1
|
Seminotti B, Brondani M, Ribeiro RT, Leipnitz G, Wajner M. Disturbance of Mitochondrial Dynamics, Endoplasmic Reticulum-Mitochondria Crosstalk, Redox Homeostasis, and Inflammatory Response in the Brain of Glutaryl-CoA Dehydrogenase-Deficient Mice: Neuroprotective Effects of Bezafibrate. Mol Neurobiol 2022; 59:4839-4853. [PMID: 35639256 DOI: 10.1007/s12035-022-02887-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/17/2022] [Indexed: 11/26/2022]
Abstract
Patients with glutaric aciduria type 1 (GA1), a neurometabolic disorder caused by deficiency of glutaryl-CoA dehydrogenase (GCDH) activity, commonly manifest acute encephalopathy associated with severe striatum degeneration and progressive cortical and striatal injury whose pathogenesis is still poorly known. We evaluated redox homeostasis, inflammatory response, mitochondrial biogenesis and dynamics, endoplasmic reticulum (ER)-mitochondria crosstalk, and ER stress in the brain of GCDH-deficient (Gcdh-/-) and wild-type (Gcdh+/+) mice fed a high Lys chow, which better mimics the human neuropathology mainly characterized by striatal lesions. Increased lipid peroxidation and altered antioxidant defenses, including decreased concentrations of reduced glutathione and increased activities of superoxide dismutase, catalase, and glutathione transferase, were observed in the striatum and cerebral cortex of Gcdh-/- mice. Augmented Iba-1 staining was also found in the dorsal striatum and neocortex, whereas the nuclear content of NF-κB was increased, and the cytosolic content of IκBα decreased in the striatum of the mutant animals, indicating a pro-inflammatory response. Noteworthy, in vivo treatment with the pan-PPAR agonist bezafibrate normalized these alterations. It was also observed that the ER-mitochondria crosstalk proteins VDAC1 and IP3R were reduced, whereas the ER stress protein DDIT3 was augmented in Gcdh-/- striatum, signaling disturbances of these processes. Finally, DRP1 content was elevated in the striatum of Gcdh-/- mice, indicating activated mitochondrial fission. We presume that some of these novel pathomechanisms may be involved in GA1 neuropathology and that bezafibrate should be tested as a potential adjuvant therapy for GA1.
Collapse
Affiliation(s)
- Bianca Seminotti
- 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
| | - Morgana Brondani
- 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
| | - Rafael Teixeira Ribeiro
- 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
| | - Guilhian Leipnitz
- 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
- Programa de Pós-Graduação em Fisiologia, 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, Rua Ramiro Barcelos, 2600, Prédio 21111, Porto Alegre, RS, 90035-003, Brazil
| | - 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, Rua Ramiro Barcelos, 2600, Prédio 21111, Porto Alegre, RS, 90035-003, Brazil.
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil.
| |
Collapse
|
2
|
Nonose Y, Pieper LZ, da Silva JS, Longoni A, Apel RV, Meira-Martins LA, Grings M, Leipnitz G, Souza DO, de Assis AM. Guanosine enhances glutamate uptake and oxidation, preventing oxidative stress in mouse hippocampal slices submitted to high glutamate levels. Brain Res 2020; 1748:147080. [PMID: 32866546 DOI: 10.1016/j.brainres.2020.147080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/31/2020] [Accepted: 08/21/2020] [Indexed: 01/01/2023]
Abstract
Glutamate (Glu) is the main mammalian brain neurotransmitter. Concerning the glutamatergic neurotransmission, excessive levels of glutamate in the synaptic cleft are extremally harmful. This phenomenon, named as excitotoxicity is involved in various acute and chronic brain diseases. Guanosine (GUO), an endogenous guanine nucleoside, possesses neuroprotective effects in several experimental models of glutamatergic excitotoxicity, an effect accompanied by an increase in astrocytic glutamate uptake. Therefore, the objective of this study was to investigate the involvement of an additional putative parameter, glutamate oxidation to CO2, involved in ex-vivo GUO neuroprotective effects in mouse hippocampal slices submitted to glutamatergic excitotoxicity. Mice were sacrificed by decapitation, the hippocampi were removed and sliced. The slices were incubated for various times and concentrations of Glu and GUO. First, the concentration of Glu that produced an increase in L-[14C(U)]-Glu oxidation to CO2 without cell injury was determined at different time points (between 0 and 90 min); 1000 μM Glu increased Glu oxidation between 30 and 60 min of incubation without cell injury. Under these conditions (Glu concentration and incubation time), 100 μM GUO increased Glu oxidation (35%). Additionally, 100 μM GUO increased L-[3,4-3H]-glutamate uptake (45%) in slices incubated with 1000 μM Glu (0-30 min). Furthermore, 1000 μM Glu increased reactive species levels, SOD activity, and decreased GPx activity, and GSH content after 30 and 60 min; 100 μM GUO prevented these effects. This is the first study demonstrating that GUO simultaneously promoted an increase in the uptake and utilization of Glu in excitotoxicity-like conditions preventing redox imbalance.
Collapse
Affiliation(s)
- Y Nonose
- Graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil
| | - L Z Pieper
- Graduate Program in Health and Behavior, Center of Health Science, Universidade Católica de Pelotas - UCPel, Pelotas, RS 96015-560, Brazil
| | - J S da Silva
- Graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil
| | - A Longoni
- Graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil; Graduate Program in Health and Behavior, Center of Health Science, Universidade Católica de Pelotas - UCPel, Pelotas, RS 96015-560, Brazil
| | - R V Apel
- Graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil
| | - L A Meira-Martins
- Graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil
| | - M Grings
- Graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil
| | - G Leipnitz
- Graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil
| | - D O Souza
- Graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil.
| | - A M de Assis
- Graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS 90035-003, Brazil; Graduate Program in Health and Behavior, Center of Health Science, Universidade Católica de Pelotas - UCPel, Pelotas, RS 96015-560, Brazil
| |
Collapse
|
3
|
Odorcyk FK, Duran-Carabali LE, Rocha DS, Sanches EF, Martini AP, Venturin GT, Greggio S, da Costa JC, Kucharski LC, Zimmer ER, Netto CA. Differential glucose and beta-hydroxybutyrate metabolism confers an intrinsic neuroprotection to the immature brain in a rat model of neonatal hypoxia ischemia. Exp Neurol 2020; 330:113317. [PMID: 32304750 DOI: 10.1016/j.expneurol.2020.113317] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022]
Abstract
Neonatal hypoxia ischemia (HI) is the main cause of newborn mortality and morbidity. Preclinical studies have shown that the immature rat brain is more resilient to HI injury, suggesting innate mechanisms of neuroprotection. During neonatal period brain metabolism experience changes that might greatly affect the outcome of HI injury. Therefore, the aim of the present study was to investigate how changes in brain metabolism interfere with HI outcome in different stages of CNS development. For this purpose, animals were divided into 6 groups: HIP3, HIP7 and HIP11 (HI performed at postnatal days 3, 7 and 11, respectively), and their respective shams. In vivo [18F]FDG micro positron emission tomography (microPET) imaging was performed 24 and 72 h after HI, as well as ex-vivo assessments of glucose and beta-hydroxybutyrate (BHB) oxidation. At adulthood behavioral tests and histology were performed. Behavioral and histological analysis showed greater impairments in HIP11 animals, while HIP3 rats were not affected. Changes in [18F]FDG metabolism were found only in the lesion area of HIP11, where a substantial hypometabolism was detected. Furthermore, [18F]FDG hypometabolism predicted impaired cognition and worst histological outcomes at adulthood. Finally, substrate oxidation assessments showed that glucose oxidation remained unaltered and higher level of BHB oxidation found in P3 animals, suggesting a more resilient metabolism. Overall, present results show [18F]FDG microPET predicts long-term injury outcome and suggests that higher BHB utilization is one of the mechanisms that confer the intrinsic neuroprotection to the immature brain and should be explored as a therapeutic target for treatment of HI.
Collapse
Affiliation(s)
- F K Odorcyk
- Graduate Program in Phisiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - L E Duran-Carabali
- Graduate Program in Phisiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - D S Rocha
- Graduate Program in Phisiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - E F Sanches
- Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - A P Martini
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - G T Venturin
- Preclinical Research Center, Brain Institute (BraIns) of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - S Greggio
- Preclinical Research Center, Brain Institute (BraIns) of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - J C da Costa
- Preclinical Research Center, Brain Institute (BraIns) of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - L C Kucharski
- Graduate Program in Phisiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - E R Zimmer
- Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Graduate Program in Pharmacology and therapeutics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Department of Pharmacology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - C A Netto
- Graduate Program in Phisiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| |
Collapse
|
4
|
Guazzelli PA, Cittolin-Santos GF, Meira-Martins LA, Grings M, Nonose Y, Lazzarotto GS, Nogara D, da Silva JS, Fontella FU, Wajner M, Leipnitz G, Souza DO, de Assis AM. Acute Liver Failure Induces Glial Reactivity, Oxidative Stress and Impairs Brain Energy Metabolism in Rats. Front Mol Neurosci 2020; 12:327. [PMID: 31998076 PMCID: PMC6968792 DOI: 10.3389/fnmol.2019.00327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/18/2019] [Indexed: 01/02/2023] Open
Abstract
Acute liver failure (ALF) implies a severe and rapid liver dysfunction that leads to impaired liver metabolism and hepatic encephalopathy (HE). Recent studies have suggested that several brain alterations such as astrocytic dysfunction and energy metabolism impairment may synergistically interact, playing a role in the development of HE. The purpose of the present study is to investigate early alterations in redox status, energy metabolism and astrocytic reactivity of rats submitted to ALF. Adult male Wistar rats were submitted either to subtotal hepatectomy (92% of liver mass) or sham operation to induce ALF. Twenty-four hours after the surgery, animals with ALF presented higher plasmatic levels of ammonia, lactate, ALT and AST and lower levels of glucose than the animals in the sham group. Animals with ALF presented several astrocytic morphological alterations indicating astrocytic reactivity. The ALF group also presented higher mitochondrial oxygen consumption, higher enzymatic activity and higher ATP levels in the brain (frontoparietal cortex). Moreover, ALF induced an increase in glutamate oxidation concomitant with a decrease in glucose and lactate oxidation. The increase in brain energy metabolism caused by astrocytic reactivity resulted in augmented levels of reactive oxygen species (ROS) and Poly [ADP-ribose] polymerase 1 (PARP1) and a decreased activity of the enzymes superoxide dismutase and glutathione peroxidase (GSH-Px). These findings suggest that in the early stages of ALF the brain presents a hypermetabolic state, oxidative stress and astrocytic reactivity, which could be in part sustained by an increase in mitochondrial oxidation of glutamate.
Collapse
Affiliation(s)
- Pedro Arend Guazzelli
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Giordano Fabricio Cittolin-Santos
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Leo Anderson Meira-Martins
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Mateus Grings
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Yasmine Nonose
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Gabriel S Lazzarotto
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Daniela Nogara
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Jussemara S da Silva
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Fernanda U Fontella
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Moacir Wajner
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Guilhian Leipnitz
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Diogo O Souza
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Adriano Martimbianco de Assis
- Post-graduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil.,Post-graduate Program in Health and Behavior, Health Sciences Centre, Universidade Católica de Pelotas-UCPel, Pelotas, Brazil
| |
Collapse
|
5
|
Delgado CA, Balbueno Guerreiro GB, Diaz Jacques CE, de Moura Coelho D, Sitta A, Manfredini V, Wajner M, Vargas CR. Prevention by L-carnitine of DNA damage induced by 3-hydroxy-3-methylglutaric and 3-methylglutaric acids and experimental evidence of lipid and DNA damage in patients with 3-hydroxy-3-methylglutaric aciduria. Arch Biochem Biophys 2019; 668:16-22. [PMID: 31047871 DOI: 10.1016/j.abb.2019.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/25/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022]
Abstract
3-hydroxy-3-methylglutaric aciduria (HMGA) is an inherited disorder of the leucine catabolic pathway in which occurs a deficiency of the 3-hydroxy-3-methylglutaryl-CoA lyase enzyme. Therefore, the organic acids 3-hydroxy-3-methylglutaric (HMG) and 3-methylglutaric (MGA), mainly, accumulate in tissues of affected patients. Lately, much attention has been focused on free radicals as mediators of tissue damage in human diseases, causing lipid peroxidation, protein oxidation and DNA damage. The treatment of this disease is based in a restricted protein ingest and supplementation with l-carnitine (LC), an antioxidant and detoxifying agent. In the present work, we investigated the in vitro oxidative damage to DNA induced by the accumulation of organic acids and oxidative stress parameters in vivo of patients with 3-HMG, as well as the effect of the recommended therapy. The in vitro DNA damage was analyzed by the alkaline comet assay in leukocytes incubated with HMG and MGA (1 mM, 2.5 mM and 5 mM) and co-incubated with LC (90 μM and 150 μM). The in vivo urinary 15-F2t-isoprostane levels and urinary oxidized guanine species were measured by ELISA kits in patient's urine before and after the treatment with LC. HMG and MGA induced a DNA damage index (DI) significantly higher than that of the control group. The DI was significantly reduced in the presence of LC. It was also verified a significant increase of oxidized guanine species and urinary isoprostane levels, biomarker of oxidative DNA damage and lipid peroxidation respectively, in patients before treatment. After the treatment and supplementation with LC, patients presented significantly lower levels of those biomarkers. Analyzing the data together, we can conclude that HMGA patients present oxidative lipid and DNA damage, which is induced by HMG and MGA, and the antioxidant therapy with LC can prevent that kind of injuries.
Collapse
Affiliation(s)
- Camila Aguilar Delgado
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, UFRGS, Av. Ipiranga, 27522, CEP, 90610-000, Porto Alegre, RS, Brazil; Serviço de Genética Médica, HCPA, R.Ramiro Barcelos, 2350, CEP, 90035-003, Porto Alegre, RS, Brazil.
| | - Gilian Batista Balbueno Guerreiro
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, UFRGS, Av. Ipiranga, 27522, CEP, 90610-000, Porto Alegre, RS, Brazil; Serviço de Genética Médica, HCPA, R.Ramiro Barcelos, 2350, CEP, 90035-003, Porto Alegre, RS, Brazil
| | - Carlos Eduardo Diaz Jacques
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, R.Ramiro Barcelos, 2600, CEP, 90035-03, Porto Alegre, RS, Brazil; Serviço de Genética Médica, HCPA, R.Ramiro Barcelos, 2350, CEP, 90035-003, Porto Alegre, RS, Brazil
| | - Daniella de Moura Coelho
- Serviço de Genética Médica, HCPA, R.Ramiro Barcelos, 2350, CEP, 90035-003, Porto Alegre, RS, Brazil
| | - Angela Sitta
- Serviço de Genética Médica, HCPA, R.Ramiro Barcelos, 2350, CEP, 90035-003, Porto Alegre, RS, Brazil
| | - Vanusa Manfredini
- Programa de Pós-Graduação em Bioquímica, BR 427 Km 585, 118, Universidade Federal do Pampa, CEP, 97500-970, Uruguaiana, RS, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, R.Ramiro Barcelos, 2600, CEP, 90035-03, Porto Alegre, RS, Brazil; Serviço de Genética Médica, HCPA, R.Ramiro Barcelos, 2350, CEP, 90035-003, Porto Alegre, RS, Brazil
| | - Carmen Regla Vargas
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, UFRGS, Av. Ipiranga, 27522, CEP, 90610-000, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, R.Ramiro Barcelos, 2600, CEP, 90035-03, Porto Alegre, RS, Brazil; Serviço de Genética Médica, HCPA, R.Ramiro Barcelos, 2350, CEP, 90035-003, Porto Alegre, RS, Brazil.
| |
Collapse
|
6
|
Guerreiro G, Amaral AU, Ribeiro RT, Faverzani J, Groehs AC, Sitta A, Deon M, Wajner M, Vargas CR. l-Carnitine prevents oxidative stress in striatum of glutaryl-CoA dehydrogenase deficient mice submitted to lysine overload. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2420-2427. [PMID: 31181292 DOI: 10.1016/j.bbadis.2019.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/24/2019] [Accepted: 06/03/2019] [Indexed: 11/25/2022]
Abstract
The deficiency of the enzyme glutaryl-CoA dehydrogenase leads to predominant accumulation of glutaric acid (GA) in the organism and is known as glutaric acidemia type I (GA1). Despite the mechanisms of brain damage involved in GA1 are not fully understood, oxidative stress may be involved in this process. Treatment is based on protein/lysine (Lys) restriction and l-carnitine (L-car) supplementation. L-car was recently shown to have an important antioxidant role. A knockout mice model (Gcdh-/-) submitted to a dietary overload of Lys was developed to better understand the GA1 pathogenesis. In this study, we evaluated L-car and glutarylcarnitine levels, the lipid and protein damage, reactive oxygen species (ROS) production and antioxidant enzymes activities in striatum of Gcdh-/- and wild-type (WT) mice. We also determined the effect of the L-car treatment on these parameters. Thirty-day-old Gcdh-/- and WT mice were fed a normal chow (0.9% Lys) or submitted to a high Lys diet (4.7%) for 72 h. Additionally, these animals were administered with three intraperitoneal injections of saline or L-car in different times. Gcdh-/- mice were deficient in L-car and presented a higher glutarylcarnitine levels. They also presented lipid and protein damage, an increased ROS production and altered antioxidant enzymes compared to WT mice. Additionally, mice exposed to Lys overload presented higher alterations in these parameters than mice under normal diet, which were significantly decreased or normalized in those receiving L-car. Thus, we demonstrated a new beneficial effect of the L-car treatment attenuating or abolishing the oxidative stress process in Gcdh-/- mice.
Collapse
Affiliation(s)
- Gilian Guerreiro
- Faculdade de Farmácia, UFRGS, Av. Ipiranga 2752, 90610-000 Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, UFRGS, Av. Ipiranga, 2752, 90610-000 Porto Alegre, RS, Brazil.
| | - Alexandre U Amaral
- Programa de Pós-Graduação em CB:Bioquímica, UFRGS, Rua Ramiro Barcelos, 2600, 90035 000 Porto Alegre, RS, Brazil
| | - Rafael Teixeira Ribeiro
- Programa de Pós-Graduação em CB:Bioquímica, UFRGS, Rua Ramiro Barcelos, 2600, 90035 000 Porto Alegre, RS, Brazil
| | - Jéssica Faverzani
- Faculdade de Farmácia, UFRGS, Av. Ipiranga 2752, 90610-000 Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, UFRGS, Av. Ipiranga, 2752, 90610-000 Porto Alegre, RS, Brazil
| | - Ana Carolina Groehs
- Faculdade de Farmácia, UFRGS, Av. Ipiranga 2752, 90610-000 Porto Alegre, RS, Brazil
| | - Angela Sitta
- Serviço de Genética Médica, HCPA, UFRGS, Rua Ramiro Barcelos, 2350, 90035-903 Porto Alegre, RS, Brazil
| | - Marion Deon
- Serviço de Genética Médica, HCPA, UFRGS, Rua Ramiro Barcelos, 2350, 90035-903 Porto Alegre, RS, Brazil
| | - Moacir Wajner
- Serviço de Genética Médica, HCPA, UFRGS, Rua Ramiro Barcelos, 2350, 90035-903 Porto Alegre, RS, Brazil; Programa de Pós-Graduação em CB:Bioquímica, UFRGS, Rua Ramiro Barcelos, 2600, 90035 000 Porto Alegre, RS, Brazil
| | - Carmen Regla Vargas
- Faculdade de Farmácia, UFRGS, Av. Ipiranga 2752, 90610-000 Porto Alegre, RS, Brazil; Serviço de Genética Médica, HCPA, UFRGS, Rua Ramiro Barcelos, 2350, 90035-903 Porto Alegre, RS, Brazil; Programa de Pós-Graduação em CB:Bioquímica, UFRGS, Rua Ramiro Barcelos, 2600, 90035 000 Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, UFRGS, Av. Ipiranga, 2752, 90610-000 Porto Alegre, RS, Brazil.
| |
Collapse
|
7
|
Pathogenesis of brain damage in glutaric acidemia type I: Lessons from the genetic mice model. Int J Dev Neurosci 2019; 78:215-221. [DOI: 10.1016/j.ijdevneu.2019.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/18/2019] [Accepted: 05/18/2019] [Indexed: 11/22/2022] Open
|
8
|
|
9
|
Guerreiro G, Faverzani J, Jacques CED, Marchetti DP, Sitta A, de Moura Coelho D, Kayser A, Kok F, Athayde L, Manfredini V, Wajner M, Vargas CR. Oxidative damage in glutaric aciduria type I patients and the protective effects of l-carnitine treatment. J Cell Biochem 2018; 119:10021-10032. [PMID: 30129250 DOI: 10.1002/jcb.27332] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/28/2018] [Indexed: 12/13/2022]
Abstract
The deficiency of the enzyme glutaryl-CoA dehydrogenase, known as glutaric acidemia type I (GA-I), leads to the accumulation of glutaric acid (GA) and glutarilcarnitine (C5DC) in the tissues and body fluids, unleashing important neurotoxic effects. l-carnitine (l-car) is recommended for the treatment of GA-I, aiming to induce the excretion of toxic metabolites. l-car has also demonstrated an important role as antioxidant and anti-inflammatory in some neurometabolic diseases. This study evaluated GA-I patients at diagnosis moment and treated the oxidative damage to lipids, proteins, and the inflammatory profile, as well as in vivo and in vitro DNA damage, reactive nitrogen species (RNS), and antioxidant capacity, verifying if the actual treatment with l-car (100 mg kg-1 day-1 ) is able to protect the organism against these processes. Significant increases of GA and C5DC were observed in GA-I patients. A deficiency of carnitine in patients before the supplementation was found. GA-I patients presented significantly increased levels of isoprostanes, di-tyrosine, urinary oxidized guanine species, and the RNS, as well as a reduced antioxidant capacity. The l-car supplementation induced beneficial effects reducing these biomarkers levels and increasing the antioxidant capacity. GA, in three different concentrations, significantly induced DNA damage in vitro, and the l-car was able to prevent this damage. Significant increases of pro-inflammatory cytokines IL-6, IL-8, GM-CSF, and TNF-α were shown in patients. Thus, the beneficial effects of l-car presented in the treatment of GA-I are due not only by increasing the excretion of accumulated toxic metabolites, but also by preventing oxidative damage.
Collapse
Affiliation(s)
- Gilian Guerreiro
- Faculdade de Farmácia, UFRGS, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Farmacêuticas, UFRGS, Porto Alegre, RS, Brazil
| | - Jéssica Faverzani
- Faculdade de Farmácia, UFRGS, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Farmacêuticas, UFRGS, Porto Alegre, RS, Brazil
| | | | | | - Angela Sitta
- Serviço de Genética Médica, HCPA, UFRGS, Porto Alegre, RS, Brazil
| | | | - Aline Kayser
- Faculdade de Farmácia, UFRGS, Porto Alegre, RS, Brazil
| | - Fernando Kok
- Departamento de Neurologia, Unidade de Neurogenética, Escola de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Larissa Athayde
- Departamento de Neurologia, Unidade de Neurogenética, Escola de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Vanusa Manfredini
- Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, CEP, Uruguaiana, RS, Brazil
| | - Moacir Wajner
- Serviço de Genética Médica, HCPA, UFRGS, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em CB:Bioquímica, UFRGS, Porto Alegre, RS, Brazil
| | - Carmen Regla Vargas
- Faculdade de Farmácia, UFRGS, Porto Alegre, RS, Brazil.,Serviço de Genética Médica, HCPA, UFRGS, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em CB:Bioquímica, UFRGS, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Farmacêuticas, UFRGS, Porto Alegre, RS, Brazil
| |
Collapse
|
10
|
Dimer NW, Ferreira BK, Agostini JF, Gomes ML, Kist LW, Malgarin F, Carvalho-Silva M, Gomes LM, Rebelo J, Frederico MJS, Silva FRMB, Rico EP, Bogo MR, Streck EL, Ferreira GC, Schuck PF. Brain bioenergetics in rats with acute hyperphenylalaninemia. Neurochem Int 2018; 117:188-203. [PMID: 29454001 DOI: 10.1016/j.neuint.2018.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 12/21/2017] [Accepted: 01/03/2018] [Indexed: 12/15/2022]
Abstract
Phenylketonuria (PKU) is a disorder of phenylalanine (Phe) metabolism caused by deficient phenylalanine hydroxylase (PAH) activity. The deficiency results in increased levels of Phe and its metabolites in fluids and tissues of patients. PKU patients present neurological signs and symptoms including hypomyelination and intellectual deficit. This study assessed brain bioenergetics at 1 h after acute Phe administration to induce hyperphenylalaninemia (HPA) in rats. Wistar rats were randomized in two groups: HPA animals received a single subcutaneous administration of Phe (5.2 μmol/g) plus p-Cl-Phe (PAH inhibitor) (0.9 μmol/g); control animals received a single injection of 0.9% NaCl. In cerebral cortex, HPA group showed lower mitochondrial mass, lower glycogen levels, as well as lower activities of complexes I-III and IV, ATP synthase and citrate synthase. Higher levels of free Pi and phospho-AMPK, and higher activities of LDH, α-ketoglutarate dehydrogenase and isocitrate dehydrogenase were also reported in cerebral cortex of HPA animals. In striatum, HPA animals had higher LDH (pyruvate to lactate) and isocitrate dehydrogenase activities, and lower activities of α-ketoglutarate dehydrogenase and complex IV, as well as lower phospho-AMPK immunocontent. In hippocampus, HPA rats had higher mRNA expression for MFN1 and higher activities of α-ketoglutarate dehydrogenase and isocitrate dehydrogenase, but decreased activities of pyruvate dehydrogenase and complexes I and IV. In conclusion, our data demonstrated impaired bioenergetics in cerebral cortex, striatum and hippocampus of HPA rats.
Collapse
Affiliation(s)
- Nádia Weber Dimer
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Bruna Klippel Ferreira
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil; Laboratório de Neuroenergética e Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jotele Fontana Agostini
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Maria Luiza Gomes
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Luiza Wilges Kist
- Laboratório de Biologia Genômica e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Malgarin
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Milena Carvalho-Silva
- Laboratório de Bioenergética, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Lara Mezari Gomes
- Laboratório de Bioenergética, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Joyce Rebelo
- Laboratório de Bioenergética, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Marisa Jádna Silva Frederico
- Laboratório de Hormônios e Transdução de Sinais, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Fátima Regina Mena Barreto Silva
- Laboratório de Hormônios e Transdução de Sinais, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Eduardo Pacheco Rico
- Laboratório de Sinalização Neural e Psicofarmacologia, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Mauricio Reis Bogo
- Laboratório de Biologia Genômica e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Emilio Luiz Streck
- Laboratório de Bioenergética, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gustavo Costa Ferreira
- Laboratório de Neuroenergética e Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patrícia Fernanda Schuck
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.
| |
Collapse
|
11
|
Pierozan P, Colín-González AL, Biasibetti H, da Silva JC, Wyse A, Wajner M, Santamaria A. Toxic Synergism Between Quinolinic Acid and Glutaric Acid in Neuronal Cells Is Mediated by Oxidative Stress: Insights to a New Toxic Model. Mol Neurobiol 2017; 55:5362-5376. [PMID: 28936789 DOI: 10.1007/s12035-017-0761-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/31/2017] [Indexed: 12/31/2022]
Abstract
It has been shown that synergistic toxic effects of quinolinic acid (QUIN) and glutaric acid (GA), both in isolated nerve endings and in vivo conditions, suggest the contribution of these metabolites to neurodegeneration. However, this synergism still requires a detailed characterization of the mechanisms involved in cell damage during its occurrence. In this study, the effects of subtoxic concentrations of QUIN and/or GA were tested in neuronal cultures, co-cultures (neuronal cells + astrocytes), and mixed cultures (neuronal cells + astrocytes + microglia) from rat cortex and striatum. The exposure of different cortical and striatal cell cultures to QUIN + GA resulted in cell death and stimulated different markers of oxidative stress, including reactive oxygen species (ROS) formation; changes in the activity of antioxidant enzymes such as superoxide dismutase, catalase, and glutathione peroxidase; and depletion of endogenous antioxidants such as -SH groups and glutathione. The co-incubation of neuronal cultures with QUIN + GA plus the N-methyl-D-aspartate antagonist MK-801 prevented cell death but not ROS formation, whereas the antioxidant melatonin reduced both parameters. Our results demonstrated that QUIN and GA can create synergistic scenarios, inducing toxic effects on some parameters of cell viability via the stimulation of oxidative damage. Therefore, it is likely that oxidative stress may play a major causative role in the synergistic actions exerted by QUIN + GA in a variety of cell culture conditions involving the interaction of different neural types.
Collapse
Affiliation(s)
- Paula Pierozan
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ana Laura Colín-González
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, SSA, Insurgentes Sur 3877, 14269, Mexico City, Mexico
| | - Helena Biasibetti
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Janaina Camacho da Silva
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Angela Wyse
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Moacir Wajner
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, SSA, Insurgentes Sur 3877, 14269, Mexico City, Mexico.
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, SSA, Insurgentes Sur 3877, 14269, Mexico City, Mexico.
| |
Collapse
|
12
|
L-Carnitine and Acetyl-L-carnitine Roles and Neuroprotection in Developing Brain. Neurochem Res 2017; 42:1661-1675. [PMID: 28508995 DOI: 10.1007/s11064-017-2288-7] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 12/30/2022]
Abstract
L-Carnitine functions to transport long chain fatty acyl-CoAs into the mitochondria for degradation by β-oxidation. Treatment with L-carnitine can ameliorate metabolic imbalances in many inborn errors of metabolism. In recent years there has been considerable interest in the therapeutic potential of L-carnitine and its acetylated derivative acetyl-L-carnitine (ALCAR) for neuroprotection in a number of disorders including hypoxia-ischemia, traumatic brain injury, Alzheimer's disease and in conditions leading to central or peripheral nervous system injury. There is compelling evidence from preclinical studies that L-carnitine and ALCAR can improve energy status, decrease oxidative stress and prevent subsequent cell death in models of adult, neonatal and pediatric brain injury. ALCAR can provide an acetyl moiety that can be oxidized for energy, used as a precursor for acetylcholine, or incorporated into glutamate, glutamine and GABA, or into lipids for myelination and cell growth. Administration of ALCAR after brain injury in rat pups improved long-term functional outcomes, including memory. Additional studies are needed to better explore the potential of L-carnitine and ALCAR for protection of developing brain as there is an urgent need for therapies that can improve outcome after neonatal and pediatric brain injury.
Collapse
|
13
|
Nonose Y, Gewehr PE, Almeida RF, da Silva JS, Bellaver B, Martins LAM, Zimmer ER, Greggio S, Venturin GT, Da Costa JC, Quincozes-Santos A, Pellerin L, de Souza DO, de Assis AM. Cortical Bilateral Adaptations in Rats Submitted to Focal Cerebral Ischemia: Emphasis on Glial Metabolism. Mol Neurobiol 2017; 55:2025-2041. [PMID: 28271402 DOI: 10.1007/s12035-017-0458-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 02/13/2017] [Indexed: 11/30/2022]
Abstract
This study was performed to evaluate the bilateral effects of focal permanent ischemia (FPI) on glial metabolism in the cerebral cortex. Two and 9 days after FPI induction, we analyze [18F]FDG metabolism by micro-PET, astrocyte morphology and reactivity by immunohistochemistry, cytokines and trophic factors by ELISA, glutamate transporters by RT-PCR, monocarboxylate transporters (MCTs) by western blot, and substrate uptake and oxidation by ex vivo slices model. The FPI was induced surgically by thermocoagulation of the blood in the pial vessels of the motor and sensorimotor cortices in adult (90 days old) male Wistar rats. Neurochemical analyses were performed separately on both ipsilateral and contralateral cortical hemispheres. In both cortical hemispheres, we observed an increase in tumor necrosis factor alpha (TNF-α), interleukin-1β (IL-1β), and glutamate transporter 1 (GLT-1) mRNA levels; lactate oxidation; and glutamate uptake and a decrease in brain-derived neurotrophic factor (BDNF) after 2 days of FPI. Nine days after FPI, we observed an increase in TNF-α levels and a decrease in BDNF, GLT-1, and glutamate aspartate transporter (GLAST) mRNA levels in both hemispheres. Additionally, most of the unilateral alterations were found only in the ipsilateral hemisphere and persisted until 9 days post-FPI. They include diminished in vivo glucose uptake and GLAST expression, followed by increased glial fibrillary acidic protein (GFAP) gray values, astrocyte reactivity, and glutamate oxidation. Astrocytes presented signs of long-lasting reactivity, showing a radial morphology. In the intact hemisphere, there was a decrease in MCT2 levels, which did not persist. Our study shows the bilateralism of glial modifications following FPI, highlighting the role of energy metabolism adaptations on brain recovery post-ischemia.
Collapse
Affiliation(s)
- Yasmine Nonose
- Postgraduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 - Anexo Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Pedro E Gewehr
- Postgraduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 - Anexo Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Roberto F Almeida
- Postgraduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 - Anexo Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Jussemara S da Silva
- Postgraduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 - Anexo Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Bruna Bellaver
- Postgraduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 - Anexo Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Leo A M Martins
- Postgraduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 - Anexo Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Eduardo R Zimmer
- Postgraduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 - Anexo Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.,Brain Institute of Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, 90619-900, Brazil
| | - Samuel Greggio
- Brain Institute of Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, 90619-900, Brazil
| | - Gianina T Venturin
- Brain Institute of Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, 90619-900, Brazil
| | - Jaderson C Da Costa
- Brain Institute of Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, 90619-900, Brazil
| | - André Quincozes-Santos
- Postgraduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 - Anexo Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Luc Pellerin
- Department of Physiology, University of Lausanne, 1005, Lausanne, Switzerland
| | - Diogo O de Souza
- Postgraduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 - Anexo Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Adriano M de Assis
- Postgraduate Program in Biological Sciences: Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600 - Anexo Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
14
|
de Assis AM, da Silva JS, Rech A, Longoni A, Nonose Y, Repond C, de Bittencourt Pasquali MA, Moreira JCF, Souza DO, Pellerin L. Cerebral Ketone Body Oxidation Is Facilitated by a High Fat Diet Enriched with Advanced Glycation End Products in Normal and Diabetic Rats. Front Neurosci 2016; 10:509. [PMID: 27877108 PMCID: PMC5099525 DOI: 10.3389/fnins.2016.00509] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/24/2016] [Indexed: 12/30/2022] Open
Abstract
Diabetes mellitus (DM) causes important modifications in the availability and use of different energy substrates in various organs and tissues. Similarly, dietary manipulations such as high fat diets also affect systemic energy metabolism. However, how the brain adapts to these situations remains unclear. To investigate these issues, control and alloxan-induced type I diabetic rats were fed either a standard or a high fat diet enriched with advanced glycation end products (AGEs) (HAGE diet). The HAGE diet increased their levels of blood ketone bodies, and this effect was exacerbated by DM induction. To determine the effects of diet and/or DM induction on key cerebral bioenergetic parameters, both ketone bodies (β-hydroxybutyric acid) and lactate oxidation were measured. In parallel, the expression of Monocarboxylate Transporter 1 (MCT1) and 2 (MCT2) isoforms in hippocampal and cortical slices from rats submitted to these diets was assessed. Ketone body oxidation increased while lactate oxidation decreased in hippocampal and cortical slices in both control and diabetic rats fed a HAGE diet. In parallel, the expression of both MCT1 and MCT2 increased only in the cerebral cortex in diabetic rats fed a HAGE diet. These results suggest a shift in the preferential cerebral energy substrate utilization in favor of ketone bodies in animals fed a HAGE diet, an effect that, in DM animals, is accompanied by the enhanced expression of the related transporters.
Collapse
Affiliation(s)
- Adriano M de Assis
- Postgraduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul Porto Alegre, Brazil
| | - Jussemara S da Silva
- Postgraduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul Porto Alegre, Brazil
| | - Anderson Rech
- Postgraduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul Porto Alegre, Brazil
| | - Aline Longoni
- Postgraduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul Porto Alegre, Brazil
| | - Yasmine Nonose
- Postgraduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul Porto Alegre, Brazil
| | - Cendrine Repond
- Department of Physiology, University of Lausanne Lausanne, Switzerland
| | - Matheus A de Bittencourt Pasquali
- Postgraduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do SulPorto Alegre, Brazil; Department of Biochemistry, Institute of Tropical Medicine, Federal University of Rio Grande do NorteNatal, Brazil
| | - José C F Moreira
- Postgraduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do SulPorto Alegre, Brazil; Department of Biochemistry, Federal University of Rio Grande do SulPorto Alegre, Brazil
| | - Diogo O Souza
- Postgraduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do SulPorto Alegre, Brazil; Department of Biochemistry, Federal University of Rio Grande do SulPorto Alegre, Brazil
| | - Luc Pellerin
- Department of Physiology, University of Lausanne Lausanne, Switzerland
| |
Collapse
|
15
|
Rodrigues MDN, Seminotti B, Zanatta Â, de Mello Gonçalves A, Bellaver B, Amaral AU, Quincozes-Santos A, Goodman SI, Woontner M, Souza DO, Wajner M. Higher Vulnerability of Menadione-Exposed Cortical Astrocytes of Glutaryl-CoA Dehydrogenase Deficient Mice to Oxidative Stress, Mitochondrial Dysfunction, and Cell Death: Implications for the Neurodegeneration in Glutaric Aciduria Type I. Mol Neurobiol 2016; 54:4795-4805. [PMID: 27510504 DOI: 10.1007/s12035-016-0023-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 08/01/2016] [Indexed: 11/27/2022]
Abstract
Patients affected by glutaric aciduria type I (GA-I) show progressive cortical leukoencephalopathy whose pathogenesis is poorly known. In the present work, we exposed cortical astrocytes of wild-type (Gcdh +/+ ) and glutaryl-CoA dehydrogenase knockout (Gcdh -/- ) mice to the oxidative stress inducer menadione and measured mitochondrial bioenergetics, redox homeostasis, and cell viability. Mitochondrial function (MTT and JC1-mitochondrial membrane potential assays), redox homeostasis (DCFH oxidation, nitrate and nitrite production, GSH concentrations and activities of the antioxidant enzymes SOD and GPx), and cell death (propidium iodide incorporation) were evaluated in primary cortical astrocyte cultures of Gcdh +/+ and Gcdh -/- mice unstimulated and stimulated by menadione. We also measured the pro-inflammatory response (TNFα levels, IL1-β and NF-ƙB) in unstimulated astrocytes obtained from these mice. Gcdh -/- mice astrocytes were more vulnerable to menadione-induced oxidative stress (decreased GSH concentrations and altered activities of the antioxidant enzymes), mitochondrial dysfunction (decrease of MTT reduction and JC1 values), and cell death as compared with Gcdh +/+ astrocytes. A higher inflammatory response (TNFα, IL1-β and NF-ƙB) was also observed in Gcdh -/- mice astrocytes. These data indicate a higher susceptibility of Gcdh -/- cortical astrocytes to oxidative stress and mitochondrial dysfunction, probably leading to cell death. It is presumed that these pathomechanisms may contribute to the cortical leukodystrophy observed in GA-I patients.
Collapse
Affiliation(s)
- Marília Danyelle Nunes Rodrigues
- Departamento e PPG Bioquímica, ICBS/Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos N° 2600, Anexo, Porto Alegre, RS, CEP90035-003, Brazil
| | - Bianca Seminotti
- Departamento e PPG Bioquímica, ICBS/Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos N° 2600, Anexo, Porto Alegre, RS, CEP90035-003, Brazil
| | - Ângela Zanatta
- Departamento e PPG Bioquímica, ICBS/Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos N° 2600, Anexo, Porto Alegre, RS, CEP90035-003, Brazil
| | - Aline de Mello Gonçalves
- Departamento e PPG Bioquímica, ICBS/Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos N° 2600, Anexo, Porto Alegre, RS, CEP90035-003, Brazil
| | - Bruna Bellaver
- Departamento e PPG Bioquímica, ICBS/Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos N° 2600, Anexo, Porto Alegre, RS, CEP90035-003, Brazil
| | - Alexandre Umpierrez Amaral
- Departamento e PPG Bioquímica, ICBS/Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos N° 2600, Anexo, Porto Alegre, RS, CEP90035-003, Brazil
| | - André Quincozes-Santos
- Departamento e PPG Bioquímica, ICBS/Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos N° 2600, Anexo, Porto Alegre, RS, CEP90035-003, Brazil
| | | | - Michael Woontner
- Department of Pediatrics, University of Colorado Denver, Aurora, CO, USA
| | - Diogo Onofre Souza
- Departamento e PPG Bioquímica, ICBS/Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos N° 2600, Anexo, Porto Alegre, RS, CEP90035-003, Brazil
| | - Moacir Wajner
- Departamento e PPG Bioquímica, ICBS/Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos N° 2600, Anexo, Porto Alegre, RS, CEP90035-003, Brazil.
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| |
Collapse
|
16
|
Mishur RJ, Khan M, Munkácsy E, Sharma L, Bokov A, Beam H, Radetskaya O, Borror M, Lane R, Bai Y, Rea SL. Mitochondrial metabolites extend lifespan. Aging Cell 2016; 15:336-48. [PMID: 26729005 PMCID: PMC4783347 DOI: 10.1111/acel.12439] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2015] [Indexed: 12/26/2022] Open
Abstract
Disruption of mitochondrial respiration in the nematode Caenorhabditis elegans can extend lifespan. We previously showed that long-lived respiratory mutants generate elevated amounts of α-ketoacids. These compounds are structurally related to α-ketoglutarate, suggesting they may be biologically relevant. Here, we show that provision of several such metabolites to wild-type worms is sufficient to extend their life. At least one mode of action is through stabilization of hypoxia-inducible factor-1 (HIF-1). We also find that an α-ketoglutarate mimetic, 2,4-pyridinedicarboxylic acid (2,4-PDA), is alone sufficient to increase the lifespan of wild-type worms and this effect is blocked by removal of HIF-1. HIF-1 is constitutively active in isp-1(qm150) Mit mutants, and accordingly, 2,4-PDA does not further increase their lifespan. Incubation of mouse 3T3-L1 fibroblasts with life-prolonging α-ketoacids also results in HIF-1α stabilization. We propose that metabolites that build up following mitochondrial respiratory dysfunction form a novel mode of cell signaling that acts to regulate lifespan.
Collapse
Affiliation(s)
- Robert J. Mishur
- The Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
- Department of PhysiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Maruf Khan
- The Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
- Department of PhysiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Erin Munkácsy
- The Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
- Department of Cellular & Structural BiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Lokendra Sharma
- Department of Cellular & Structural BiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
- Biotechnology ProgrammeCenter for Biological SciencesCentral University of South BiharPatna800014India
| | - Alex Bokov
- Department of Epidemiology and BiostatisticsUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Haley Beam
- The Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Oxana Radetskaya
- The Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Megan Borror
- The Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Rebecca Lane
- The Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Yidong Bai
- Department of Cellular & Structural BiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| | - Shane L. Rea
- The Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
- Department of PhysiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
| |
Collapse
|
17
|
Seminotti B, Amaral AU, Ribeiro RT, Rodrigues MDN, Colín-González AL, Leipnitz G, Santamaría A, Wajner M. Oxidative Stress, Disrupted Energy Metabolism, and Altered Signaling Pathways in Glutaryl-CoA Dehydrogenase Knockout Mice: Potential Implications of Quinolinic Acid Toxicity in the Neuropathology of Glutaric Acidemia Type I. Mol Neurobiol 2015; 53:6459-6475. [PMID: 26607633 DOI: 10.1007/s12035-015-9548-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 11/17/2015] [Indexed: 12/13/2022]
Abstract
We investigated the effects of an acute intrastriatal QUIN administration on cellular redox and bioenergetics homeostasis, as well as on important signaling pathways in the striatum of wild-type (Gcdh +/+ , WT) and knockout mice for glutaryl-CoA dehydrogenase (Gcdh -/- ) fed a high lysine (Lys, 4.7 %) chow. QUIN increased lactate release in both Gcdh +/+ and Gcdh -/- mice and reduced the activities of complex IV and creatine kinase only in the striatum of Gcdh -/- mice. QUIN also induced lipid and protein oxidative damage and increased the generation of reactive nitrogen species, as well as the activities of the antioxidant enzymes glutathione peroxidase, superoxide dismutase 2, and glutathione-S-transferase in WT and Gcdh -/- animals. Furthermore, QUIN induced DCFH oxidation (reactive oxygen species production) and reduced GSH concentrations (antioxidant defenses) in Gcdh -/- . An early increase of Akt and phospho-Erk 1/2 in the cytosol and Nrf2 in the nucleus was also observed, as well as a decrease of cytosolic Keap1caused by QUIN, indicating activation of the Nrf2 pathway mediated by Akt and phospho-Erk 1/2, possibly as a compensatory protective mechanism against the ongoing QUIN-induced toxicity. Finally, QUIN increased NF-κB and diminished IκBα expression, evidencing a pro-inflammatory response. Our data show a disruption of energy and redox homeostasis associated to inflammation induced by QUIN in the striatum of Gcdh -/- mice submitted to a high Lys diet. Therefore, it is presumed that QUIN may possibly contribute to the pathophysiology of striatal degeneration in children with glutaric aciduria type I during inflammatory processes triggered by infections or vaccinations.
Collapse
Affiliation(s)
- Bianca Seminotti
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos N° 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Alexandre Umpierrez Amaral
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos N° 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Rafael Teixeira Ribeiro
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos N° 2600-Anexo, CEP 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 N° 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Ana Laura Colín-González
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez, SSA, México, DF, México
| | - Guilhian Leipnitz
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos N° 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez, SSA, México, DF, México
| | - Moacir Wajner
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos N° 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil. .,Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| |
Collapse
|
18
|
Colín-González A, Paz-Loyola A, Serratos I, Seminotti B, Ribeiro C, Leipnitz G, Souza D, Wajner M, Santamaría A. Toxic synergism between quinolinic acid and organic acids accumulating in glutaric acidemia type I and in disorders of propionate metabolism in rat brain synaptosomes: Relevance for metabolic acidemias. Neuroscience 2015; 308:64-74. [DOI: 10.1016/j.neuroscience.2015.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/29/2015] [Accepted: 09/01/2015] [Indexed: 12/11/2022]
|
19
|
Colín-González AL, Paz-Loyola AL, Serratos IN, Seminotti B, Ribeiro CAJ, Leipnitz G, Souza DO, Wajner M, Santamaría A. The effect of WIN 55,212-2 suggests a cannabinoid-sensitive component in the early toxicity induced by organic acids accumulating in glutaric acidemia type I and in related disorders of propionate metabolism in rat brain synaptosomes. Neuroscience 2015; 310:578-88. [PMID: 26431622 DOI: 10.1016/j.neuroscience.2015.09.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 09/03/2015] [Accepted: 09/18/2015] [Indexed: 10/23/2022]
Abstract
Several physiological processes in the CNS are regulated by the endocannabinoid system (ECS). Cannabinoid receptors (CBr) and CBr agonists have been involved in the modulation of the N-methyl-D-aspartate receptor (NMDAr) activation. Glutaric (GA), 3-hydroxyglutaric (3-OHGA), methylmalonic (MMA) and propionic (PA) acids are endogenous metabolites produced and accumulated in the brain of children affected by severe organic acidemias (OAs) with neurodegeneration. Oxidative stress and excitotoxicity have been involved in the toxic pattern exerted by these organic acids. Studying the early pattern of toxicity exerted by these metabolites is crucial to explain the extent of damage that they can produce in the brain. Herein, we investigated the effects of the synthetic CBr agonist WIN 55,212-2 (WIN) on early markers of GA-, 3-OHGA-, MMA- and PA-induced toxicity in brain synaptosomes from adult (90-day-old) and adolescent (30-day-old) rats. As pre-treatment, WIN exerted protective effects on the GA- and MMA-induced mitochondrial dysfunction, and prevented the reactive oxygen species (ROS) formation and lipid peroxidation induced by all metabolites. Our findings support a protective and modulatory role of cannabinoids in the early toxic events elicited by toxic metabolites involved in OAs.
Collapse
Affiliation(s)
- A L Colín-González
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico
| | - A L Paz-Loyola
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico
| | - I N Serratos
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico; Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, Mexico
| | - B Seminotti
- Departamento de Bioquímica, Instituto de Ciências Básicas da Sáude, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - C A J Ribeiro
- Departamento de Bioquímica, Instituto de Ciências Básicas da Sáude, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - G Leipnitz
- Departamento de Bioquímica, Instituto de Ciências Básicas da Sáude, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - D O Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Sáude, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - M Wajner
- Departamento de Bioquímica, Instituto de Ciências Básicas da Sáude, 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.
| | - A Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico.
| |
Collapse
|
20
|
Amaral AU, Cecatto C, Seminotti B, Ribeiro CA, Lagranha VL, Pereira CC, de Oliveira FH, de Souza DG, Goodman S, Woontner M, Wajner M. Experimental evidence that bioenergetics disruption is not mainly involved in the brain injury of glutaryl-CoA dehydrogenase deficient mice submitted to lysine overload. Brain Res 2015; 1620:116-29. [DOI: 10.1016/j.brainres.2015.05.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 04/29/2015] [Accepted: 05/05/2015] [Indexed: 11/29/2022]
|
21
|
Duarte J, Schuck PF, Wenk GL, Ferreira GC. Metabolic disturbances in diseases with neurological involvement. Aging Dis 2014; 5:238-55. [PMID: 25110608 DOI: 10.14336/ad.2014.0500238] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 11/26/2013] [Accepted: 11/27/2013] [Indexed: 12/19/2022] Open
Abstract
Degeneration of specific neuronal populations and progressive nervous system dysfunction characterize neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. These findings are also reported in inherited diseases such as phenylketonuria and glutaric aciduria type I. The involvement of mitochondrial dysfunction in these diseases was reported, elicited by genetic alterations, exogenous toxins or buildup of toxic metabolites. In this review we shall discuss some metabolic alterations related to the pathophysiology of diseases with neurological involvement and aging process. These findings may help identifying early disease biomarkers and lead to more effective therapies to improve the quality of life of the patients affected by these devastating illnesses.
Collapse
Affiliation(s)
| | - Patrícia F Schuck
- Laboratory of inborn errors of metabolism, Universidade do Extremo Sul Catarinense, Brazil
| | - Gary L Wenk
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Gustavo C Ferreira
- Laboratory of inborn errors of metabolism, Universidade do Extremo Sul Catarinense, Brazil
| |
Collapse
|
22
|
Tian F, Fu X, Gao J, Ying Y, Hou L, Liang Y, Ning Q, Luo X. Glutaric acid-mediated apoptosis in primary striatal neurons. BIOMED RESEARCH INTERNATIONAL 2014; 2014:484731. [PMID: 24900967 PMCID: PMC4036723 DOI: 10.1155/2014/484731] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/20/2014] [Accepted: 04/21/2014] [Indexed: 11/28/2022]
Abstract
Glutaric acid (GA) has been implicated in the mechanism of neurodegeneration in glutaric aciduria type I. In the present study, the potential cytotoxic effects of GA (0.1~50 mM for 24~96 h) were examined in cultured primary rat striatal neurons. Results showed increase in the number of cells labeled by annexin-V or with apoptotic features shown by Hoechst/PI staining and transmission electron microscopy (TEM) and upregulation of the expression of mRNA as well as the active protein fragments caspase 3, suggesting involvement of the caspase 3-dependent apoptotic pathway in GA-induced striatal neuronal death. This effect was in part suppressed by the N-methyl-D-aspartate (NMDA) receptor antagonist MK-801 but not the α -amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) antagonist 6-cyano-7-nitroquinoxalone-2,3-dione (CNQX). Thus, GA may trigger neuronal damage partially through apoptotic pathway and via activation of NMDA receptors in cultured primary striatal neurons.
Collapse
Affiliation(s)
- Fengyan Tian
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xi Fu
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jinzhi Gao
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yanqin Ying
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ling Hou
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yan Liang
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Qin Ning
- Laboratory of Infectious Immunology, Department of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
23
|
Gao J, Zhang C, Fu X, Yi Q, Tian F, Ning Q, Luo X. Effects of targeted suppression of glutaryl-CoA dehydrogenase by lentivirus-mediated shRNA and excessive intake of lysine on apoptosis in rat striatal neurons. PLoS One 2013; 8:e63084. [PMID: 23658800 PMCID: PMC3642093 DOI: 10.1371/journal.pone.0063084] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 03/29/2013] [Indexed: 12/31/2022] Open
Abstract
In glutaric aciduria type 1 (GA1), glutaryl-CoA dehydrogenase (GCDH) deficiency has been shown to be responsible for the accumulation of glutaric acid and striatal degeneration. However, the mechanisms by which GA1 induces striatal degeneration remain unclear. In this study, we aimed to establish a novel neuronal model of GA1 and to investigate the effects of GCDH deficiency and lysine-related metabolites on the viability of rat striatal neurons. Thus we constructed a lentiviral vector containing short hairpin RNA targeted against the GCDH gene expression (lentivirus-shRNA) in neurons. A virus containing a scrambled short hairpin RNA construct served as a control. Addition of lysine (5 mmol/L) was used to mimic hypermetabolism. Cell viability was measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. Apoptosis was assessed using Hoechst33342 staining and Annexin V-PE/7-AAD staining. The mitochondrial membrane potential (MPP) was monitored using tetramethylrhodamine methyl ester. The expression levels of caspases 3, 8, and 9 were determined by Western blotting. We found that lentivirus-shRNA induced apoptosis and decreased MMP levels in neurons, and addition of 5 mmol/L lysine enhanced this effect markedly. Lentivirus-shRNA upregulated the protein levels of caspases 3 and 9 regardless of the presence of 5 mmol/L lysine. The expression level of caspase 8 was higher in neurons co-treated with lentivirus-shRNA and 5 mmol/L lysine than in control. Benzyloxy-carbonyl-Val-Ala-Asp(OMe)-fluoromethylketone, a pan-caspase inhibitor, blocked the apoptosis induced by lentivirus-shRNA and 5 mmol/L lysine to a great extent. These results indicate that the targeted suppression of GCDH by lentivirus-mediated shRNA and excessive intake of lysine may be a useful cell model of GA1. These also suggest that GA1-induced striatal degeneration is partially caspase-dependent.
Collapse
MESH Headings
- Amino Acid Metabolism, Inborn Errors/enzymology
- Amino Acid Metabolism, Inborn Errors/metabolism
- Amino Acid Metabolism, Inborn Errors/pathology
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Base Sequence
- Biological Transport/genetics
- Brain Diseases, Metabolic/enzymology
- Brain Diseases, Metabolic/metabolism
- Brain Diseases, Metabolic/pathology
- Caspase Inhibitors/pharmacology
- Cell Survival/drug effects
- Cell Survival/genetics
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/genetics
- Gene Knockdown Techniques
- Glutaryl-CoA Dehydrogenase/deficiency
- Glutaryl-CoA Dehydrogenase/genetics
- Glutaryl-CoA Dehydrogenase/metabolism
- Lentivirus/genetics
- Lysine/metabolism
- Lysine/pharmacology
- Membrane Potential, Mitochondrial/drug effects
- Membrane Potential, Mitochondrial/genetics
- Neostriatum/cytology
- Neurons/cytology
- Neurons/drug effects
- Neurons/metabolism
- RNA, Small Interfering/genetics
- Rats
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Jinzhi Gao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cai Zhang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Fu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Yi
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengyan Tian
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail:
| |
Collapse
|
24
|
Neurotoxic effects of trans-glutaconic acid in rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:607610. [PMID: 23606926 PMCID: PMC3625603 DOI: 10.1155/2013/607610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/03/2013] [Accepted: 03/04/2013] [Indexed: 02/01/2023]
Abstract
trans-Glutaconic acid (tGA) is an unsaturated C5-dicarboxylic acid which may be found accumulated in glutaric aciduria type I, whose pathophysiology is still uncertain. In the present work it was investigated the in vitro effect of increasing tGA concentrations on neurochemical and oxidative stress parameters in rat cerebral cortex. We observed that Na+, K+-ATPase activity was reduced by tGA, but not creatine kinase, respiratory chain complex IV, and ATP synthase activities. On the other hand, tGA significantly increased lipid peroxidation (thiobarbituric acid-reactive species levels and spontaneous chemiluminescence), as well as protein oxidative damage (oxidation of sulfhydryl groups). tGA also significantly decreased nonenzymatic antioxidant defenses (TRAP and reduced glutathione levels). Our data suggest that tGA may be neurotoxic in rat brain.
Collapse
|
25
|
Seminotti B, Amaral AU, da Rosa MS, Fernandes CG, Leipnitz G, Olivera-Bravo S, Barbeito L, Ribeiro CAJ, de Souza DOG, Woontner M, Goodman SI, Koeller DM, Wajner M. Disruption of brain redox homeostasis in glutaryl-CoA dehydrogenase deficient mice treated with high dietary lysine supplementation. Mol Genet Metab 2013; 108:30-9. [PMID: 23218171 DOI: 10.1016/j.ymgme.2012.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 11/01/2012] [Accepted: 11/01/2012] [Indexed: 10/27/2022]
Abstract
Deficiency of glutaryl-CoA dehydrogenase (GCDH) activity or glutaric aciduria type I (GA I) is an inherited neurometabolic disorder biochemically characterized by predominant accumulation of glutaric acid and 3-hydroxyglutaric acid in the brain and other tissues. Affected patients usually present acute striatum necrosis during encephalopathic crises triggered by metabolic stress situations, as well as chronic leukodystrophy and delayed myelination. Considering that the mechanisms underlying the brain injury in this disease are not yet fully established, in the present study we investigated important parameters of oxidative stress in the brain (cerebral cortex, striatum and hippocampus), liver and heart of 30-day-old GCDH deficient knockout (Gcdh(-/-)) and wild type (WT) mice submitted to a normal lysine (Lys) (0.9% Lys), or high Lys diets (2.8% or 4.7% Lys) for 60 h. It was observed that the dietary supplementation of 2.8% and 4.7% Lys elicited noticeable oxidative stress, as verified by an increase of malondialdehyde concentrations (lipid oxidative damage) and 2-7-dihydrodichlorofluorescein (DCFH) oxidation (free radical production), as well as a decrease of reduced glutathione levels and alteration of various antioxidant enzyme activities (antioxidant defenses) in the cerebral cortex and the striatum, but not in the hippocampus, the liver and the heart of Gcdh(-/-) mice, as compared to WT mice receiving the same diets. Furthermore, alterations of oxidative stress parameters in the cerebral cortex and striatum were more accentuated in symptomatic, as compared to asymptomatic Gcdh(-/-) mice exposed to 4.7% Lys overload. Histopathological studies performed in the cerebral cortex and striatum of these animals exposed to high dietary Lys revealed increased expression of oxidative stress markers despite the absence of significant structural damage. The results indicate that a disruption of redox homeostasis in the cerebral cortex and striatum of young Gcdh(-/-) mice exposed to increased Lys diet may possibly represent an important pathomechanism of brain injury in GA I patients under metabolic stress.
Collapse
Affiliation(s)
- Bianca Seminotti
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Amaral AU, Seminotti B, Cecatto C, Fernandes CG, Busanello ENB, Zanatta Â, Kist LW, Bogo MR, de Souza DOG, Woontner M, Goodman S, Koeller DM, Wajner M. Reduction of Na+, K+-ATPase activity and expression in cerebral cortex of glutaryl-CoA dehydrogenase deficient mice: a possible mechanism for brain injury in glutaric aciduria type I. Mol Genet Metab 2012; 107:375-82. [PMID: 22999741 DOI: 10.1016/j.ymgme.2012.08.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 08/22/2012] [Accepted: 08/22/2012] [Indexed: 10/27/2022]
Abstract
Mitochondrial dysfunction has been proposed to play an important role in the neuropathology of glutaric acidemia type I (GA I). However, the relevance of bioenergetics disruption and the exact mechanisms responsible for the cortical leukodystrophy and the striatum degeneration presented by GA I patients are not yet fully understood. Therefore, in the present work we measured the respiratory chain complexes activities I-IV, mitochondrial respiratory parameters state 3, state 4, the respiratory control ratio and dinitrophenol (DNP)-stimulated respiration (uncoupled state), as well as the activities of α-ketoglutarate dehydrogenase (α-KGDH), creatine kinase (CK) and Na+, K+-ATPase in cerebral cortex, striatum and hippocampus from 30-day-old Gcdh-/- and wild type (WT) mice fed with a normal or a high Lys (4.7%) diet. When a baseline (0.9% Lys) diet was given, we verified mild alterations of the activities of some respiratory chain complexes in cerebral cortex and hippocampus, but not in striatum from Gcdh-/- mice as compared to WT animals. Furthermore, the mitochondrial respiratory parameters and the activities of α-KGDH and CK were not modified in all brain structures from Gcdh-/- mice. In contrast, we found a significant reduction of Na(+), K(+)-ATPase activity associated with a lower degree of its expression in cerebral cortex from Gcdh-/- mice. Furthermore, a high Lys (4.7%) diet did not accentuate the biochemical alterations observed in Gcdh-/- mice fed with a normal diet. Since Na(+), K(+)-ATPase activity is required for cell volume regulation and to maintain the membrane potential necessary for a normal neurotransmission, it is presumed that reduction of this enzyme activity may represent a potential underlying mechanism involved in the brain swelling and cortical abnormalities (cortical atrophy with leukodystrophy) observed in patients affected by GA I.
Collapse
Affiliation(s)
- Alexandre Umpierrez Amaral
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Amaral AU, Cecatto C, Seminotti B, Zanatta Â, Fernandes CG, Busanello ENB, Braga LM, Ribeiro CAJ, de Souza DOG, Woontner M, Koeller DM, Goodman S, Wajner M. Marked reduction of Na(+), K(+)-ATPase and creatine kinase activities induced by acute lysine administration in glutaryl-CoA dehydrogenase deficient mice. Mol Genet Metab 2012; 107:81-6. [PMID: 22578804 DOI: 10.1016/j.ymgme.2012.04.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 04/17/2012] [Accepted: 04/17/2012] [Indexed: 12/23/2022]
Abstract
Glutaric acidemia type I (GA I) is an inherited neurometabolic disorder caused by a severe deficiency of the mitochondrial glutaryl-CoA dehydrogenase activity leading to accumulation of predominantly glutaric (GA) and 3-hydroxyglutaric (3HGA) acids in the brain and other tissues. Affected patients usually present with hypotonia and brain damage and acute encephalopathic episodes whose pathophysiology is not yet fully established. In this study we investigated important parameters of cellular bioenergetics in brain, heart and skeletal muscle from 15-day-old glutaryl-CoA dehydrogenase deficient mice (Gcdh(-/-)) submitted to a single intra-peritoneal injection of saline (Sal) or lysine (Lys - 8 μmol/g) as compared to wild type (WT) mice. We evaluated the activities of the respiratory chain complexes II, II-III and IV, α-ketoglutarate dehydrogenase (α-KGDH), creatine kinase (CK) and synaptic Na(+), K(+)-ATPase. No differences of all evaluated parameters were detected in the Gcdh(-/-) relatively to the WT mice injected at baseline (Sal). Furthermore, mild increases of the activities of some respiratory chain complexes (II-III and IV) were observed in heart and skeletal muscle of Gcdh(-/-) and WT mice after Lys administration. However, the most marked effects provoked by Lys administration were marked decreases of the activities of Na(+), K(+)-ATPase in brain and CK in brain and skeletal muscle of Gcdh(-/-) mice. In contrast, brain α-KGDH activity was not altered in WT and Gcdh(-/-) injected with Sal or Lys. Our results demonstrate that reduction of Na(+), K(+)-ATPase and CK activities may play an important role in the pathogenesis of the neurodegenerative changes in GA I.
Collapse
Affiliation(s)
- Alexandre Umpierrez Amaral
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Jafari P, Braissant O, Bonafé L, Ballhausen D. The unsolved puzzle of neuropathogenesis in glutaric aciduria type I. Mol Genet Metab 2011; 104:425-37. [PMID: 21944461 DOI: 10.1016/j.ymgme.2011.08.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 08/23/2011] [Accepted: 08/23/2011] [Indexed: 12/22/2022]
Abstract
Glutaric aciduria type I (GA-I) is a cerebral organic aciduria caused by deficiency of glutaryl-Co-A dehydrogenase (GCDH). GCDH deficiency leads to accumulation of glutaric acid (GA) and 3-hydroxyglutaric acid (3-OHGA), two metabolites that are believed to be neurotoxic, in brain and body fluids. The disorder usually becomes clinically manifest during a catabolic state (e.g. intercurrent illness) with an acute encephalopathic crisis that results in striatal necrosis and in a permanent dystonic-dyskinetic movement disorder. The results of numerous in vitro and in vivo studies have pointed to three main mechanisms involved in the metabolite-mediated neuronal damage: excitotoxicity, impairment of energy metabolism and oxidative stress. There is evidence that during a metabolic crisis GA and its metabolites are produced endogenously in the CNS and accumulate because of limiting transport mechanisms across the blood-brain barrier. Despite extensive experimental work, the relative contribution of the proposed pathogenic mechanisms remains unclear and specific therapeutic approaches have yet to be developed. Here, we review the experimental evidence and try to delineate possible pathogenetic models and approaches for future studies.
Collapse
Affiliation(s)
- Paris Jafari
- Inborn Errors of Metabolism, Molecular Pediatrics, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1011 Lausanne, Switzerland
| | | | | | | |
Collapse
|
29
|
Wyse ATS, Netto CA. Behavioral and neurochemical effects of proline. Metab Brain Dis 2011; 26:159-72. [PMID: 21643764 DOI: 10.1007/s11011-011-9246-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 05/12/2011] [Indexed: 12/11/2022]
Abstract
Proline is an amino acid with an essential role for primary metabolism and physiologic functions. Hyperprolinemia results from the deficiency of specific enzymes for proline catabolism, leading to tissue accumulation of this amino acid. Hyperprolinemic patients can present neurological symptoms and brain abnormalities, whose aetiopathogenesis is poorly understood. This review addresses some of the findings obtained, mainly from animal studies, indicating that high proline levels may be associated to neuropathophysiology of some disorders. In this context, it has been suggested that energy metabolism deficit, Na(+),K(+)-ATPase, kinase creatine, oxidative stress, excitotoxicity, lipid content, as well as purinergic and cholinergic systems are involved in the effect of proline on brain damage and spatial memory deficit. The discussion focuses on the relatively low antioxidant defenses of the brain and the vulnerability of neural tissue to reactive species. This offers new perspectives for potential therapeutic strategies for this condition, which may include the early use of appropriate antioxidants as a novel adjuvant therapy, besides the usual treatment based on special diets poor in proline.
Collapse
Affiliation(s)
- Angela T S Wyse
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil.
| | | |
Collapse
|
30
|
Schmidt B, de Assis AM, Battu CE, Rieger DK, Hansen F, Sordi F, Longoni A, Hoefel AL, Farina M, Gonçalves CA, Souza DO, Santos Perry ML. Effects of glyoxal or methylglyoxal on the metabolism of amino acids, lactate, glucose and acetate in the cerebral cortex of young and adult rats. Brain Res 2010; 1315:19-24. [DOI: 10.1016/j.brainres.2009.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 11/30/2009] [Accepted: 12/03/2009] [Indexed: 11/25/2022]
|
31
|
Tonin AM, Ferreira GC, Schuck PF, Viegas CM, Zanatta A, Leipnitz G, Seminotti B, Duvall Wannmacher CM, Wajner M. Inhibition of creatine kinase activity by lysine in rat cerebral cortex. Metab Brain Dis 2009; 24:349-60. [PMID: 19370404 DOI: 10.1007/s11011-009-9131-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Accepted: 11/12/2008] [Indexed: 10/20/2022]
Abstract
Accumulation of lysine (Lys) in tissues and biochemical fluids is the biochemical hallmark of patients affected by familial hyperlysinemia (FH) and also by other inherited neurometabolic disorders. In the present study, we investigated the in vitro effect of Lys on various parameters of energy metabolism in cerebral cortex of 30-day-old Wistar rats. We verified that total (tCK) and cytosolic creatine kinase activities were significantly inhibited by Lys, in contrast to the mitochondrial isoform which was not affected by this amino acid. Furthermore, the inhibitory effect of Lys on tCK activity was totally prevented by reduced glutathione, suggesting a possible role of reactive species oxidizing critical thiol groups of the enzyme. In contrast, Lys did not affect (14)CO(2) production from [U-(14)C] glucose (aerobic glycolytic pathway) and [1-(14)C] acetic acid (citric acid cycle activity) neither the various activities of the electron transfer chain and synaptic Na(+)K(+)-ATPase at concentrations as high as 5.0 mM. Considering the importance of creatine kinase (CK) activity for brain energy metabolism homeostasis and especially ATP transfer and buffering, our results suggest that inhibition of this enzyme by Lys may contribute to the neurological signs presented by symptomatic patients affected by FH and other neurodegenerative disorders in which Lys accumulates.
Collapse
Affiliation(s)
- Anelise Miotti Tonin
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Rua Ramiro Barcelos, RS, Brasil
| | | | | | | | | | | | | | | | | |
Collapse
|