lipase inhibitor, Orlistat, which led to significant reduction (P-value <0.001 on 2-sample unpaired t-test) of mean preplasmapheresis phytanic acid levels with retardation of the progression of most of their dermatological and neurological symptoms.
Collapse
Affiliation(s)
- Nimalie J. Perera
- Department of Clinical Biochemistry, Royal Prince Alfred Hospital, Sydney, Camperdown, NSW 2050, Australia
| | - Barry Lewis
- Department of Medicine, University of Sydney, NSW 2006, Australia
| | - Huy Tran
- Department of Biochemistry, John Hunter Hospital, Newcastle, NSW 2305, Australia
| | - Michael Fietz
- National Referral Laboratory for Lysosomal, Peroxisomal and Related Genetic Disorders, SA Pathology at The Women's and Children's Hospital, North Adelaide, SA 5006, Australia
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA 5005, Australia
| | - David R. Sullivan
- Department of Clinical Biochemistry, Royal Prince Alfred Hospital, Sydney, Camperdown, NSW 2050, Australia
- *David R. Sullivan:
| |
Collapse
|
30
|
Barranco I, Gómez-Laguna J, Rodríguez-Gómez IM, Salguero FJ, Pallarés FJ, Bernabé A, Carrasco L. Immunohistochemical detection of extrinsic and intrinsic mediators of apoptosis in porcine paraffin-embedded tissues. Vet Immunol Immunopathol 2010; 139:210-6. [PMID: 21074278 DOI: 10.1016/j.vetimm.2010.10.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2010] [Revised: 09/07/2010] [Accepted: 10/14/2010] [Indexed: 10/18/2022]
Abstract
Apoptosis is a strictly regulated mechanism of cell death that involves a complex network of biochemical pathways. Whether a cell undergoes apoptosis or not depends on a delicate balance of anti- and pro-apoptotic stimuli. This phenomenon can be induced by two different pathways: intrinsic and extrinsic pathways. The main aim of this study was to determine the ideal fixative and antigen retrieval method in porcine paraffin embedded tissues for the immunohistochemical detection of apoptosis mediators, from both extrinsic and intrinsic pathways. Tonsil, retropharyngeal lymph node and lung tissue samples were fixed in 10% neutral buffered formalin, Bouin solution and zinc salts fixative (ZSF) and different unmasking methods were carried out. Both 10% neutral buffered formalin and ZSF resulted as the fixatives of election to study apoptosis phenomena. Tween 20 (0.01% in PBS), citrate buffer (microwave, pH 6.0) and/or protease type XIV were the antigen retrieval methods which displayed better labelling. Our results allow to deep in the knowledge of apoptosis and its role in the pathogenesis of porcine diseases.
Collapse
Affiliation(s)
- Inmaculada Barranco
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Cordoba University, 14014 Cordoba, Spain
| | | | | | | | | | | | | |
Collapse
|
31
|
In vitro evidence that phytanic acid compromises Na(+),K(+)-ATPase activity and the electron flow through the respiratory chain in brain cortex from young rats. Brain Res 2010; 1352:231-8. [PMID: 20624373 DOI: 10.1016/j.brainres.2010.07.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 06/29/2010] [Accepted: 07/05/2010] [Indexed: 11/19/2022]
Abstract
Phytanic acid (Phyt) tissue concentrations are increased in Refsum disease and other peroxisomal disorders characterized by neurologic damage and brain abnormalities. The present work investigated the in vitro effects of Phyt, at concentrations found in these peroxisomal disorders, on important parameters of energy metabolism in brain cortex of young rats. The parameters analyzed were CO(2) production from labeled acetate and glucose, the activities of the citric acid cycle enzymes citrate synthase, aconitase, isocitrate dehydrogenase, alpha-ketoglutarate dehydrogenase, succinate dehydrogenase, fumarase and malate dehydrogenase, as well as of the respiratory chain complexes I-IV, creatine kinase and Na(+),K(+)-ATPase. Our results show that Phyt did not alter citric acid cycle enzyme activities, or CO(2) production from acetate, reflecting no impairment of the functionality of the citric acid cycle. In contrast, respiratory chain activities were reduced at complexes I, II, I-III, II-III and IV. Membrane synaptical Na(+),K(+)-ATPase activity was also reduced by Phyt, with no alteration of creatine kinase activity. Considering the importance of the electron flow through the respiratory chain for brain energy metabolism (oxidative phosphorylation) and of Na(+),K(+)-ATPase activity for maintaining membrane potential necessary for neurotransmission, the data indicate that Phyt impairs brain bioenergetics at the level of energy formation, as well as neurotransmission. It is presumed that Phyt-induced impairment of these important systems may be involved at least in part in the neurological damage found in patients affected by disorders in which brain Phyt concentrations are increased.
Collapse
|
32
|
Neurochemical evidence that phytanic acid induces oxidative damage and reduces the antioxidant defenses in cerebellum and cerebral cortex of rats. Life Sci 2010; 87:275-80. [PMID: 20619275 DOI: 10.1016/j.lfs.2010.06.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 06/02/2010] [Accepted: 06/23/2010] [Indexed: 11/21/2022]
Abstract
AIMS In the present work we investigated the in vitro effects of phytanic acid (Phyt), that accumulates in Refsum disease and other peroxisomal diseases, on important parameters of oxidative stress in cerebellum and cerebral cortex from young rats. MAIN METHODS The parameters thiobarbituric acid-reactive substances levels (TBA-RS; lipid peroxidation), carbonyl formation and sulfhydryl oxidation (protein oxidative damage) and the concentrations of the most important nonenzymatic antioxidant defense reduced glutathione (GSH) were determined. KEY FINDINGS It was observed that Phyt significantly increased TBA-RS levels in both cerebral structures. This effect was prevented by the antioxidants alpha-tocopherol and melatonin, suggesting the involvement of free radicals. Phyt also provoked protein oxidative damage in both cerebellum and cerebral cortex, as determined by increased carbonyl content and sulfhydryl oxidation. Furthermore, Phyt significantly diminished the concentrations of GSH, while melatonin and alpha-tocopherol treatment totally blocked this effect. We also verified that Phyt does not behave as a direct acting oxidant, since Phyt did not oxidize commercial solutions of GSH and reduced cytochrome c to Phyt in a free cell medium. SIGNIFICANCE Our data indicate that oxidative stress is elicited in vitro by Phyt, a mechanism that may contribute at least in part to the pathophysiology of Refsum disease and other peroxisomal disorders where Phyt is accumulated.
Collapse
|
33
|
Peroxisomes, lipid metabolism and lipotoxicity. Biochim Biophys Acta Mol Cell Biol Lipids 2010; 1801:272-80. [DOI: 10.1016/j.bbalip.2010.01.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 01/07/2010] [Accepted: 01/07/2010] [Indexed: 12/26/2022]
|
34
|
Rönicke S, Kruska N, Kahlert S, Reiser G. The influence of the branched-chain fatty acids pristanic acid and Refsum disease-associated phytanic acid on mitochondrial functions and calcium regulation of hippocampal neurons, astrocytes, and oligodendrocytes. Neurobiol Dis 2009; 36:401-10. [DOI: 10.1016/j.nbd.2009.08.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 08/09/2009] [Accepted: 08/14/2009] [Indexed: 10/20/2022] Open
|
35
|
Li X, Chen BD. Histone Deacetylase Inhibitor M344 Inhibits Cell Proliferation and Induces Apoptosis in Human THP-1 Leukemia Cells. AMERICAN JOURNAL OF BIOMEDICAL SCIENCES 2009; 1:352-363. [PMID: 20526416 PMCID: PMC2880493 DOI: 10.5099/aj090400352] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Histone acetylation plays an important role in the silencing and activation of genes involved in tumoregenesis. Trichostatin A, originally identified as an anti-fungal drug, is a potent inhibitor of histone deacetylase (HDAC) with potential anti-tumor activity. In this study, we investigated the effect of M344, an amide analogues of trichostatin A, on the growth and differentiation of THP-1 human leukemia cells. We showed that at low doses, (< 0.2 muM), M344 could inhibit the growth of THP-1 cells at G1 phase in vitro with low cytotoxic effect. Low dose of M344 exerted some differentiating effect on THP-1 cells as judged by the expression of c-fms proto-oncogene (M-CSF receptor) and appearance of adherent cells. Growth arrest induced by M344 is associated with increased levels of cyclin-dependent protein kinase inhibitor p21 and cyclin E, in agreement with G1 phase arrest. At higher doses (2 muM), M344 could induce THP-1 cells to undergo apoptosis, which was associated with the cleavage of PARP, cytochrome c release and activation of both caspases-8, -9, followed by the activation of caspase-3. In addition, M344 could increase the levels of pro-apoptotic protein Bax but decreased the levels of anti-apoptotic protein XIAP. M344 is a potent activator of NF-kappaB transcription factor. RT-PCR assay showed that the M344 could transiently increase IL-1 expression yet markedly decreased TNF-alpha expression. Our results show that M344 is a potent growth inhibitor and inducer of apoptosis in human leukemia cells and suggest potential therapeutic strategies of HDAC inhibitors for patients with leukemias.
Collapse
Affiliation(s)
| | - Ben D. Chen
- Department of Internal medicine and Karmanos Cancer Institute, Wayne State University School of Medicine, 550 E. Canfield, Detroit, MI 48201
| |
Collapse
|
36
|
Thompson SA, Calvin J, Hogg S, Ferdinandusse S, Wanders RJA, Barker RA. Relapsing encephalopathy in a patient with α-methylacyl-CoA racemase deficiency. BMJ Case Rep 2009; 2009:bcr08.2008.0814. [PMID: 21686617 DOI: 10.1136/bcr.08.2008.0814] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
α-Methylacyl-CoA racemase (AMACR) deficiency is a rare disorder of fatty acid metabolism which has recently been described in three adult cases. We have identified a further patient with clinical features of a relapsing encephalopathy, seizures and cognitive decline over a 40 year period. Biochemical studies revealed grossly elevated plasma pristanic acid levels, and a deficiency of AMACR in skin fibroblasts. Sequence analysis of AMACR cDNA identified a homozygous point mutation (c154T>C). This case adds to the phenotypic variation seen in this peroxisomal disorder and highlights the importance of screening for plasma pristanic acid levels in patients with unexplained relapsing encephalopathies.
Collapse
Affiliation(s)
- Sian A Thompson
- Addenbrooke's Hospital, Department of Neurology, Hills Road, Cambridge, CB2 0QQ, UK
| | | | | | | | | | | |
Collapse
|
37
|
Cignarella A, Bolego C, Pelosi V, Meda C, Krust A, Pinna C, Gaion RM, Vegeto E, Maggi A. Distinct roles of estrogen receptor-alpha and beta in the modulation of vascular inducible nitric-oxide synthase in diabetes. J Pharmacol Exp Ther 2009; 328:174-82. [PMID: 18832649 PMCID: PMC2685905 DOI: 10.1124/jpet.108.143511] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Accepted: 10/01/2008] [Indexed: 12/15/2022] Open
Abstract
Estrogen is known to affect vascular function and diabetes development, but the relative contribution of estrogen receptor (ER) isoforms is unclear. The aim of this study was to determine how individual ER isoforms modulate inflammatory enzymes in the vascular wall of control and streptozotocin (STZ)-injected rodents. Primary cultures of rat aortic smooth muscle cells (SMCs) were stimulated with inflammatory agents in the presence or absence of increasing concentrations of the ER alpha and ER beta-selective agonists 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT) and diarylpropionitrile (DPN), respectively. The production of inducible nitric-oxide synthase (iNOS), a classical indicator of vascular inflammation, was significantly reduced by PPT in control but not diabetic SMCs, whereas it was further enhanced by DPN treatment in both groups. This distinct action profile was not related to changes in ER transcriptional activity. However, extracellular signal-regulated kinase 1/2 signaling was activated by DPN but not by PPT in cytokine-treated SMCs. In cultured aortic rings from both normoglycemic and STZ-diabetic mice, pharmacological activation of ER alpha attenuated cytokine-driven iNOS induction by 30 to 50%. Vascular iNOS levels were decreased consistently when adding 1 nM 17beta-estradiol to aortic tissues from ER beta- but not ER alpha-knockout mice. These findings suggest a possible role for ER alpha-selective ligands in reducing vascular inflammatory responses under normo- and hyperglycemic conditions.
Collapse
Affiliation(s)
- Andrea Cignarella
- Department of Pharmacology and Anaesthesiology, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Artwohl M, Lindenmair A, Roden M, Waldhäusl WK, Freudenthaler A, Klosner G, Ilhan A, Luger A, Baumgartner-Parzer SM. Fatty acids induce apoptosis in human smooth muscle cells depending on chain length, saturation, and duration of exposure. Atherosclerosis 2008; 202:351-62. [PMID: 18597758 DOI: 10.1016/j.atherosclerosis.2008.05.030] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 05/04/2008] [Accepted: 05/06/2008] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Plasma free fatty acid (FFA) concentrations are increased in states of insulin resistance. Therefore, this study evaluated apoptosis and underlying mechanisms induced by selected nutritional FFAs, a defined FFA-mix, and human plasma containing high FFA concentrations in human smooth muscle cells (HSMCs). RESEARCH DESIGN AND METHODS HSMCs were incubated (24-72 h) with selected FFAs (100-300 micromol/l), an FFA-mix (palmitic-/stearic-/oleic-/linoleic-/alpha-linolenic acid=2.6/1/3.6/9/1; 300-900 micromol/l), or with high FFA-plasma (600 micromol/l) versus respective control cultures. Apoptosis, caspase activation, and protein expression were determined by DNA-fragmentation assays, flow cytometry, and Western blots, respectively. RESULTS Exposure (24h) of HSMCs to 300 micromol/l stearic-, oleic-, linoleic-, alpha-linolenic-, and arachidonic acid induced apoptosis, correlating (p<0.01) with the FFAs' chain length (r=0.602) and number of FFA double bonds (r=0.956). After 48 h, 100 micromol/l of all tested FFAs - including palmitic acid - were already sufficient to trigger HSMCs' cell death. FFA-exposure resulted in activation of caspases and apoptosis was completely abolished by co-incubation with caspase inhibitors and negatively correlated (p<0.01) with the base-excision repair protein XRCC1 (r=-0.765) and with c-myc's antagonist mad (r=-0.916), whereas positive correlations (p<0.01) were found for protein expression of the proto-oncogene c-myc (r=0.972) and the transcription factor E2F-1 (r=0.971). Exposure of HSMCs to the defined FFA-mix and to plasma samples from individuals with elevated plasma FFAs supported the results obtained by defined FFA stimulation. CONCLUSIONS Since smooth muscle cells surround the macrophage/foam cell/lipid-laden artheromatous core of atherosclerotic lesions with a protective fibrous cap, their FFA-induced HSMC apoptosis could contribute to progression of atherosclerosis by thinning of the fibrous cap and subsequent plaque destabilization.
Collapse
Affiliation(s)
- Michaela Artwohl
- Department of Internal Medicine III, Clinical Division of Endocrinology and Metabolism, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Pinna C, Cignarella A, Sanvito P, Pelosi V, Bolego C. Prolonged Ovarian Hormone Deprivation Impairs the Protective Vascular Actions of Estrogen Receptor αAgonists. Hypertension 2008; 51:1210-7. [DOI: 10.1161/hypertensionaha.107.106807] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The vascular consequences of estrogen treatment may be driven by its initiation timing. We tested the hypothesis that the duration of ovarian hormone deprivation before estrogen reintroduction affects the role of estrogen as mediator of endothelial function and vascular relaxation in nondiseased vessels. Rats were ovariectomized and implanted with 17β-estradiol (E
2
) or oil capsules 1, 4, and 8 months after surgery. After the longest hypoestrogenicity period, acetylcholine-mediated aortic relaxation was attenuated and insensitive to E
2
administration despite endothelial integrity. Whereas no rapid vasorelaxant responses were elicited by an estrogen receptor (ER) β–selective agonist, responses to E
2
and an ERα selective agonist waned postovariectomy at any given time and were restored by E
2
treatment after 1 and 4 months but not 8 months postovariectomy. Accordingly, endothelial ERα mRNA and protein expression declined ≈6-fold after prolonged hypoestrogenicity and was restored by estrogen replacement starting 1 month but not 8 months postovariectomy. Furthermore, the amount of active phosphorylated endothelial NO synthase rose significantly after E
2
replacement after 1 and 4 months but not 8 months postovariectomy. The present findings document that the functional impairment of the ERα/endothelial NO synthase signaling network after an extended period of hypoestrogenicity was not restored by E
2
administration, providing experimental support to early initiation of estrogen replacement with preferential ERα targeting to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Christian Pinna
- From the Department of Pharmacological Sciences (C.P., A.C., P.S., V.P., C.B.), University of Milan, Milan, Italy; and the Department of Pharmacology and Anaesthesiology (A.C., P.S., V.P., C.B.), University of Padova, Padova, Italy
| | - Andrea Cignarella
- From the Department of Pharmacological Sciences (C.P., A.C., P.S., V.P., C.B.), University of Milan, Milan, Italy; and the Department of Pharmacology and Anaesthesiology (A.C., P.S., V.P., C.B.), University of Padova, Padova, Italy
| | - Paola Sanvito
- From the Department of Pharmacological Sciences (C.P., A.C., P.S., V.P., C.B.), University of Milan, Milan, Italy; and the Department of Pharmacology and Anaesthesiology (A.C., P.S., V.P., C.B.), University of Padova, Padova, Italy
| | - Valeria Pelosi
- From the Department of Pharmacological Sciences (C.P., A.C., P.S., V.P., C.B.), University of Milan, Milan, Italy; and the Department of Pharmacology and Anaesthesiology (A.C., P.S., V.P., C.B.), University of Padova, Padova, Italy
| | - Chiara Bolego
- From the Department of Pharmacological Sciences (C.P., A.C., P.S., V.P., C.B.), University of Milan, Milan, Italy; and the Department of Pharmacology and Anaesthesiology (A.C., P.S., V.P., C.B.), University of Padova, Padova, Italy
| |
Collapse
|
40
|
Schroeder F, Atshaves BP, McIntosh AL, Gallegos AM, Storey SM, Parr RD, Jefferson JR, Ball JM, Kier AB. Sterol carrier protein-2: new roles in regulating lipid rafts and signaling. BIOCHIMICA ET BIOPHYSICA ACTA 2007; 1771:700-18. [PMID: 17543577 PMCID: PMC1989133 DOI: 10.1016/j.bbalip.2007.04.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Revised: 03/28/2007] [Accepted: 04/03/2007] [Indexed: 12/31/2022]
Abstract
Sterol carrier protein-2 (SCP-2) was independently discovered as a soluble protein that binds and transfers cholesterol as well as phospholipids (nonspecific lipid transfer protein, nsLTP) in vitro. Physiological functions of this protein are only now beginning to be resolved. The gene encoding SCP-2 also encodes sterol carrier protein-x (SCP-x) arising from an alternate transcription site. In vitro and in vivo SCP-x serves as a peroxisomal 3-ketoacyl-CoA thiolase in oxidation of branched-chain lipids (cholesterol to form bile acids; branched-chain fatty acid for detoxification). While peroxisomal SCP-2 facilitates branched-chain lipid oxidation, the role(s) of extraperoxisomal (up to 50% of total) are less clear. Studies using transfected fibroblasts overexpressing SCP-2 and hepatocytes from SCP-2/SCP-x gene-ablated mice reveal that SCP-2 selectively remodels the lipid composition, structure, and function of lipid rafts/caveolae. Studies of purified SCP-2 and in cells show that SCP-2 has high affinity for and selectively transfers many lipid species involved in intracellular signaling: fatty acids, fatty acyl CoAs, lysophosphatidic acid, phosphatidylinositols, and sphingolipids (sphingomyelin, ceramide, mono-di-and multi-hexosylceramides, gangliosides). SCP-2 selectively redistributes these signaling lipids between lipid rafts/caveolae and intracellular sites. These findings suggest SCP-2 serves not only in cholesterol and phospholipid transfer, but also in regulating multiple lipid signaling pathways in lipid raft/caveolae microdomains of the plasma membrane.
Collapse
Affiliation(s)
- Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX 77843-4466, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Tang XH, Suh MJ, Li R, Gudas LJ. Cell proliferation inhibition and alterations in retinol esterification induced by phytanic acid and docosahexaenoic acid. J Lipid Res 2007; 48:165-76. [PMID: 17068359 DOI: 10.1194/jlr.m600419-jlr200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated the effects of two natural dietary retinoid X receptor (RXR) ligands, phytanic acid (PA) and docosahexaenoic acid (DHA), on proliferation and on the metabolism of retinol (vitamin A) in both cultured normal human prostate epithelial cells (PrECs) and PC-3 prostate carcinoma cells. PA and DHA inhibited the proliferation of the parental PC-3 cells and PC-3 cells engineered to overexpress human lecithin:retinol acyltransferase (LRAT) in both the absence and presence of retinol. A synthetic RXR-specific ligand also inhibited PC-3 cell proliferation, whereas all-trans retinoic acid (ATRA) did not. PA and DHA treatment increased the levels of retinyl esters (REs) in both PrECs and PC-3 cells and generated novel REs that eluted on reverse-phase HPLC at 54.0 and 50.5 min, respectively. Mass spectrometric analyses demonstrated that these novel REs were retinyl phytanate (54.0 min) and retinyl docosahexaenoate (50.5 min). Neither PA nor DHA increased LRAT mRNA levels in these cells. In addition, we demonstrate that retinyl phytanate was generated by LRAT in the presence of PA and retinol; however, retinyl docosahexaenoate was produced by another enzyme in the presence of DHA and retinol.
Collapse
Affiliation(s)
- Xiao-Han Tang
- Department of Pharmacology, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | |
Collapse
|
42
|
Pinna C, Bolego C, Sanvito P, Pelosi V, Baetta R, Corsini A, Gaion RM, Cignarella A. Raloxifene elicits combined rapid vasorelaxation and long-term anti-inflammatory actions in rat aorta. J Pharmacol Exp Ther 2006; 319:1444-51. [PMID: 16943257 DOI: 10.1124/jpet.106.106062] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Previous studies reported the ability of raloxifene to acutely relax arterial and venous vessels, but the underlying mechanisms are controversial. Anti-inflammatory effects of the drug have been reported in nonvascular tissues. Therefore, the aim of this study was to investigate the nature of short- and long-term effects of raloxifene on selected aspects of vascular function in rat aorta. Isometric tension changes in response to raloxifene were recorded in aortic rings from ovariectomized female rats that underwent estrogen replacement, whereas long-term experiments were performed in isolated aortic smooth muscle cells (SMCs). Raloxifene (0.1 pM-0.1 microM) induced acute vasorelaxation through endothelium- and nitric oxide (NO)-dependent, prostanoid-independent mechanisms. The relaxant response to raloxifene was significantly weaker than that to 17beta-estradiol and was sensitive to neither the nonselective estrogen receptor antagonist ICI 182,780 [7,17-[9[(4,4,5,5,5-pentafluoropentyl)sulfinyl]nonyl]estra-1,3,5(10)-triene-3,17-diol] nor a selective estrogen receptor (ER) alpha antagonist. This rapid vasorelaxant effect was retained in aortic rings from rats treated with 0.1 mg/kg, but not 1 mg/kg, lipopolysaccharide, 4 h before sacrifice. In cultured aortic SMCs, raloxifene treatment (1 nM-1 microM) for 24 h reduced inducible NO synthase activation in response to cytokines. This effect was prevented by the selective ERalpha antagonist and was associated with up-regulation of ERalpha protein levels, which dropped markedly upon cytokine stimulation. These findings illustrate the relevance of classic ER-dependent pathways to the vascular anti-inflammatory effects rather than to the nongenomic vasorelaxation induced by raloxifene and may assist in the design of novel ER isoform-selective estrogen-receptor modulators targeted to the vascular system.
Collapse
Affiliation(s)
- Christian Pinna
- Department of Pharmacological Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Cignarella A, Minici C, Bolego C, Pinna C, Sanvito P, Gaion RM, Puglisi L. Potential pro-inflammatory action of resveratrol in vascular smooth muscle cells from normal and diabetic rats. Nutr Metab Cardiovasc Dis 2006; 16:322-329. [PMID: 16829340 DOI: 10.1016/j.numecd.2005.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2005] [Revised: 04/21/2005] [Accepted: 05/26/2005] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIM Based on the reported cardioprotective effects of resveratrol, a polyphenolic antioxidant abundant in grapes that binds to estrogen receptors, and the well-characterized anti-inflammatory properties of 17beta-estradiol, the effects of resveratrol on the functional expression of inflammatory enzymes were assessed in vascular smooth muscle cells (SMC) from normoglycaemic and streptozotocin-diabetic rats. METHODS AND RESULTS SMC were isolated from the aorta four weeks after treating rats with streptozotocin or its vehicle. In SMC exposed to a cytokine mixture for 24h, unexpectedly, treatment with resveratrol (0.1-100microM) as well as the structurally related isoflavone genistein (1nM-1microM) enhanced expression of inducible NO synthase (iNOS). Genistein failed to mimic the elevated iNOS activity induced by resveratrol. Inhibition of estrogen receptors by the pure antiestrogen ICI 182,780 reversed the action of resveratrol on iNOS. In addition, resveratrol failed to alter cyclooxygenase-2 protein levels but reduced the accumulation of prostaglandin E(2) in the culture medium of SMC from normoglycaemic, but not diabetic rats. CONCLUSIONS These results indicate that resveratrol, at concentrations approaching putative peak plasma levels in vivo, exhibited no anti-inflammatory properties in vascular SMC from normal and diabetic rats. By contrast, resveratrol displayed a potential pro-inflammatory activity in settings of vascular inflammation.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Aorta/enzymology
- Aorta/metabolism
- Diabetes Mellitus, Experimental
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Estradiol/pharmacology
- Genistein/pharmacology
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Nitric Oxide Synthase Type II/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Estrogen/metabolism
- Resveratrol
- Stilbenes/pharmacology
- Tissue Culture Techniques
Collapse
Affiliation(s)
- Andrea Cignarella
- Department of Pharmacological Sciences, University of Milan, via G. Balzaretti 9, I-20133 Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
44
|
Hostetler HA, Kier AB, Schroeder F. Very-long-chain and branched-chain fatty acyl-CoAs are high affinity ligands for the peroxisome proliferator-activated receptor alpha (PPARalpha). Biochemistry 2006; 45:7669-81. [PMID: 16768463 PMCID: PMC2593851 DOI: 10.1021/bi060198l] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Very-long-chain fatty acids (VLCFA) and branched-chain fatty acids (BCFA) are potent inducers of the peroxisome proliferator-activated receptor PPARalpha, a nuclear receptor that enhances transcription of peroxisomal enzymes mediating beta-oxidation of these potentially toxic fatty acids. However, it is not known whether the respective free fatty acids or their activated metabolites, i.e., CoA thioesters, (i) are the endogenous high-affinity PPARalpha ligands, (ii) alter PPARalpha conformation, and (iii) alter recruitment of coregulatory proteins to PPARalpha. As shown by quenching of PPARalpha intrinsic amino acid fluorescence, PPARalpha exhibited high affinity (3-29 nM Kds) for the CoA thioesters of the common (C20-C24) VLCFA. In contrast, with the exception of arachidonic acid (Kd = 20 nM), PPARalpha only weakly bound the VLCFA. PPARalpha also exhibited higher affinity for the CoA thioesters of BCFA (phytanoyl-CoA, pristanoyl-CoA; Kds near 11 nM) than for the respective free branched-chain fatty acids. As shown by circular dichroism, the high affinity VLCFA-CoA and BCFA-CoA strongly altered PPARalpha conformation. Likewise, the high affinity VLCFA-CoA and BCFA-CoA altered cofactor recruitment to PPARalpha as shown by coimmunoprecipitation from liver homogenates. In contrast, nearly all the respective free fatty acids elicited only weak conformational changes in PPARalpha and did not alter cofactor recruitment to PPARalpha. In summary, the CoA thioesters of very-long-chain and branched-chain fatty acids are much more potent PPARalpha ligands than the free acids, resulting in altered PPARalpha conformation and cofactor recruitment. Since these are hallmarks of ligand-activated nuclear receptors, this suggests that the CoA thioesters are the active forms of these PPARalpha ligands.
Collapse
Affiliation(s)
- Heather A Hostetler
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, Texas 77843-4466, USA
| | | | | |
Collapse
|
45
|
Schönfeld P, Kahlert S, Reiser G. A study of the cytotoxicity of branched-chain phytanic acid with mitochondria and rat brain astrocytes. Exp Gerontol 2006; 41:688-96. [PMID: 16616447 DOI: 10.1016/j.exger.2006.02.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 02/24/2006] [Accepted: 02/27/2006] [Indexed: 11/29/2022]
Abstract
Phytanic acid, a saturated fatty acid of 20-carbon-atoms with isoprenoic structure, is formed from the phytol-side chain of chlorophyll in ruminants. Degradation of phytanic acid is blocked in Refsum disease by several enzymatic defects of peroxisomal degradation of branched-chain fatty acids. Refsum disease is an inherited neurological disorder progressively developing from early childhood to adultness. Clinical signs are attributed to toxicity of phytanic acid, which accumulates to unusually high levels in the tissue and serum of patients suffering from untreated Refsum disease. We report here that hippocampal astrocytes isolated from rat brain, which were exposed to phytanic acid (50 microM) die within a few hours. In situ depolarization of mitochondria and an increase of cytosolic Ca2+ precede cell death. Therefore, we also investigated the influence of phytanic acid on physiology of mitochondria isolated from rat brain. Mitochondria become functionally impaired by phytanic acid, as indicated by uncoupling (resting state), inhibition of the electron transport (state 3), stimulation of ROS-generation, decline of Ca2+ loading and severe release of cytochrome c. Thus, phytanic acid seems to initiate astrocyte cell death by activating the mitochondrial route of apoptosis.
Collapse
Affiliation(s)
- Peter Schönfeld
- Institut für Biochemie, Otto-von-Guericke-Universität Magdeburg, Leipziger Str. 44, Medizinische Fakultät, D-39120 Magdeburg, Germany.
| | | | | |
Collapse
|
46
|
Reiser G, Schönfeld P, Kahlert S. Mechanism of toxicity of the branched‐chain fatty acid phytanic acid, a marker of Refsum disease, in astrocytes involves mitochondrial impairment. Int J Dev Neurosci 2006; 24:113-22. [PMID: 16386870 DOI: 10.1016/j.ijdevneu.2005.11.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Accepted: 11/10/2005] [Indexed: 10/25/2022] Open
Abstract
Phytanic acid is a saturated branched-chain fatty acid, which is formed by bacterial degradation of chlorophyll in the intestinal tract of ruminants. The methyl group in beta-position prevents degradation of phytanic acid by the beta-oxidation pathway. Therefore, degradation of phytanic acid is initiated by alpha-oxidation in peroxisomes. The inherited peroxisomal disorder Refsum disease is characterised by accumulation of phytanic acid. Unusually high concentrations of phytanic acid can be found in the plasma of Refsum disease patients, who suffer from neurodegeneration and muscle dystrophy. Phytanic acid has been suggested to be causally involved in the clinical symptoms. To elucidate the pathogenic mechanism, we investigated the influence of phytanic acid in rat hippocampal astrocytes by monitoring the cytosolic Ca(2+) concentration, the mitochondrial membrane potential (Deltapsi(m)), the generation of reactive oxygen species as well as the cellular ATP level. In response to phytanic acid (100 microM) cytosolic Ca(2+) was quickly increased. The phytanic acid-evoked Ca(2+) response was transient and involved activation of intracellular Ca(2+) stores. In isolated rat brain mitochondria, phytanic acid dissipated Deltapsi(m) in a reversible and dose-dependent manner. Moreover, phytanic acid released cytochrome c from mitochondria. Depending on the mitochondrial activity state, phytanic acid either stimulated or inhibited the electron flux within the respiratory chain. In addition, phytanic acid induced substantial generation of reactive oxygen species in isolated mitochondria as well as in intact cells. Phytanic acid caused cell death of astrocytes within a few hours of exposure. In conclusion, we suggest that phytanic acid initiates astrocyte cell death by activating the mitochondrial route of apoptosis.
Collapse
Affiliation(s)
- Georg Reiser
- Institut für Neurobiochemie, Otto-von-Guericke-Universität Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | | | | |
Collapse
|
47
|
Bolego C, Cignarella A, Sanvito P, Pelosi V, Pellegatta F, Puglisi L, Pinna C. The acute estrogenic dilation of rat aorta is mediated solely by selective estrogen receptor-alpha agonists and is abolished by estrogen deprivation. J Pharmacol Exp Ther 2005; 313:1203-8. [PMID: 15722404 DOI: 10.1124/jpet.104.082867] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Estrogen is known to induce rapid vasodilatory response in isolated arteries. Because estrogen is a nonselective receptor agonist, the involvement of estrogen receptor (ER) subtypes in acute estrogenic responses has remained elusive. Acute administration of the selective ERalpha agonist 4,4',4''-(4-propyl-[(1)H]pyrazole-1,3,5-triyl) tris-phenol (PPT) to precontracted aortic rings from intact female rats dose-dependently induced an ER-dependent vascular relaxation fully overlapping to that induced by 17beta-estradiol. By contrast, the selective ERbeta agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN) had no acute effect on vasomotion. This short-term vasorelaxant action of PPT was abolished by the NO synthase inhibitor N(omega)-nitro-l-arginine methyl ester and by endothelium removal. In aortic tissues from ovariectomized (OVX) rats, however, neither 17beta-estradiol nor PPT induced acute vascular relaxation. The effect of PPT was restored in preparations from estrogen-replaced OVX rats, whereas DPN remained ineffective even after estrogen replacement. PPT acted through an ER-dependent mechanism, as shown by impaired response in the presence of the anti-estrogen ICI 182,780 (7alpha,17beta-[9[(4,4,5,5,5-pentafluoropentyl)sulfinyl]nonyl]estra-1,3,5(10)-triene-3,17-diol). Accordingly, isolated rat aortic endothelial cells expressed both ERalpha and ERbeta. These data show that selective ERalpha but not ERbeta agonists reproduced the acute vasodilation of estrogen via a receptor-mediated pathway in the aorta from intact as well as 17beta-estradiol-replaced OVX rats. This beneficial effect was undetectable in tissues from OVX rats. Selective pharmacological targeting of ER subtypes may thus represent a novel and promising approach in the treatment of vascular disease.
Collapse
Affiliation(s)
- Chiara Bolego
- Department of Pharmacological Sciences, University of Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
48
|
Kahlert S, Schönfeld P, Reiser G. The Refsum disease marker phytanic acid, a branched chain fatty acid, affects Ca2+ homeostasis and mitochondria, and reduces cell viability in rat hippocampal astrocytes. Neurobiol Dis 2005; 18:110-8. [PMID: 15649701 DOI: 10.1016/j.nbd.2004.08.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2004] [Revised: 06/03/2004] [Accepted: 08/27/2004] [Indexed: 12/29/2022] Open
Abstract
The saturated branched chain fatty acid, phytanic acid, a degradation product of chlorophyll, accumulates in Refsum disease, an inherited peroxisomal disorder with neurological clinical features. To elucidate the pathogenic mechanism, we investigated the influence of phytanic acid on cellular physiology of rat hippocampal astrocytes. Phytanic acid (100 microM) induced an immediate transient increase in cytosolic Ca2+ concentration, followed by a plateau. The peak of this biphasic Ca2+ response was largely independent of extracellular Ca2+, indicating activation of cellular Ca2+ stores by phytanic acid. Phytanic acid depolarized mitochondria without causing in situ swelling of mitochondria. The slow decrease of mitochondrial potential is not consistent with fast and simultaneous opening of the mitochondrial permeability transition pore. However, phytanic acid induced substantial generation of reactive oxygen species. Phytanic acid caused astroglia cell death after a few hours of exposure. We suggest that the cytotoxic effect of phytanic acid seems to be due to a combined action on Ca2+ regulation, mitochondrial depolarization, and increased ROS generation in brain cells.
Collapse
Affiliation(s)
- Stefan Kahlert
- Institut für Neurobiochemie Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Germany
| | | | | |
Collapse
|
49
|
van den Brink DM, van Miert JNI, Dacremont G, Rontani JF, Wanders RJA. Characterization of the final step in the conversion of phytol into phytanic acid. J Biol Chem 2005; 280:26838-44. [PMID: 15866875 DOI: 10.1074/jbc.m501861200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phytol is a branched-chain fatty alcohol that is a naturally occurring precursor of phytanic acid, a fatty acid involved in the pathogenesis of Refsum disease. The conversion of phytol into phytanic acid is generally believed to take place via three enzymatic steps that involve 1) oxidation to its aldehyde, 2) further oxidation to phytenic acid, and 3) reduction of the double bond at the 2,3 position, yielding phytanic acid. Our recent investigations of this mechanism have elucidated the enzymatic steps leading to phytenic acid production, but the final step of the pathway has not been investigated so far. In this study, we describe the characterization of phytenic acid reduction in rat liver. NADPH-dependent conversion of phytenic acid into phytanic acid was detected, although at a slow rate. However, it was shown that phytenic acid can be activated to its CoA ester and that reduction of phytenoyl-CoA is much more efficient than that of phytenic acid. Furthermore, in rat hepatocytes cultured in the presence of phytol, phytenoyl-CoA could be detected, showing that it is a bona fide intermediate of phytol degradation. Subcellular fractionation experiments revealed that phytenoyl-CoA reductase activity is present in peroxisomes and mitochondria. With these findings, we have accomplished the full elucidation of the mechanism by which phytol is converted into phytanic acid.
Collapse
Affiliation(s)
- Daan M van den Brink
- Department of Clinical Chemistry and Pediatrics, University of Amsterdam, Academic Medical Center, Emma Children's Hospital, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
50
|
Schönfeld P, Kahlert S, Reiser G. In brain mitochondria the branched-chain fatty acid phytanic acid impairs energy transduction and sensitizes for permeability transition. Biochem J 2005; 383:121-8. [PMID: 15198638 PMCID: PMC1134050 DOI: 10.1042/bj20040583] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2004] [Revised: 06/04/2004] [Accepted: 06/15/2004] [Indexed: 12/27/2022]
Abstract
Phytanic acid (3,7,11,15-tetramethylhexadecanoic acid) accumulates at high levels throughout the body in the adult form of Refsum disease, a peroxisomal genetic disorder. However, it is still unclear why increased levels of phytanic acid have cytotoxic effects. In the present study, we examined the influence of non-esterified phytanic acid on energy-related functions of mitochondria from adult rat brain. Phytanic acid at low concentrations (5-20 microM, i.e. 5-20 nmol/mg of mitochondrial protein) de-energized mitochondria, as indicated by depolarization, stimulation of non-phosphorylating oxygen uptake and inhibition of the reduction of the tetrazolium dye 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide. The unbranched homologue palmitic acid exerted much smaller effects. In addition, phytanic acid reduced state 3 respiration, which was partly due to inhibition of the ADP/ATP carrier. Phytanic acid decreased the rate of adenine nucleotide exchange and increased the degree of control, which the ADP/ATP carrier has on state 3 respiration. Important for functional consequences is the finding that mitochondria, which are preloaded with small amounts of Ca2+ (100 nmol/mg of protein), became highly sensitized to rapid permeability transition even when only low concentrations of phytanic acid (below 5 microM) were applied. In conclusion, the incorporation of phytanic acid into the inner mitochondrial membrane increases the membrane H+ conductance and disturbs the protein-linked functions in energy coupling. This is most probably essential for the short-term toxicity of phytanic acid. Thus in neural tissue, which becomes enriched with phytanic acid, the reduction in mitochondrial ATP supply and the facilitation of the opening of the permeability transition pore are two major mechanisms by which the branched-chain fatty acid phytanic acid induces the onset of degenerative processes.
Collapse
Affiliation(s)
- Peter Schönfeld
- Medizinische Fakultät, Institut für Biochemie, Otto-von-Guericke-Universität Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany.
| | | | | |
Collapse
|