1
|
Tracy KM, Shishido Y, Petrovic M, Murphy A, Adesanya T, Fortier AK, Harris TR, Cortelli M, Tucker WD, François SA, Petree B, Raietparvar K, Simon V, Johnson CA, Simonds E, Poland J, Glomp GA, Crannell C, Liang J, Marshall A, Hinton A, Shaver CM, Demarest CT, Ukita R, Shah AS, Rizzari M, Montenovo M, Rauf MA, McReynolds M, Bacchetta M. 10 degree C static storage of porcine donation after circulatory death livers improves biliary viability and mitigates ischemia-reperfusion injury. Am J Transplant 2025:S1600-6135(25)00147-9. [PMID: 40120647 DOI: 10.1016/j.ajt.2025.03.018] [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: 11/12/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Optimized static cold storage has the potential to improve the preservation of organs most vulnerable to ischemia-reperfusion injury. Data from lung transplantation suggest that storage at 10 °C improves mitochondrial preservation and subsequent allograft function compared with conventional storage on ice. Using a porcine model of donation after circulatory death, we compared static storage of livers at 10 °C to ice. Livers (N = 5 per group) underwent 10 hours of storage followed by 4 hours of normothermic machine perfusion (NMP) for real-time allograft assessment. Allografts were compared using established NMP viability criteria, tissue immunostaining, and tissue metabolomics. Livers stored at 10 °C demonstrated lower portal venous vascular resistance and greater hepatic artery vasoresponsiveness. Lactate clearance during NMP was similar between the groups. Livers stored at 10 °C showed favorable biochemical parameters of biliary viability, including greater bile volume, pH, and bicarbonate. Metabolomics analysis revealed increased aerobic respiration, improved electron transport chain function, and less DNA damage after reperfusion of livers stored at 10 °C. Static storage of donation after circulatory death livers with extended cold ischemic time at 10 °C demonstrates superior allograft function with evidence of improved biliary viability and mitochondrial function compared with ice. These data suggest that storage at 10 °C should be considered for translation to clinical practice.
Collapse
Affiliation(s)
- Kaitlyn M Tracy
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yutaka Shishido
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mark Petrovic
- Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Alexandria Murphy
- Department of Biochemistry and Molecular Biology, The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - TiOluwanimi Adesanya
- Vanderbilt University, Nashville, Tennessee, USA; Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Timothy R Harris
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael Cortelli
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - William D Tucker
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sean A François
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Brandon Petree
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Victoria Simon
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Carl A Johnson
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - John Poland
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Christian Crannell
- Department of Surgery, Division of Kidney and Pancreas Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jiancong Liang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andrea Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Ciara M Shaver
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Caitlin T Demarest
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rei Ukita
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ashish S Shah
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael Rizzari
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Martin Montenovo
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Muhammad Ameen Rauf
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Melanie McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Matthew Bacchetta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA; Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
2
|
Poleto KH, Janner DE, Dahleh MMM, Poetini MR, Fernandes EJ, Musachio EAS, de Almeida FP, Amador ECDM, Reginaldo JC, Carriço MRS, Roehrs R, Prigol M, Guerra GP. p-Coumaric acid potential in restoring neuromotor function and oxidative balance through the Parkin pathway in a Parkinson disease-like model in Drosophila melanogaster. Food Chem Toxicol 2024; 193:115002. [PMID: 39276910 DOI: 10.1016/j.fct.2024.115002] [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: 03/05/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
p-Coumaric acid is a significant phenolic compound known for its potent antioxidant activity. Thus, this study investigated the effects of p-coumaric acid on the behavioral and neurochemical changes induced in Drosophila melanogaster by exposure to rotenone in a Parkinson disease (PD)-like model. The flies were divided into four groups and maintained for seven days on different diets: a standard diet (control), a diet containing rotenone (500 μM), a control diet to which p-coumaric acid was added on the fourth day (0.3 μM), and a diet initially containing rotenone (500 μM) with p-coumaric acid added on the fourth day (0.3 μM). Exposure to p-coumaric acid ameliorated locomotor impairment and reduced mortality induced by rotenone. Moreover, p-coumaric acid normalized oxidative stress markers (ROS, TBARS, SOD, CAT, GST, and NPSH), mitigated oxidative damage, and reflected in the recovery of dopamine levels, AChE activity, and cellular viability post-rotenone exposure. Additionally, p-coumaric acid restored the immunoreactivity of Parkin and Nrf2. The results affirm that p-coumaric acid effectively mitigates PD-like model-induced damage, underscoring its antioxidant potency and potential neuroprotective effect.
Collapse
Affiliation(s)
- Kétnne Hanna Poleto
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil; Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil
| | - Dieniffer Espinosa Janner
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil; Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil
| | - Mustafa Munir Mustafa Dahleh
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil; Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil
| | - Márcia Rósula Poetini
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil; Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil
| | - Eliana Jardim Fernandes
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil; Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil
| | - Elize Aparecida Santos Musachio
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil; Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil
| | - Francielli Polet de Almeida
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil; Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil
| | - Elen Caroline de Matos Amador
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil
| | - Jocemara Corrêa Reginaldo
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil
| | - Murilo Ricardo Sigal Carriço
- Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil; Environmental and Toxicological Chemical Analysis Laboratory, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil
| | - Rafael Roehrs
- Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil; Environmental and Toxicological Chemical Analysis Laboratory, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil
| | - Marina Prigol
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil; Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil
| | - Gustavo Petri Guerra
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, Itaqui, Rio Grande do Sul, Brazil; Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana Campus, Uruguaiana, Rio Grande do Sul, Brazil.
| |
Collapse
|
3
|
Glänzel NM, Parmeggiani B, Grings M, Seminotti B, Brondani M, Bobermin LD, Ribeiro CAJ, Quincozes-Santos A, Vockley J, Leipnitz G. Myelin Disruption, Neuroinflammation, and Oxidative Stress Induced by Sulfite in the Striatum of Rats Are Mitigated by the pan-PPAR agonist Bezafibrate. Cells 2023; 12:1557. [PMID: 37371027 DOI: 10.3390/cells12121557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Sulfite predominantly accumulates in the brain of patients with isolated sulfite oxidase (ISOD) and molybdenum cofactor (MoCD) deficiencies. Patients present with severe neurological symptoms and basal ganglia alterations, the pathophysiology of which is not fully established. Therapies are ineffective. To elucidate the pathomechanisms of ISOD and MoCD, we investigated the effects of intrastriatal administration of sulfite on myelin structure, neuroinflammation, and oxidative stress in rat striatum. Sulfite administration decreased FluoromyelinTM and myelin basic protein staining, suggesting myelin abnormalities. Sulfite also increased the staining of NG2, a protein marker of oligodendrocyte progenitor cells. In line with this, sulfite also reduced the viability of MO3.13 cells, which express oligodendroglial markers. Furthermore, sulfite altered the expression of interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-10 (IL-10), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and heme oxygenase-1 (HO-1), indicating neuroinflammation and redox homeostasis disturbances. Iba1 staining, another marker of neuroinflammation, was also increased by sulfite. These data suggest that myelin changes and neuroinflammation induced by sulfite contribute to the pathophysiology of ISOD and MoCD. Notably, post-treatment with bezafibrate (BEZ), a pan-PPAR agonist, mitigated alterations in myelin markers and Iba1 staining, and IL-1β, IL-6, iNOS and HO-1 expression in the striatum. MO3.13 cell viability decrease was further prevented. Moreover, pre-treatment with BEZ also attenuated some effects. These findings show the modulation of PPAR as a potential opportunity for therapeutic intervention in these disorders.
Collapse
Affiliation(s)
- Nícolas Manzke Glänzel
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - Belisa Parmeggiani
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - Mateus Grings
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - Bianca Seminotti
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Morgana Brondani
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - Larissa D Bobermin
- Programa de Pós-Graduação Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - César A J Ribeiro
- Natural and Humanities Sciences Center, Universidade Federal do ABC, São Bernardo do Campo 09606-070, SP, Brazil
| | - André Quincozes-Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
- Programa de Pós-Graduação Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - Jerry Vockley
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
- Programa de Pós-Graduação Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| |
Collapse
|
4
|
Kasperek MC, Mailing L, Piccolo BD, Moody B, Lan R, Gao X, Hernandez‐Saavedra D, Woods JA, Adams SH, Allen JM. Exercise training modifies xenometabolites in gut and circulation of lean and obese adults. Physiol Rep 2023; 11:e15638. [PMID: 36945966 PMCID: PMC10031301 DOI: 10.14814/phy2.15638] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/23/2023] Open
Abstract
Regular, moderate exercise modifies the gut microbiome and contributes to human metabolic and immune health. The microbiome may exert influence on host physiology through the microbial production and modification of metabolites (xenometabolites); however, this has not been extensively explored. We hypothesized that 6 weeks of supervised, aerobic exercise 3×/week (60%-75% heart rate reserve [HRR], 30-60 min) in previously sedentary, lean (n = 14) and obese (n = 10) adults would modify both the fecal and serum xenometabolome. Serum and fecal samples were collected pre- and post-6 week intervention and analyzed by liquid chromatography/tandem mass spectrometry (LC-MS/MS). Linear mixed models (LMMs) identified multiple fecal and serum xenometabolites responsive to exercise training. Further cluster and pathway analysis revealed that the most prominent xenometabolic shifts occurred within aromatic amino acid (ArAA) metabolic pathways. Fecal and serum ArAA derivatives correlated with body composition (lean mass), markers of insulin sensitivity (insulin, HOMA-IR) and cardiorespiratory fitness (V ̇ O 2 max $$ \dot{\mathrm{V}}{\mathrm{O}}_{2\max } $$ ), both at baseline and in response to exercise training. Two serum aromatic microbial-derived amino acid metabolites that were upregulated following the exercise intervention, indole-3-lactic acid (ILA: fold change: 1.2, FDR p < 0.05) and 4-hydroxyphenyllactic acid (4-HPLA: fold change: 1.3, FDR p < 0.05), share metabolic pathways within the microbiota and were associated with body composition and markers of insulin sensitivity at baseline and in response to training. These data provide evidence of physiologically relevant shifts in microbial metabolism that occur in response to exercise training, and reinforce the view that host metabolic health influences gut microbiota population and function. Future studies should consider the microbiome and xenometabolome when investigating the health benefits of exercise.
Collapse
Affiliation(s)
- Mikaela C. Kasperek
- Division of Nutritional SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Kinesiology and Community HealthUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Lucy Mailing
- Division of Nutritional SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Brian D. Piccolo
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Becky Moody
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Renny Lan
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Xiaotian Gao
- Department of Kinesiology and Community HealthUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Diego Hernandez‐Saavedra
- Department of Kinesiology and Community HealthUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Jeffrey A. Woods
- Division of Nutritional SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Kinesiology and Community HealthUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Sean H. Adams
- Department of SurgeryUniversity of California, Davis School of MedicineSacramentoCaliforniaUSA
- Center for Alimentary and Metabolic ScienceUniversity of California, DavisSacramentoCaliforniaUSA
| | - Jacob M. Allen
- Division of Nutritional SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Kinesiology and Community HealthUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
5
|
Liang X, Fu Y, Cao WT, Wang Z, Zhang K, Jiang Z, Jia X, Liu CY, Lin HR, Zhong H, Miao Z, Gou W, Shuai M, Huang Y, Chen S, Zhang B, Chen YM, Zheng JS. Gut microbiome, cognitive function and brain structure: a multi-omics integration analysis. Transl Neurodegener 2022; 11:49. [PMID: 36376937 PMCID: PMC9661756 DOI: 10.1186/s40035-022-00323-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Microbiome-gut-brain axis may be involved in the progression of age-related cognitive impairment and relevant brain structure changes, but evidence from large human cohorts is lacking. This study was aimed to investigate the associations of gut microbiome with cognitive impairment and brain structure based on multi-omics from three independent populations. METHODS We included 1430 participants from the Guangzhou Nutrition and Health Study (GNHS) with both gut microbiome and cognitive assessment data available as a discovery cohort, of whom 272 individuals provided fecal samples twice before cognitive assessment. We selected 208 individuals with baseline microbiome data for brain magnetic resonance imaging during the follow-up visit. Fecal 16S rRNA and shotgun metagenomic sequencing, targeted serum metabolomics, and cytokine measurements were performed in the GNHS. The validation analyses were conducted in an Alzheimer's disease case-control study (replication study 1, n = 90) and another community-based cohort (replication study 2, n = 1300) with cross-sectional dataset. RESULTS We found protective associations of specific gut microbial genera (Odoribacter, Butyricimonas, and Bacteroides) with cognitive impairment in both the discovery cohort and the replication study 1. Result of Bacteroides was further validated in the replication study 2. Odoribacter was positively associated with hippocampal volume (β, 0.16; 95% CI 0.06-0.26, P = 0.002), which might be mediated by acetic acids. Increased intra-individual alterations in gut microbial composition were found in participants with cognitive impairment. We also identified several serum metabolites and inflammation-associated metagenomic species and pathways linked to impaired cognition. CONCLUSIONS Our findings reveal that specific gut microbial features are closely associated with cognitive impairment and decreased hippocampal volume, which may play an important role in dementia development.
Collapse
Affiliation(s)
- Xinxiu Liang
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Yuanqing Fu
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Wen-Ting Cao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
- School of Public Health, Hainan Medical University, Haikou, 571199, China
| | - Zhihong Wang
- Chinese Center for Disease Control and Prevention, National Institute for Nutrition and Health, Beijing, 100050, China
- Key Laboratory of Trace Element Nutrition, National Health Commission, Beijing, 100050, China
| | - Ke Zhang
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Zengliang Jiang
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China
| | - Xiaofang Jia
- Chinese Center for Disease Control and Prevention, National Institute for Nutrition and Health, Beijing, 100050, China
- Key Laboratory of Trace Element Nutrition, National Health Commission, Beijing, 100050, China
| | - Chun-Ying Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Hong-Rou Lin
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Haili Zhong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zelei Miao
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Wanglong Gou
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Menglei Shuai
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Yujing Huang
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Bing Zhang
- Chinese Center for Disease Control and Prevention, National Institute for Nutrition and Health, Beijing, 100050, China.
- Key Laboratory of Trace Element Nutrition, National Health Commission, Beijing, 100050, China.
| | - Yu-Ming Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Ju-Sheng Zheng
- School of Life Sciences, Westlake University, 18 Shilongshan Rd, Cloud Town, Hangzhou, 310024, China.
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, 310024, China.
| |
Collapse
|
6
|
Wajner M, Vargas CR, Amaral AU. Disruption of mitochondrial functions and oxidative stress contribute to neurologic dysfunction in organic acidurias. Arch Biochem Biophys 2020; 696:108646. [PMID: 33098870 DOI: 10.1016/j.abb.2020.108646] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 02/08/2023]
Abstract
Organic acidurias (OADs) are inherited disorders of amino acid metabolism biochemically characterized by accumulation of short-chain carboxylic acids in tissues and biological fluids of the affected patients and clinically by predominant neurological manifestations. Some of these disorders are amenable to treatment, which significantly decreases mortality and morbidity, but it is still ineffective to prevent long-term neurologic and systemic complications. Although pathogenesis of OADs is still poorly established, recent human and animal data, such as lactic acidosis, mitochondrial morphological alterations, decreased activities of respiratory chain complexes and altered parameters of oxidative stress, found in tissues from patients and from genetic mice models with these diseases indicate that disruption of critical mitochondrial functions and oxidative stress play an important role in their pathophysiology. Furthermore, organic acids that accumulate in the most prevalent OADs were shown to compromise bioenergetics, by decreasing ATP synthesis, mitochondrial membrane potential, reducing equivalent content and calcium retention capacity, besides inducing mitochondrial swelling, reactive oxygen and nitrogen species generation and apoptosis. It is therefore presumed that secondary mitochondrial dysfunction and oxidative stress caused by major metabolites accumulating in OADs contribute to tissue damage in these pathologies.
Collapse
Affiliation(s)
- Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; 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.
| | - Carmen Regla Vargas
- Programa de Pós-Graduação em Ciências Biológicas: 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; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alexandre Umpierrez Amaral
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Ciências Biológicas, Universidade Regional Integrada do Alto Uruguai e das Missões, Erechim, RS, Brazil
| |
Collapse
|
7
|
Jones DE, Perez L, Ryan RO. 3-Methylglutaric acid in energy metabolism. Clin Chim Acta 2019; 502:233-239. [PMID: 31730811 DOI: 10.1016/j.cca.2019.11.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 12/16/2022]
Abstract
3-methylglutaric (3MG) acid is a conspicuous C6 dicarboxylic organic acid classically associated with two distinct leucine pathway enzyme deficiencies. 3MG acid is excreted in urine of individuals harboring deficiencies in 3-hydroxy-3-methylglutaryl (HMG) CoA lyase (HMGCL) or 3-methylglutaconyl CoA hydratase (AUH). Whereas 3MG CoA is not part of the leucine catabolic pathway, it is likely formed via a side reaction involving reduction of the α-ß trans double bond in the leucine pathway intermediate, 3-methylglutaconyl CoA. While the metabolic basis for the accumulation of 3MG acid in subjects with deficiencies in HMGCL or AUH is apparent, the occurrence of 3MG aciduria in a host of unrelated inborn errors of metabolism associated with compromised mitochondrial energy metabolism is less clear. Herein, a novel mitochondrial biosynthetic pathway termed "the acetyl CoA diversion pathway", provides an explanation. The pathway is initiated by defective electron transport chain function which, ultimately, inhibits acetyl CoA entry into the TCA cycle. When this occurs, 3MG acid is synthesized in five steps from acetyl CoA via a novel reaction sequence, providing a metabolic rationale for the connection between 3MG aciduria and compromised mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Dylan E Jones
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV 89557, United States
| | - Leanne Perez
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV 89557, United States
| | - Robert O Ryan
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, NV 89557, United States.
| |
Collapse
|
8
|
Vamecq J, Papegay B, Nuyens V, Boogaerts J, Leo O, Kruys V. Mitochondrial dysfunction, AMPK activation and peroxisomal metabolism: A coherent scenario for non-canonical 3-methylglutaconic acidurias. Biochimie 2019; 168:53-82. [PMID: 31626852 DOI: 10.1016/j.biochi.2019.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022]
Abstract
The occurrence of 3-methylglutaconic aciduria (3-MGA) is a well understood phenomenon in leucine oxidation and ketogenesis disorders (primary 3-MGAs). In contrast, its genesis in non-canonical (secondary) 3-MGAs, a growing-up group of disorders encompassing more than a dozen of inherited metabolic diseases, is a mystery still remaining unresolved for three decades. To puzzle out this anthologic problem of metabolism, three clues were considered: (i) the variety of disorders suggests a common cellular target at the cross-road of metabolic and signaling pathways, (ii) the response to leucine loading test only discriminative for primary but not secondary 3-MGAs suggests these latter are disorders of extramitochondrial HMG-CoA metabolism as also attested by their failure to increase 3-hydroxyisovalerate, a mitochondrial metabolite accumulating only in primary 3-MGAs, (iii) the peroxisome is an extramitochondrial site possessing its own pool and displaying metabolism of HMG-CoA, suggesting its possible involvement in producing extramitochondrial 3-methylglutaconate (3-MG). Following these clues provides a unifying common basis to non-canonical 3-MGAs: constitutive mitochondrial dysfunction induces AMPK activation which, by inhibiting early steps in cholesterol and fatty acid syntheses, pipelines cytoplasmic acetyl-CoA to peroxisomes where a rise in HMG-CoA followed by local dehydration and hydrolysis may lead to 3-MGA yield. Additional contributors are considered, notably for 3-MGAs associated with hyperammonemia, and to a lesser extent in CLPB deficiency. Metabolic and signaling itineraries followed by the proposed scenario are essentially sketched, being provided with compelling evidence from the literature coming in their support.
Collapse
Affiliation(s)
- Joseph Vamecq
- Inserm, CHU Lille, Univ Lille, Department of Biochemistry and Molecular Biology, Laboratory of Hormonology, Metabolism-Nutrition & Oncology (HMNO), Center of Biology and Pathology (CBP) Pierre-Marie Degand, CHRU Lille, EA 7364 RADEME, University of North France, Lille, France.
| | - Bérengère Papegay
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Vincent Nuyens
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Jean Boogaerts
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Oberdan Leo
- Laboratory of Immunobiology, Department of Molecular Biology, ULB Immunology Research Center (UIRC), Free University of Brussels (ULB), Gosselies, Belgium
| | - Véronique Kruys
- Laboratory of Molecular Biology of the Gene, Department of Molecular Biology, ULB Immunology Research Center (UIRC), Free University of Brussels (ULB), Gosselies, Belgium
| |
Collapse
|
9
|
Germoush MO, Othman SI, Al-Qaraawi MA, Al-Harbi HM, Hussein OE, Al-Basher G, Alotaibi MF, Elgebaly HA, Sandhu MA, Allam AA, Mahmoud AM. Umbelliferone prevents oxidative stress, inflammation and hematological alterations, and modulates glutamate-nitric oxide-cGMP signaling in hyperammonemic rats. Biomed Pharmacother 2018; 102:392-402. [PMID: 29573618 DOI: 10.1016/j.biopha.2018.03.104] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/02/2018] [Accepted: 03/17/2018] [Indexed: 01/09/2023] Open
Abstract
Hepatic encephalopathy (HE) is a serious neuropsychiatric complication that occurs as a result of liver failure. Umbelliferone (UMB; 7-hydroxycoumarin) is a natural product with proven hepatoprotective activity; however, nothing has yet been reported on its protective effect against hyperammonemia, the main culprit behind the symptoms of HE. Here, we evaluated the effect of UMB against ammonium chloride (NH4Cl)-induced hyperammonemia, oxidative stress, inflammation and hematological alterations in rats. We demonstrated the modulatory role of UMB on the glutamate-nitric oxide (NO)-cGMP pathways in the cerebrum of rats. Rats received intraperitoneal injections of NH4Cl (3 times/week) for 8 weeks and concomitantly received 50 mg/kg UMB. NH4Cl-induced rats showed significantly elevated blood ammonia and liver function markers. Lipid peroxidation and NO were increased in the liver and cerebrum of rats while the antioxidant defenses were declined. UMB significantly reduced blood ammonia, liver function markers, lipid peroxidation and NO, and enhanced the antioxidant defenses in NH4Cl-induced rats. UMB significantly prevented anemia, leukocytosis, thrombocytopenia and prolongation of PT and aPTT. Hyperammonemic rats showed elevated levels of cerebral TNF-α, IL-1β and glutamine as well as increased activity and expression of Na+/K+-ATPase, effects that were significantly reversed by UMB. In addition, UMB down-regulated nitric oxide synthase and soluble guanylate cyclase in the cerebrum of hyperammonemic rats. In conclusion, this study provides evidence that UMB protects against hyperammonemia via attenuation of oxidative stress and inflammation. UMB prevents hyperammonemia associated hematological alterations and therefore represents a promising protective agent against the deleterious effects of excess ammonia.
Collapse
Affiliation(s)
- Mousa O Germoush
- Biology Department, Faculty of Science, Jouf University, Aljouf, Saudi Arabia
| | - Sarah I Othman
- Biology Department, Faculty of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Maha A Al-Qaraawi
- Biology Department, Faculty of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Hanan M Al-Harbi
- Biology Department, Faculty of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Omnia E Hussein
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Gadh Al-Basher
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed F Alotaibi
- Physiology Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hassan A Elgebaly
- Biology Department, Faculty of Science, Jouf University, Aljouf, Saudi Arabia
| | - Mansur A Sandhu
- Biomedical Sciences Department, Faculty of Veterinary & Animal Sciences, PMAS, Arid Agriculture University, Rawalpindi, Pakistan
| | - Ahmed A Allam
- Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Ayman M Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt; Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt.
| |
Collapse
|
10
|
Commiphora molmol Modulates Glutamate-Nitric Oxide-cGMP and Nrf2/ARE/HO-1 Pathways and Attenuates Oxidative Stress and Hematological Alterations in Hyperammonemic Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7369671. [PMID: 28744340 PMCID: PMC5506469 DOI: 10.1155/2017/7369671] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/21/2017] [Accepted: 05/28/2017] [Indexed: 12/11/2022]
Abstract
Hyperammonemia is a serious complication of liver disease and may lead to encephalopathy and death. This study investigated the effects of Commiphora molmol resin on oxidative stress, inflammation, and hematological alterations in ammonium chloride- (NH4Cl-) induced hyperammonemic rats, with an emphasis on the glutamate-NO-cGMP and Nrf2/ARE/HO-1 signaling pathways. Rats received NH4Cl and C. molmol for 8 weeks. NH4Cl-induced rats showed significant increase in blood ammonia, liver function markers, and tumor necrosis factor-alpha (TNF-α). Concurrent supplementation of C. molmol significantly decreased circulating ammonia, liver function markers, and TNF-α in hyperammonemic rats. C. molmol suppressed lipid peroxidation and nitric oxide and enhanced the antioxidant defenses in the liver, kidney, and cerebrum of hyperammonemic rats. C. molmol significantly upregulated Nrf2 and HO-1 and decreased glutamine and nitric oxide synthase, soluble guanylate cyclase, and Na+/K+-ATPase expression in the cerebrum of NH4Cl-induced hyperammonemic rats. Hyperammonemia was also associated with hematological and coagulation system alterations. These alterations were reversed by C. molmol. Our findings demonstrated that C. molmol attenuates ammonia-induced liver injury, oxidative stress, inflammation, and hematological alterations. This study points to the modulatory effect of C. molmol on glutamate-NO-cGMP and Nrf2/ARE/HO-1 pathways in hyperammonemia. Therefore, C. molmol might be a promising protective agent against hyperammonemia.
Collapse
|
11
|
Ramakrishnan A, Vijayakumar N, Renuka M. Naringin regulates glutamate-nitric oxide cGMP pathway in ammonium chloride induced neurotoxicity. Biomed Pharmacother 2016; 84:1717-1726. [PMID: 27836465 DOI: 10.1016/j.biopha.2016.10.080] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 11/15/2022] Open
Abstract
Naringin, plant bioflavonoid extracted mainly from grapefruit and other related citrus species. This study was designed to assess the neuroprotective effect of naringin on ammonium chloride (NH4Cl) induced hyperammonemic rats. Experimental hyperammonemia was induced by intraperitonial injection (i.p) of NH4Cl (100mg/kg body weight (b.w.)) thrice a week for 8 consecutive weeks. Hyperammonemic rats were treated with naringin (80mg/kg b.w.) via oral gavage. Naringin administration drastically restored the levels of blood ammonia, plasma urea, nitric oxide (NO), glutamate, glutamine, lipid peroxidation, lipid profile, activities of liver marker enzymes, antioxidant status and sodium/potassium-ATPase (Na+/K+-ATPase). In addition, naringin supplementation reverted back the pathological changes of liver, brain and kidney tissues, the expressions of Glutamine synthetase (GS), Na+/K+-ATPase, neuronal nitric oxide (nNOS) and soluble guanylate cyclase (sGC) in hyperammonemic rats. Hence, this study suggested that nargingin exhibited their protective effect against NH4Cl induced toxicity via enhancing the activities of antioxidant enzymes and inhibiting the lipid peroxidation process. Take together, this study provides data that naingin effectively reduced neurotoxicity by attenuating hyperammonemia, suggesting that naringin act as a potential therapeutic agent to treat hyperammonemic rats.
Collapse
Affiliation(s)
- Arumugam Ramakrishnan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, India
| | - Natesan Vijayakumar
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, India.
| | - Mani Renuka
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, India
| |
Collapse
|
12
|
Abstract
The three essential branched-chain amino acids (BCAAs), leucine, isoleucine and valine, share the first enzymatic steps in their metabolic pathways, including a reversible transamination followed by an irreversible oxidative decarboxylation to coenzyme-A derivatives. The respective oxidative pathways subsequently diverge and at the final steps yield acetyl- and/or propionyl-CoA that enter the Krebs cycle. Many disorders in these pathways are diagnosed through expanded newborn screening by tandem mass spectrometry. Maple syrup urine disease (MSUD) is the only disorder of the group that is associated with elevated body fluid levels of the BCAAs. Due to the irreversible oxidative decarboxylation step distal enzymatic blocks in the pathways do not result in the accumulation of amino acids, but rather to CoA-activated small carboxylic acids identified by gas chromatography mass spectrometry analysis of urine and are therefore classified as organic acidurias. Disorders in these pathways can present with a neonatal onset severe-, or chronic intermittent- or progressive forms. Metabolic instability and increased morbidity and mortality are shared between inborn errors in the BCAA pathways, while treatment options remain limited, comprised mainly of dietary management and in some cases solid organ transplantation.
Collapse
Affiliation(s)
- I Manoli
- Organic Acid Research Section, Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - C P Venditti
- Organic Acid Research Section, Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
13
|
Zandberg L, van Dyk HC, van der Westhuizen FH, van Dijk AA. A 3-methylcrotonyl-CoA carboxylase deficient human skin fibroblast transcriptome reveals underlying mitochondrial dysfunction and oxidative stress. Int J Biochem Cell Biol 2016; 78:116-129. [PMID: 27417235 DOI: 10.1016/j.biocel.2016.07.010] [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: 04/17/2015] [Revised: 07/07/2016] [Accepted: 07/11/2016] [Indexed: 01/03/2023]
Abstract
Isolated 3-methylcrotonyl-CoA carboxylase (MCC) deficiency is an autosomal recessive inherited metabolic disease of leucine catabolism with a highly variable phenotype. Apart from extensive mutation analyses of the MCCC1 and MCCC2 genes encoding 3-methylcrotonyl-CoA carboxylase (EC 6.4.1.4), molecular data on MCC deficiency gene expression studies in human tissues is lacking. For IEMs, unbiased '-omics' approaches are starting to reveal the secondary cellular responses to defects in biochemical pathways. Here we present the first whole genome expression profile of immortalized cultured skin fibroblast cells of two clinically affected MCC deficient patients and two healthy individuals generated using Affymetrix(®)HuExST1.0 arrays. There were 16191 significantly differentially expressed transcript IDs of which 3591 were well annotated and present in the predefined knowledge database of Ingenuity Pathway Analysis software used for downstream functional analyses. The most noticeable feature of this MCCA deficient skin fibroblast transcriptome was the typical genetic hallmark of mitochondrial dysfunction, decreased antioxidant response and disruption of energy homeostasis, which was confirmed by mitochondrial functional analyses. The MCC deficient transcriptome seems to predict oxidative stress that could alter the complex secondary cellular response that involve genes of the glycolysis, the TCA cycle, OXPHOS, gluconeogenesis, β-oxidation and the branched-chain fatty acid metabolism. An important emerging insight from this human MCCA transcriptome in combination with previous reports is that chronic exposure to the primary and secondary metabolites of MCC deficiency and the resulting oxidative stress might impact adversely on the quality of life and energy levels, irrespective of whether MCC deficient individuals are clinically affected or asymptomatic.
Collapse
Affiliation(s)
- L Zandberg
- Biochemistry Division, Centre for Human Metabolomics, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - H C van Dyk
- Biochemistry Division, Centre for Human Metabolomics, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - F H van der Westhuizen
- Biochemistry Division, Centre for Human Metabolomics, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - A A van Dijk
- Biochemistry Division, Centre for Human Metabolomics, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa.
| |
Collapse
|
14
|
Experimental Evidence that 3-Methylglutaric Acid Disturbs Mitochondrial Function and Induced Oxidative Stress in Rat Brain Synaptosomes: New Converging Mechanisms. Neurochem Res 2016; 41:2619-2626. [DOI: 10.1007/s11064-016-1973-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/23/2016] [Accepted: 06/01/2016] [Indexed: 12/14/2022]
|
15
|
Sorghum stem extract modulates Na+/K+-ATPase, ecto-5′-nucleotidase, and acetylcholinesterase activities. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s00580-016-2259-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
16
|
da Rosa MS, Scaini G, Damiani AP, Longaretti LM, Pereira M, Seminotti B, Zapelini HG, Schuck PF, Streck EL, de Andrade VM, Wajner M, Leipnitz G. Evidence that 3-hydroxy-3-methylglutaric and 3-methylglutaric acids induce DNA damage in rat striatum. Metab Brain Dis 2015; 30:1055-62. [PMID: 25939283 DOI: 10.1007/s11011-015-9675-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 04/23/2015] [Indexed: 01/10/2023]
Abstract
3-Hydroxy-3-methylglutaryl-CoA lyase (HL) deficiency is a rare autosomal recessive disorderaffecting the final step of leucine degradation and ketogenesis and biochemically characterized by the predominant accumulation of 3-hydroxy-3-methylglutaric (HMG) and 3-methylglutaric (MGA) acids in biological fluids and tissues of affected patients. Considering that previous studies reported that HMG and MGA have pro oxidant properties, the present study evaluated the ex vivo and in vitro effects of HMG and MGA on frequency and index of DNA damage in cerebral cortex and striatum of young rats. The ex vivo effects of both organic acids on 8-hydroxy-2'-deoxyguanosine (OHdG) levels and their in vitro effects on 2',7'-dichlorofluorescin (DCFH) oxidation and glutathione (GSH) concentrations in rat striatum were also determined. We also investigated the ex vivo effects of both organic acids on 8-hydroxy-2'-deoxyguanosine (OHdG) levels in rat striatum. In the ex vivo experiments, DNA damage was determined in striatum homogenates prepared 30 min after a single intrastriatal administration of HMG or MGA. On the other hand, the in vitro evaluation was performed after an incubation of rat cerebral cortex or striatum homogenates or slices in the presence of HMG or MGA during 1 h at 37 °C. We observed that the intrastriatal administration of HMG and MGA increased the frequency and the index of DNA damage, as well as OHdG staining in rat striatum. We also verified that MGA, but not HMG, increased DNA damage frequency and index in vitro in striatum of rats. In contrast, no alterations were verified in vitro in cerebral cortex. Finally, we found that HMG and MGA increased DCFH oxidation and decreased GSH concentrations in rat striatum. Therefore, it may be presumed that DNA damage provoked by HMG and MGA possibly via reactive species generation is involved, at least in part, in the pathophysiology of brain injury, particularly in the striatum of HL-deficient patients.
Collapse
Affiliation(s)
- Mateus Struecker da Rosa
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos N° 2600 - Attached, CEP 90035-003, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
NMDA Receptors and Oxidative Stress Induced by the Major Metabolites Accumulating in HMG Lyase Deficiency Mediate Hypophosphorylation of Cytoskeletal Proteins in Brain From Adolescent Rats: Potential Mechanisms Contributing to the Neuropathology of This Disease. Neurotox Res 2015; 28:239-52. [PMID: 26174040 DOI: 10.1007/s12640-015-9542-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/01/2015] [Accepted: 07/07/2015] [Indexed: 01/01/2023]
Abstract
Neurological symptoms and cerebral abnormalities are commonly observed in patients with 3-hydroxy-3-methylglutaryl-CoA lyase (HMG lyase) deficiency, which is biochemically characterized by predominant tissue accumulation of 3-hydroxy-3-methylglutaric (HMG), 3-methylglutaric (MGA), and 3-methylglutaconic (MGT) acids. Since the pathogenesis of this disease is poorly known, the present study evaluated the effects of these compounds on the cytoskeleton phosphorylating system in rat brain. HMG, MGA, and MGT caused hypophosphorylation of glial fibrillary acidic protein (GFAP) and of the neurofilament subunits NFL, NFM, and NFH. HMG-induced hypophosphorylation was mediated by inhibiting the cAMP-dependent protein kinase (PKA) on Ser55 residue of NFL and c-Jun kinase (JNK) by acting on KSP repeats of NFM and NFH subunits. We also evidenced that the subunit NR2B of NMDA receptor and Ca(2+) was involved in HMG-elicited hypophosphorylation of cytoskeletal proteins. Furthermore, the antioxidants L-NAME and TROLOX fully prevented both the hypophosphorylation and the inhibition of PKA and JNK caused by HMG, suggesting that oxidative damage may underlie these effects. These findings indicate that the main metabolites accumulating in HMG lyase deficiency provoke hypophosphorylation of cytoskeleton neural proteins with the involvement of NMDA receptors, Ca(2+), and reactive species. It is presumed that these alterations may contribute to the neuropathology of this disease.
Collapse
|
18
|
Fernandes CG, Rodrigues MDN, Seminotti B, Colín-González AL, Santamaria A, Quincozes-Santos A, Wajner M. Induction of a Proinflammatory Response in Cortical Astrocytes by the Major Metabolites Accumulating in HMG-CoA Lyase Deficiency: the Role of ERK Signaling Pathway in Cytokine Release. Mol Neurobiol 2015; 53:3586-3595. [PMID: 26099308 DOI: 10.1007/s12035-015-9289-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 06/04/2015] [Indexed: 12/19/2022]
Abstract
3-Hydroxy-3-methylglutaric aciduria (HMGA) is an inherited metabolic disorder caused by 3-hydroxy-3-methylglutaryl-CoA lyase deficiency. It is biochemically characterized by predominant tissue accumulation and high urinary excretion of 3-hydroxy-3-methylglutarate (HMG) and 3-methylglutarate (MGA). Affected patients commonly present acute symptoms during metabolic decompensation, including vomiting, seizures, and lethargy/coma accompanied by metabolic acidosis and hypoketotic hypoglycemia. Although neurological manifestations are common, the pathogenesis of brain injury in this disease is poorly known. Astrocytes are important for neuronal protection and are susceptible to damage by neurotoxins. In the present study, we investigated the effects of HMG and MGA on important parameters of redox homeostasis and cytokine production in cortical cultured astrocytes. The role of the metabolites on astrocyte mitochondrial function (thiazolyl blue tetrazolium bromide (MTT) reduction) and viability (propidium iodide incorporation) was also studied. Both organic acids decreased astrocytic mitochondrial function and the concentrations of reduced glutathione without altering cell viability. In contrast, they increased reactive species formation (2'-7'-dichlorofluorescein diacetate (DCFHDA) oxidation), as well as IL-1β, IL-6, and TNF α release through the ERK signaling pathway. Taken together, the data indicate that the principal compounds accumulating in HMGA induce a proinflammatory response in cultured astrocytes that may possibly be involved in the neuropathology of this disease.
Collapse
Affiliation(s)
- Carolina Gonçalves Fernandes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600 - Anexo, 90035-003, Porto Alegre, RS, Brazil
| | - Marília Danyelle Nunes Rodrigues
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600 - Anexo, 90035-003, Porto Alegre, RS, Brazil
| | - Bianca Seminotti
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600 - Anexo, 90035-003, Porto Alegre, RS, Brazil
| | - Ana Laura Colín-González
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, SSA, Mexico City, Mexico
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, SSA, Mexico City, Mexico
| | - André Quincozes-Santos
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600 - Anexo, 90035-003, Porto Alegre, RS, Brazil
| | - Moacir Wajner
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600 - Anexo, 90035-003, Porto Alegre, RS, Brazil.
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| |
Collapse
|
19
|
In vitro evaluation of neurotoxicity potential and oxidative stress responses of diazinon and its degradation products in rat brain synaptosomes. Toxicol Lett 2015; 233:29-37. [DOI: 10.1016/j.toxlet.2015.01.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 12/30/2014] [Accepted: 01/02/2015] [Indexed: 11/22/2022]
|
20
|
Dos Santos Mello M, Ribas GS, Wayhs CAY, Hammerschmidt T, Guerreiro GBB, Favenzani JL, Sitta Â, de Moura Coelho D, Wajner M, Vargas CR. Increased oxidative stress in patients with 3-hydroxy-3-methylglutaric aciduria. Mol Cell Biochem 2015; 402:149-55. [PMID: 25557019 DOI: 10.1007/s11010-014-2322-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/23/2014] [Indexed: 02/07/2023]
Abstract
3-hydroxy-3-methylglutaric aciduria (HMGA; OMIM 246450) is a rare autosomal recessive disorder, caused by the deficiency of 3-hydroxy-3-methylglutaryl-CoA lyase (4.1.3.4), which results in the accumulation of 3-hydroxy-3-methylglutaric (HMG) and 3-methylglutaric (MGA) acids in tissues and biological fluids of affected individuals. Recent in vivo and in vitro animal studies have demonstrated that the accumulation of these metabolites can disturb the cellular redox homeostasis, which can contribute to the neurological manifestations presented by the patients. So, in the present work, we investigated oxidative stress parameters in plasma and urine samples from HMGA patients, obtained at the moment of diagnosis of this disorder and during therapy with low-protein diet and L-carnitine supplementation. It was verified that untreated HMGA patients presented higher levels of urinary di-tyrosine and plasma thiobarbituric acid-reactive substances (TBA-RS), which are markers of protein and lipid oxidative damage, respectively, as well as a reduction of the urinary antioxidant capacity. Treated HMGA patients also presented an increased protein oxidative damage, as demonstrated by their higher concentrations of plasma protein carbonyl groups and urinary di-tyrosine, as well as by the reduction of total sulfhydryl groups in plasma, in relation to controls. On the other hand, HMGA patients under therapy presented normal levels of TBA-RS and urinary antioxidant capacity, which can be related, at least in part, to the antioxidant and antiperoxidative effects exerted by L-carnitine. The results of this work are the first report showing that a redox imbalance occurs in patients with HMGA what reinforces the importance of the antioxidant therapy in this disorder.
Collapse
Affiliation(s)
- Mariana Dos Santos Mello
- Programa de Pós-Graduação Ciências Farmacêuticas da Universidade Federal do Rio Grande do Sul (UFRGS), Ipiranga 2752, Porto Alegre, RS, 90610-000, Brazil,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Evidence that glycine induces lipid peroxidation and decreases glutathione concentrations in rat cerebellum. Mol Cell Biochem 2014; 395:125-34. [PMID: 24939360 DOI: 10.1007/s11010-014-2118-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 06/02/2014] [Indexed: 02/07/2023]
Abstract
Patients with non-ketotic hyperglycinemia (NKH) present severe neurological symptoms and brain abnormalities involving cerebellum. Although the pathomechanisms underlying the cerebellum damage have not been studied, high tissue levels of glycine (GLY), the biochemical hallmark of this disorder have been suggested to contribute to the neuropathology of this disease. We investigated the in vitro effects of GLY on important parameters of oxidative stress and energy metabolism in cerebellum of 30-day-old rats. Our results show that GLY increased 2',7'-dichlorofluorescin oxidation, suggesting that reactive species production are augmented by GLY in the cerebellum. However, hydrogen peroxide generation was not altered by GLY. GLY also increased thiobarbituric acid-reactive substances (TBA-RS) levels and reduced the glutathione (GSH) content, indicating that this amino acid provokes lipid oxidative damage and compromises the non-enzymatic antioxidant defenses, respectively, in cerebellum. The antioxidants melatonin and trolox (the hydrosoluble analog of vitamin E) prevented the GLY-induced increase of TBA-RS and decrease of GSH in cerebellum, indicating the involvement of hydroxyl and peroxyl radicals in these effects. The NMDA receptor antagonist MK-801 also attenuated GLY-induced decrease of GSH, suggesting that this effect is mediated through NMDA receptor. In contrast, GLY did not alter the protein carbonyl formation and total and protein-bound sulfhydryl group content, as well as catalase and superoxide dismutase activities. Furthermore, GLY did not alter the activities of the respiratory chain complexes and creatine kinase. Our present data indicate that oxidative stress elicited by GLY in vitro may be a potential pathomechanism involved in the cerebellar dysfunction observed in NKH.
Collapse
|
22
|
da Rosa MS, Seminotti B, Amaral AU, Fernandes CG, Gasparotto J, Moreira JCF, Gelain DP, Wajner M, Leipnitz G. Redox homeostasis is compromised in vivo by the metabolites accumulating in 3-hydroxy-3-methylglutaryl-CoA lyase deficiency in rat cerebral cortex and liver. Free Radic Res 2013; 47:1066-75. [DOI: 10.3109/10715762.2013.853876] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
23
|
Rauchová H, Vokurková M, Koudelová J. Hypoxia-induced lipid peroxidation in the brain during postnatal ontogenesis. Physiol Res 2013; 61:S89-101. [PMID: 22827877 DOI: 10.33549/physiolres.932374] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Reactive oxygen species (ROS) are common products of the physiological metabolic reactions, which are associated with cell signaling and with the pathogenesis of various nervous disorders. The brain tissue has the high rate of oxidative metabolic activity, high concentration of polyunsaturated fatty acids in membrane lipids, presence of iron ions and low capacity of antioxidant enzymes, which makes the brain very susceptible to ROS action and lipid peroxidation formation. Membranes of brain cortex show a higher production of thiobarbituric acid-reactive substances (TBARS) in prooxidant system (ADP.Fe(3+)/NADPH) than membranes from the heart or kidney. Lipid peroxidation influences numerous cellular functions through membrane-bound receptors or enzymes. The rate of brain cortex Na(+),K(+)-ATPase inhibition correlates well with the increase of TBARS or conjugated dienes and with changes of membrane fluidity. The experimental model of short-term hypoxia (simulating an altitude of 9000 m for 30 min) shows remarkable increase in TBARS in four different parts of the rat brain (cortex, subcortical structures, cerebellum and medulla oblongata) during the postnatal development of Wistar rat of both sexes. Young rats and males are more sensitive to oxygen changes than adult rats and females, respectively. Under normoxia or hypobaric hypoxia both ontogenetic aspects and sex differences play a major role in establishing the activity of erythrocyte catalase, which is an important part of the antioxidant defense of the organism. Rats pretreated with L-carnitine (and its derivatives) have lower TBARS levels after the exposure to hypobaric hypoxia. The protective effect of L-carnitine is comparable with the effect of tocopherol, well-known reactive species scavenger. Moreover, the plasma lactate increases after a short-term hypobaric hypoxia and decreases in L-carnitine pretreated rats. Acute hypobaric hypoxia and/or L-carnitine-pretreatment modify serum but not brain lactate dehydrogenase activity. The obtained data seem to be important because the variations in oxygen tension represent specific signals of regulating the activity of many specific systems in the organism.
Collapse
Affiliation(s)
- H Rauchová
- Centre for Cardiovascular Research, Prague, Czech Republic.
| | | | | |
Collapse
|
24
|
Zanatta A, Viegas CM, Tonin AM, Busanello ENB, Grings M, Moura AP, Leipnitz G, Wajner M. Disturbance of redox homeostasis by ornithine and homocitrulline in rat cerebellum: a possible mechanism of cerebellar dysfunction in HHH syndrome. Life Sci 2013; 93:161-8. [PMID: 23806752 DOI: 10.1016/j.lfs.2013.06.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/23/2013] [Accepted: 06/10/2013] [Indexed: 12/14/2022]
Abstract
AIMS Cerebellar ataxia is commonly observed in hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome, an inherited metabolic disorder biochemically characterized by ornithine (Orn), homocitrulline (Hcit) and ammonia accumulation. Since the pathophysiology of cerebellum damage in this disorder is still unknown, we investigated the effects of Hcit and Orn on important parameters of redox and energy homeostasis in cerebellum of young rats. MATERIAL AND METHODS We determined thiobarbituric acid-reactive substance (TBA-RS) levels, carbonyl content, nitrate and nitrite production, hydrogen peroxide production, GSH concentrations, sulfhydryl content, as well as activities of respiratory chain complexes I-IV, creatine kinase, Na(+),K(+)-ATPase, aconitase and α-ketoglutarate dehydrogenase. KEY FINDINGS Orn and Hcit significantly increased TBA-RS levels (lipid oxidation), that was totally prevented by melatonin and reduced glutathione (GSH). We also found that nitrate and nitrite production was not altered by any of the metabolites, in contrast to hydrogen peroxide production which was significantly enhanced by Hcit. Furthermore, GSH concentrations were significantly reduced by Orn and Hcit and sulfhydryl content by Orn, implying an impairment of antioxidant defenses. As regards energy metabolism, Orn and Hcit provoked a significant reduction of aconitase activity, without altering the other parameters. Furthermore, Orn-elicited reduction of aconitase activity was totally prevented by GSH, indicating that the critical groups of this enzyme were susceptible to oxidation caused by this amino acid. SIGNIFICANCE Taken together, our data indicate that redox homeostasis is disturbed by the major metabolites accumulating in HHH syndrome and that this mechanism may be implicated in the ataxia and cerebellar abnormalities observed in this disorder.
Collapse
Affiliation(s)
- Angela Zanatta
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Fernandes CG, da Rosa MS, Seminotti B, Pierozan P, Martell RW, Lagranha VL, Busanello ENB, Leipnitz G, Wajner M. In vivo experimental evidence that the major metabolites accumulating in 3-hydroxy-3-methylglutaryl-CoA lyase deficiency induce oxidative stress in striatum of developing rats: a potential pathophysiological mechanism of striatal damage in this disorder. Mol Genet Metab 2013; 109:144-53. [PMID: 23611578 DOI: 10.1016/j.ymgme.2013.03.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 03/29/2013] [Accepted: 03/29/2013] [Indexed: 12/24/2022]
Abstract
3-Hydroxy-3-methylglutaryl-CoA lyase (HL) deficiency is a genetic disorder biochemically characterized by predominant accumulation of 3-hydroxy-3-methylglutaric (HMG) and 3-methylglutaric (MGA) acids in tissues and biological fluids of affected individuals. Clinically, the patients present neurological symptoms and basal ganglia injury, whose pathomechanisms are partially understood. In the present study, we investigated the ex vivo effects of intrastriatal administration of HMG and MGA on important parameters of oxidative stress in striatum of developing rats. Our results demonstrate that HMG and MGA induce lipid and protein oxidative damage. HMG and MGA also increased 2',7'-dichlorofluorescein oxidation, whereas only HMG elicited nitric oxide production, indicating a role for reactive oxygen (HMG and MGA) and nitrogen (HMG) species in these effects. Regarding the enzymatic antioxidant defenses, both organic acids decreased reduced glutathione concentrations and the activities of superoxide dismutase and glutathione reductase and increased glutathione peroxidase activity. HMG also provoked an increase of catalase activity and a diminution of glucose-6-phosphate dehydrogenase activity. We finally observed that antioxidants fully prevented or attenuated HMG-induced alterations of the oxidative stress parameters, further indicating the participation of reactive species in these effects. We also observed that MK-801, a non-competitive antagonist of the N-methyl-D-aspartate (NMDA) receptor, prevented some of these effects, indicating the involvement of the NMDA receptor in HMG effects. The present data provide solid evidence that oxidative stress is induced in vivo by HMG and MGA in rat striatum and it is presumed that this pathomechanism may explain, at least in part, the cerebral alterations observed in HL deficiency.
Collapse
Affiliation(s)
- Carolina Gonçalves Fernandes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sood P, Priyadarshini S, Aich P. Estimation of psychological stress in humans: a combination of theory and practice. PLoS One 2013; 8:e63044. [PMID: 23690978 PMCID: PMC3654918 DOI: 10.1371/journal.pone.0063044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 03/28/2013] [Indexed: 02/07/2023] Open
Abstract
Stress has long been known to increase susceptibility to health disorders. In 2009, American Psychological Association further established association of stress to serious health problems. However, a quantitative and accurate way to evaluate and estimate stress status of individuals is still a big challenge. It has been shown, in large animal models using cattle, that psychological stress can be quantified as well as disease susceptibility could be predicted through biomarker discovery. Taking cue from those studies, we have evaluated and estimated psychological stress level of individuals theoretically and validated experimentally. Various biomarkers have also been identified which can be associated to psychological stress to predict stress status of unknown individuals.
Collapse
Affiliation(s)
- Parul Sood
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Institute of Physics Campus, Sachivalaya Marg, Odisha, India
| | - Sushri Priyadarshini
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Institute of Physics Campus, Sachivalaya Marg, Odisha, India
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Institute of Physics Campus, Sachivalaya Marg, Odisha, India
| |
Collapse
|
27
|
Zanatta Â, Moura AP, Tonin AM, Knebel LA, Grings M, Lobato VA, Ribeiro CAJ, Dutra-Filho CS, Leipnitz G, Wajner M. Neurochemical evidence that the metabolites accumulating in 3-methylcrotonyl-CoA carboxylase deficiency induce oxidative damage in cerebral cortex of young rats. Cell Mol Neurobiol 2013; 33:137-46. [PMID: 23053545 PMCID: PMC11497930 DOI: 10.1007/s10571-012-9879-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 09/13/2012] [Indexed: 12/13/2022]
Abstract
Isolated 3-methylcrotonyl-CoA carboxylase deficiency (3MCCD) is an autosomal recessive disorder of leucine metabolism biochemically characterized by accumulation of 3-methylcrotonylglycine (3MCG), 3-methylcrotonic acid (3MCA) and 3-hydroxyisovaleric acid. A considerable number of affected individuals present neurological symptoms with or without precedent crises of metabolic decompensation and brain abnormalities whose pathogenesis is poorly known. We investigated the in vitro effects of 3MCG and 3MCA on important parameters of oxidative stress in cerebral cortex of young rats. 3MCG and 3MCA significantly increased TBA-RS and carbonyl formation, indicating that these compounds provoke lipid and protein oxidation, respectively. In contrast, nitric oxide production was not affected by 3MCG and 3MCA. Furthermore, 3MCG- and 3MCA-induced elevation of TBA-RS values was fully prevented by melatonin, trolox and reduced glutathione, but not by the nitric oxide inhibitor N(ω)-nitro-L-arginine methyl ester or the combination of catalase plus superoxide dismutase, indicating that reactive oxygen species were involved in the oxidative damage caused by these compounds. We also found that the activity of the antioxidant enzymes glutathione peroxidase, catalase, superoxide dismutase and glutathione reductase were not altered in vitro by 3MCG and 3MCA. It is therefore presumed that alterations of the cellular redox homeostasis caused by the major metabolites accumulating in 3MCCD may potentially be involved in the pathophysiology of the neurological dysfunction and structural brain alterations found in patients affected by this disorder.
Collapse
Affiliation(s)
- Ângela Zanatta
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600-Anexo, Porto Alegre, RS, 90035-003 Brazil
| | - Alana Pimentel Moura
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600-Anexo, Porto Alegre, RS, 90035-003 Brazil
| | - Anelise Miotti Tonin
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600-Anexo, Porto Alegre, RS, 90035-003 Brazil
| | - Lisiane Aurélio Knebel
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600-Anexo, Porto Alegre, RS, 90035-003 Brazil
| | - Mateus Grings
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600-Anexo, Porto Alegre, RS, 90035-003 Brazil
| | - Vannessa Araújo Lobato
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600-Anexo, Porto Alegre, RS, 90035-003 Brazil
| | - César Augusto João Ribeiro
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600-Anexo, Porto Alegre, RS, 90035-003 Brazil
| | - Carlos Severo Dutra-Filho
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600-Anexo, Porto Alegre, RS, 90035-003 Brazil
| | - Guilhian Leipnitz
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600-Anexo, Porto Alegre, RS, 90035-003 Brazil
| | - Moacir Wajner
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos No 2600-Anexo, Porto Alegre, RS, 90035-003 Brazil
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|