Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Commun Biol ; 5(1): 997, 2022 09 21.
Article in English | MEDLINE | ID: mdl-36131087

ABSTRACT

Hyperprolinemia type II (HPII) is an inborn error of metabolism due to genetic variants in ALDH4A1, leading to a deficiency in Δ-1-pyrroline-5-carboxylate (P5C) dehydrogenase. This leads to an accumulation of toxic levels of P5C, an intermediate in proline catabolism. The accumulating P5C spontaneously reacts with, and inactivates, pyridoxal 5'-phosphate, a crucial cofactor for many enzymatic processes, which is thought to be the pathophysiological mechanism for HPII. Here, we describe the use of a combination of LC-QTOF untargeted metabolomics, NMR spectroscopy and infrared ion spectroscopy (IRIS) to identify and characterize biomarkers for HPII that result of the spontaneous reaction of P5C with malonic acid and acetoacetic acid. We show that these biomarkers can differentiate between HPI, caused by a deficiency of proline oxidase activity, and HPII. The elucidation of their molecular structures yields insights into the disease pathophysiology of HPII.


Subject(s)
Proline Oxidase , Proline , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors , Biomarkers , Phosphates , Proline/metabolism , Proline Oxidase/genetics , Proline Oxidase/metabolism , Pyridoxal , Pyrroles
2.
Metab Brain Dis ; 36(6): 1413-1417, 2021 08.
Article in English | MEDLINE | ID: mdl-34037900

ABSTRACT

Hyperprolinemia type II (HPII) is a rare autosomal recessive disorder of proline degradation pathway due to deficiency of delta-1-pyrroline-5-carboxylate dehydrogenase. Pathogenic variants in the ALDH4A1 gene are responsible for this disorder. We here describe an 11-month-old infant with recurrent seizures refractory to multiple antiepileptic drugs. She was hospitalized in view of acute-onset encephalopathy, exacerbation of generalized seizures following an upper respiratory infection. Laboratory investigation revealed significantly elevated proline levels in dried blood spots. DNA sample of the child was subjected to a targeted next-generation sequencing gene panel for hyperprolinemias. We detected a novel nonsense homozygous variant in the ALDH4A1 gene in the child and the heterozygous variant of the same in both the parents. Based on the location of the variant i.e. in the last exon, truncated protein is expected to be expressed by skipping nonsense-mediated decay and such point-nonsense variants could be an ideal target for readthrough drugs to correct genetic defects.


Subject(s)
1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , 1-Pyrroline-5-Carboxylate Dehydrogenase/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Epilepsy/genetics , Amino Acid Metabolism, Inborn Errors/complications , Brain/diagnostic imaging , Codon, Nonsense , DNA/genetics , Drug Resistant Epilepsy/genetics , Electroencephalography , Epilepsy/etiology , Female , Genetic Variation , Humans , Infant , Magnetic Resonance Imaging , Proline/blood , Proline/genetics
3.
BMC Neurol ; 19(1): 345, 2019 Dec 29.
Article in English | MEDLINE | ID: mdl-31884946

ABSTRACT

BACKGROUND: Hyperprolinemia type 2 (HPII) is a rare autosomal recessive disorder of the proline metabolism, that affects the ALDH4A1 gene. So far only four different pathogenic mutations are known. The manifestation is mostly in neonatal age, in early infancy or early childhood. CASE PRESENTATION: The 64-years female patient had a long history of abdominal pain, and episode of an acute neuritis. Ten years later she was admitted into the neurological intensive-care-unit with acute abdominal pain, multiple generalized epileptic seizures, a vertical gaze palsy accompanied by extensive lactic acidosis in serum 26.0 mmol/l (reference: 0.55-2.2 mmol/l) and CSF 12.01 mmol/l (reference: 1.12-2.47 mmol/l). Due to repeated epileptic seizures and secondary complications a long-term sedation with a ventilation therapy over 20 days was administered. A diagnostic work-up revealed up to 400-times increased prolin-level in urine CSF and blood. Furthermore, a low vitamin-B6 serum value was found, consistent with a HPII causing secondary pyridoxine deficiency and seizures. The ALDH4A1 gene sequencing confirmed two previously unknown compound heterozygous variants (ALDH4A1 gene (NM_003748.3) Intron 1: c.62 + 1G > A - heterozygous and ALDH4A1 gene (NM_003748.3) Exon 5 c.349G > C, p.(Asp117His) - heterozygous). Under high-dose vitamin-B6 therapy no further seizures occurred. CONCLUSION: We describe two novel ALDH4A1-variants in an adult patient with hyperprolinemia type II causing secondary pyridoxine deficiency and seizures. Severe and potentially life-threatening course of this treatable disease emphasizes the importance of diagnostic vigilance and thorough laboratory work-up including gene analysis even in cases with atypical late manifestation.


Subject(s)
1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/genetics , 1-Pyrroline-5-Carboxylate Dehydrogenase/genetics , Acidosis, Lactic/etiology , Adult , Amino Acid Metabolism, Inborn Errors/complications , Female , Humans , Mutation , Status Epilepticus/etiology
4.
Arch Biochem Biophys ; 632: 142-157, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28712849

ABSTRACT

Proline has important roles in multiple biological processes such as cellular bioenergetics, cell growth, oxidative and osmotic stress response, protein folding and stability, and redox signaling. The proline catabolic pathway, which forms glutamate, enables organisms to utilize proline as a carbon, nitrogen, and energy source. FAD-dependent proline dehydrogenase (PRODH) and NAD+-dependent glutamate semialdehyde dehydrogenase (GSALDH) convert proline to glutamate in two sequential oxidative steps. Depletion of PRODH and GSALDH in humans leads to hyperprolinemia, which is associated with mental disorders such as schizophrenia. Also, some pathogens require proline catabolism for virulence. A unique aspect of proline catabolism is the multifunctional proline utilization A (PutA) enzyme found in Gram-negative bacteria. PutA is a large (>1000 residues) bifunctional enzyme that combines PRODH and GSALDH activities into one polypeptide chain. In addition, some PutAs function as a DNA-binding transcriptional repressor of proline utilization genes. This review describes several attributes of PutA that make it a remarkable flavoenzyme: (1) diversity of oligomeric state and quaternary structure; (2) substrate channeling and enzyme hysteresis; (3) DNA-binding activity and transcriptional repressor function; and (4) flavin redox dependent changes in subcellular location and function in response to proline (functional switching).


Subject(s)
1-Pyrroline-5-Carboxylate Dehydrogenase/chemistry , Bacterial Proteins/chemistry , Flavoproteins/chemistry , Gram-Negative Bacteria/enzymology , Membrane Proteins/chemistry , Proline Oxidase/chemistry , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , 1-Pyrroline-5-Carboxylate Dehydrogenase/genetics , 1-Pyrroline-5-Carboxylate Dehydrogenase/metabolism , Amino Acid Metabolism, Inborn Errors , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/genetics , Flavin-Adenine Dinucleotide/metabolism , Flavoproteins/genetics , Flavoproteins/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proline/chemistry , Proline/genetics , Proline/metabolism , Proline Oxidase/genetics , Proline Oxidase/metabolism
5.
J Child Adolesc Ment Health ; 26(2): 109-24, 2014.
Article in English | MEDLINE | ID: mdl-25391710

ABSTRACT

Childhood onset schizophrenia (COS) is diagnosed before the age of 13 years, and early onset schizophrenia (EOS) is diagnosed before the age of 18 years. EOS is considered extremely rare and its prevalence in comparison to the worldwide prevalence of schizophrenia (1%) has not adequately been studied. Patients who experience the first episode of psychosis need to be treated early and optimally to lessen the morbidity and improve the outcome of the illness. Treatment needs to be a combination of both pharmacological and non-pharmacological modalities. Pharmacological intervention is necessary for remission, improvement of positive symptoms and to aid with the efficacy of psychosocial interventions. There is a lack of efficacy and safety data of the use of antipsychotic medication in children, with most of the information available being extrapolations of adult data. An increased use of atypical antipsychotic drugs in the treatment of EOS has been accompanied by growing concern about the appropriate use and associated side effects in children and adolescents. This update highlights new developments, concepts and treatment trends in EOS.


Subject(s)
Schizophrenia/therapy , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Administration, Oral , Adolescent , Algorithms , Amino Acid Metabolism, Inborn Errors/chemically induced , Antipsychotic Agents/therapeutic use , Basal Ganglia Diseases/chemically induced , Cardiovascular Diseases/chemically induced , Child , Cognition Disorders/psychology , Cognitive Behavioral Therapy/methods , Delayed-Action Preparations , Humans , Marijuana Abuse/complications , Metabolic Syndrome/chemically induced , Neuroimaging/methods , Neuroleptic Malignant Syndrome/etiology , Proline Oxidase/deficiency , Puberty/physiology , Risk Factors , Schizophrenia/diagnosis , Schizophrenia/etiology , Seizures/chemically induced , Treatment Outcome , Weight Gain/drug effects
6.
Int J Biochem Cell Biol ; 54: 20-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24980685

ABSTRACT

The present study investigated the effects of hyperprolinemia on oxidative damage to biomolecules (protein, lipids and DNA) and the antioxidant status in blood of rats. The influence of the antioxidants on the effects elicited by proline was also examined. Wistar rats received two daily injections of proline and/or vitamin E plus C (6th-28th day of life) and were killed 12h after the last injection. Results showed that hyperprolinemia induced a significant oxidative damage to proteins, lipids and DNA demonstrated by increased carbonyl content, malondialdehyde levels and a greater damage index in comet assay, respectively. The concomitant antioxidants administration to proline treatment completely prevented oxidative damage to proteins, but partially prevented lipids and DNA damage. We also observed that the non-enzymatic antioxidant potential was decreased by proline treatment and partially prevented by antioxidant supplementation. The plasma levels of vitamins E and C significantly increased in rats treated exogenously with these vitamins but, interestingly, when proline was administered concomitantly with vitamin E plus C, the levels of these vitamins were similar to those found in plasma of control and proline rats. Our findings suggest that hyperprolinemia promotes oxidative damage to the three major classes of macromolecules in blood of rats. These effects were accomplished by decrease in non-enzymatic antioxidant potential and decrease in vitamins administered exogenously, which significantly decreased oxidative damage to biomolecules studied. These data suggest that antioxidants may be an effective adjuvant therapeutic to limit oxidative damage caused by proline.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Antioxidants/pharmacology , DNA Damage/drug effects , DNA/chemistry , Lipids/chemistry , Oxidative Stress/drug effects , Proline Oxidase/deficiency , Proteins/chemistry , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Animals , Ascorbic Acid/pharmacology , Dietary Supplements , Male , Malondialdehyde/metabolism , Oxidation-Reduction , Proline/chemistry , Rats , Rats, Wistar , Vitamin E/pharmacology , Vitamins/pharmacology
7.
Pediatr Int ; 56(4): 492-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24931297

ABSTRACT

There are two classifications of hereditary hyperprolinemia: type I (HPI) and type II (HPII). Each type is caused by an autosomal recessive inborn error of the proline metabolic pathway. HPI is caused by an abnormality in the proline-oxidizing enzyme (POX). HPII is caused by a deficiency of Δ-1-pyrroline-5-carboxylate (P5C) dehydrogenase (P5CDh). The clinical features of HPI are unclear. Nephropathy, uncontrolled seizures, mental retardation or schizophrenia have been reported in HPI, but a benign phenotype without neurological problems has also been reported. The clinical features of HPII are also unclear. In addition, the precise incidences of HPI and HPII are unknown. Only two cases of HPI and one case of HPII have been identified in Japan through a questionnaire survey and by a study of previous reports. This suggests that hyperprolinemia is a very rare disease in Japan, consistent with earlier reports in Western countries. The one case of HPII found in Japan was diagnosed in an individual with influenza-associated encephalopathy. This suggests that HPII might reduce the threshold for convulsions, thereby increasing the sensitivity of individuals with influenza-associated encephalopathy. The current study presents diagnostic criteria for HPI and HPII, based on plasma proline level, with or without measurements of urinary P5C. In the future, screening for HPI and HPII in healthy individuals, or patients with relatively common diseases such as developmental disabilities, epilepsy, schizophrenia or behavioral problems will be important.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Proline/metabolism , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/epidemiology , Humans , Japan , Proline Oxidase/genetics
8.
Psychiatr Genet ; 24(4): 172-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24842239

ABSTRACT

The neuropsychiatric phenotype associated with hyperprolinemia type I (HPI) is still under debate. To our knowledge, no long-term follow-up on patients with HPI has been reported so far. We have previously described the clinical, biochemical, and molecular features of four patients with HPI. Here, we report on the neuropsychiatric and genotype features of an expanded sample of 10 patients with HPI with a mean follow-up duration of 11 years. Epileptic manifestations and/or cognitive impairment were prevalent at onset, but they were subsequently replaced by psychiatric disorders. Social behavior and relational skills were considerably impaired in the majority of cases. Learning disability was present in one patient. The complex neurochemical effects of proline on the central nervous system and genotype/phenotype correlations were discussed.


Subject(s)
Amino Acid Metabolism, Inborn Errors/psychology , Neuropsychological Tests , Proline Oxidase/deficiency , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Adolescent , Amino Acid Metabolism, Inborn Errors/blood , Child , Female , Follow-Up Studies , Humans , Intelligence Tests , Male , Proline/blood , Proline Oxidase/blood , Time Factors , Young Adult
9.
Schizophr Res ; 156(1): 15-22, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24787057

ABSTRACT

25-Hydroxyvitamin D (25(OH)D) deficits have been associated with schizophrenia susceptibility and supplementation has been recommended for those at-risk. Although the mechanism by which a deficit confers risk is unknown, vitamin D is a potent transcriptional modulator and can regulate proline dehydrogenase (PRODH) expression. PRODH maps to chromosome 22q11, a region conferring the highest known genetic risk of schizophrenia, and encodes proline oxidase, which catalyzes proline catabolism. l-Proline is a neuromodulator at glutamatergic synapses, and peripheral hyperprolinemia has been associated with decreased IQ, cognitive impairment, schizoaffective disorder, and schizophrenia. We investigated the relationship between 25(OH)D and schizophrenia, comparing fasting plasma 25(OH)D in 64 patients and 90 matched controls. We then tested for a mediating effect of hyperprolinemia on the association between 25(OH)D and schizophrenia. 25(OH)D levels were significantly lower in patients, and 25(OH)D insufficiency associated with schizophrenia (OR 2.1, adjusted p=0.044, 95% CI: 1.02-4.46). Moreover, 25(OH)D insufficient subjects had three times greater odds of hyperprolinemia than those with optimal levels (p=0.035, 95% CI: 1.08-8.91), and formal testing established that hyperprolinemia is a significantly mediating phenotype that may explain over a third of the effect of 25(OH)D insufficiency on schizophrenia risk. This study presents a mechanism by which 25(OH)D insufficiency confers risk of schizophrenia; via proline elevation due to reduced PRODH expression, and a concomitant dysregulation of neurotransmission. Although definitive causality cannot be confirmed, these findings strongly support vitamin D supplementation in patients, particularly for those with elevated proline, who may represent a large subgroup of the schizophrenia population.


Subject(s)
Amino Acid Metabolism, Inborn Errors/complications , Genetic Predisposition to Disease/genetics , Proline Oxidase/deficiency , Schizophrenia , Vitamin D Deficiency/complications , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Adolescent , Adult , Aged , Amino Acid Metabolism, Inborn Errors/blood , Fasting/blood , Female , Humans , Male , Middle Aged , Models, Statistical , Mutation/genetics , Proline/metabolism , Proline Oxidase/blood , Proline Oxidase/genetics , Risk Factors , Schizophrenia/blood , Schizophrenia/etiology , Schizophrenia/genetics , Vitamin D/analogs & derivatives , Vitamin D/blood , Young Adult
10.
J Inherit Metab Dis ; 37(3): 383-90, 2014 May.
Article in English | MEDLINE | ID: mdl-24173411

ABSTRACT

Although hyperprolinemia type-II has a discriminative metabolic phenotype and is frequently associated with neurological system involvement, the casual relation between the metabolic abnormalities and the clinical features, except for those of the secondary B6 deficiency, has been frequently debated. In order to evaluate disease frequency and the neuro-metabolic outcome we searched our laboratory database between 1992 and 2010, including 20,991 urinary organic acid profiles. From these individuals 16,720 parallel blood samples were available, and were investigated by serum amino acid analysis. We also evaluated the clinical, neurological, psychological features, laboratory data and vitamin levels and therapeutic effect in metabolically confirmed hyperprolinemia. Due to the mitochondrial localization of both ALDH4A1 and PRODH mitochondrial enzyme complex activity was evaluated and oxygen consumption was measured to assess ATP production in patient-fibroblasts. The Mitochondrial Disease Score was used to evaluate clinical mitochondrial dysfunction. The child behavior checklist was used to screen for psychopathology. We found four patients with increased urinary P5C diagnosed with hyperprolinemia type II, and only one patient had hyperprolinemia type I. All children with hyperprolinemia type II had low normal B6 concentration, and three of the patients had biochemical markers suggesting mitochondrial dysfunction. Mitochondrial dysfunction was confirmed in a muscle biopsy in one case. Intellectual disability was found in two adolescent patients. All patients showed seizures and significant behavioral problems, including anxiety and hallucinations. The clinical course was non-progressive and independent from the B6 concentration and B6 therapy. Hyperprolinemia is a rare inborn error. Individuals with hyperprolinemia should be monitored closely due to their frequent behavioral problems.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Mitochondrial Diseases/etiology , Proline Oxidase/deficiency , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Adenosine Triphosphate/biosynthesis , Adolescent , Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/psychology , Amino Acid Metabolism, Inborn Errors/therapy , Biopsy , Child , Female , Humans , Male , Mitochondria/metabolism , Mitochondrial Diseases/diagnosis , Muscles/pathology , Proline Oxidase/metabolism , Treatment Outcome , Vitamin B 6/administration & dosage
11.
Ann Clin Lab Sci ; 43(1): 31-6, 2013.
Article in English | MEDLINE | ID: mdl-23462603

ABSTRACT

BACKGROUND: Hyperprolinemia is a rare inherited metabolic disorder characterized by a high proline level in blood and/or urine and various neuropsychiatric symptoms. Type I hyperprolinemia is caused by a proline oxidase deficiency, which is encoded by the PRODH gene on chromosome 22q11. Herein, we present a study of Korean patients with type I hyperprolinemia who were diagnosed during newborn screening by tandem mass spectrometry and confirmed by molecular analysis. METHODS: Four neonates were referred to our hospital for workup of high proline levels in newborn screening test. We analyzed the biochemical findings and the PRODH gene was amplified by long-range PCR to confirm molecular genetic abnormalities. RESULTS: All patients had high plasma proline levels, ranging from 742 to 1192 µmol/L (reference range, 77.4 - 244.6 µmol/L). In molecular analysis, 4 disease-associated mutant alleles were identified: c.1414G>A (p.A472T), c.1279G>A (p.V427M), c.1357C>T (p.R453C) and c.1562A>G (p.Q521R). All mutations were missense and c.1279G>A included the majority of mutant alleles. No relationships between type of mutation and clinical outcomes were observed. CONCLUSION: We found that distinct molecular alterations of the PRODH gene result in abnormal proline levels. Newborn screening and molecular analysis are necessary to identify patients before clinical expression of metabolic disease.


Subject(s)
Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Asian People/genetics , Proline Oxidase/genetics , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , DNA Mutational Analysis , Female , Humans , Infant, Newborn , Male , Mutation/genetics , Proline Oxidase/deficiency , Republic of Korea
12.
Neurochem Res ; 37(1): 205-13, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21935728

ABSTRACT

This study investigated the effects of acute and chronic hyperprolinemia on glutamate uptake, as well as some mechanisms underlying the proline effects on glutamatergic system in rat cerebral cortex. The protective role of guanosine on effects mediated by proline was also evaluated. Results showed that acute and chronic hyperprolinemia reduced glutamate uptake, Na(+), K(+)-ATPase activity, ATP levels and increased lipoperoxidation. GLAST and GLT-1 immunocontent were increased in acute, but not in chronic hyperprolinemic rats. Our data suggest that the effects of proline on glutamate uptake may be mediated by lipid peroxidation and disruption of Na(+), K(+)-ATPase activity, but not by decreasing in glutamate transporters. This probably induces excitotoxicity and subsequent energy deficit. Guanosine was effective to prevent most of the effects promoted by proline, reinforcing its modulator role in counteracting the glutamate toxicity. However, further studies are needed to assess the modulatory effects of guanosine on experimental hyperprolinemia.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Brain/physiopathology , Glutamic Acid/metabolism , Guanosine/pharmacology , Homeostasis , Neuroprotective Agents/pharmacology , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Proline Oxidase/deficiency , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
13.
J Cell Biochem ; 113(1): 174-83, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21882227

ABSTRACT

The present study investigated the effects of chronic hyperprolinemia on oxidative and metabolic status in liver and serum of rats. Wistar rats received daily subcutaneous injections of proline from their 6th to 28th day of life. Twelve hours after the last injection the rats were sacrificed and liver and serum were collected. Results showed that hyperprolinemia induced a significant reduction in total antioxidant potential and thiobarbituric acid-reactive substances. The activities of the antioxidant enzymes catalase and superoxide dismutase were significantly increased after chronic proline administration, while glutathione (GSH) peroxidase activity, dichlorofluorescin oxidation, GSH, sulfhydryl, and carbonyl content remained unaltered. Histological analyses of the liver revealed that proline treatment induced changes of the hepatic microarchitecture and increased the number of inflammatory cells and the glycogen content. Biochemical determination also demonstrated an increase in glycogen concentration, as well as a higher synthesis of glycogen in liver of hyperprolinemic rats. Regarding to hepatic metabolism, it was observed an increase on glucose oxidation and a decrease on lipid synthesis from glucose. However, hepatic lipid content and serum glucose levels were not changed. Proline administration did not alter the aminotransferases activities and serum markers of hepatic injury. Our findings suggest that hyperprolinemia alters the liver homeostasis possibly by induction of a mild degree of oxidative stress and metabolic changes. The hepatic alterations caused by proline probably do not implicate in substantial hepatic tissue damage, but rather demonstrate a process of adaptation of this tissue to oxidative stress. However, the biological significance of these findings requires additional investigation.


Subject(s)
Amino Acid Metabolism, Inborn Errors/chemically induced , Amino Acid Metabolism, Inborn Errors/metabolism , Liver/metabolism , Oxidative Stress , Proline/administration & dosage , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Animals , Antioxidants/analysis , Blood Glucose/analysis , Catalase/metabolism , Female , Fluoresceins/metabolism , Glutathione/analysis , Glutathione Peroxidase/metabolism , Glycogen/biosynthesis , Lipids/biosynthesis , Male , Proline Oxidase/deficiency , Proline Oxidase/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/analysis
14.
Prog Neuropsychopharmacol Biol Psychiatry ; 36(2): 258-63, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22019856

ABSTRACT

Hyperprolinemia is an inherited disorder of proline metabolism and patients affected by this disease may present neurological manifestations, including seizures and cognitive dysfunctions. Moreover, an association between adulthood schizoaffective disorders and moderate hyperprolinemia has been reported. However, the mechanisms underlying these behavioral phenotypes still remain unclear. In the present study, we investigated the effect of proline treatments on behavioral parameters in zebrafish, such as locomotor activity, anxiety, and social interaction. Adult zebrafish (Danio rerio) were exposed to proline (1.5 and 3.0 mM) during 1h or 7 days (short- or long-term treatments, respectively). Short-term proline exposure did not promote significant changes on the behavioral parameters observed. Long-term exposure at 1.5 mM proline significantly increased the number of line crossing (47%), the total distance (29%), and the mean speed (33%) when compared to control group. A significant increase in the time spent in the upper portion of the test tank was also observed after this treatment (91%), which may be interpreted as an indicator of anxiolytic behavior. Proline at 1.5 mM also induced social interaction impairment (78%), when compared to the untreated group after long-term treatment. Moreover, these proline-induced behavioral changes in zebrafish were completely reversed by acute administration of an atypical antipsychotic drug (sulpiride), but not by a typical (haloperidol). These findings demonstrate that proline is able to induce schizophrenia-like symptoms in zebrafish, which reinforce the use of this species as a complementary vertebrate model for studying behavioral phenotypes associated with neurological dysfunctions characteristic of metabolic diseases.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Antipsychotic Agents/therapeutic use , Anxiety/drug therapy , Locomotion/drug effects , Proline/antagonists & inhibitors , Proline/pharmacology , Social Behavior , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/chemically induced , Amino Acid Metabolism, Inborn Errors/complications , Animals , Antipsychotic Agents/pharmacology , Anxiety/complications , Disease Models, Animal , Drug Interactions , Female , Haloperidol/pharmacology , Haloperidol/therapeutic use , Male , Proline Oxidase/deficiency , Sulpiride/pharmacology , Sulpiride/therapeutic use , Time Factors , Zebrafish
15.
Neurochem Res ; 36(12): 2306-15, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21792675

ABSTRACT

This study investigated whether physical exercise would reverse proline-induced performance deficits in water maze tasks, as well as its effects on brain-derived neurotrophic factor (BDNF) immunocontent and brain acetylcholinesterase (AChE) activity in Wistar rats. Proline administration followed partial time (6th-29th day of life) or full time (6th-60th day of life) protocols. Treadmill exercise was performed from 30th to 60th day of life, when behavioral testing was started. After that, animals were sacrificed for BDNF and AChE determination. Results show that proline impairs cognitive performance, decreases BDNF in cerebral cortex and hippocampus and increases AChE activity in hippocampus. All reported effects were prevented by exercise. These results suggest that cognitive, spatial learning/memory, deficits caused by hyperprolinemia may be associated, at least in part, to the decrease in BDNF levels and to the increase in AChE activity, as well as support the role of physical exercise as a potential neuroprotective strategy.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Cognition Disorders/therapy , Maze Learning/drug effects , Memory/drug effects , Physical Conditioning, Animal , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Acetylcholinesterase/metabolism , Amino Acid Metabolism, Inborn Errors/psychology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Male , Proline Oxidase/deficiency , Rats , Rats, Wistar
16.
Proc Natl Acad Sci U S A ; 108(32): 13041-6, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21788482

ABSTRACT

The larva of the drosophilid fly, Chymomyza costata, is probably the most complex metazoan organism that can survive submergence in liquid nitrogen (-196 °C) in a fully hydrated state. We examined the associations between the physiological and biochemical parameters of differently acclimated larvae and their freeze tolerance. Entering diapause is an essential and sufficient prerequisite for attaining high levels of survival in liquid nitrogen (23% survival to adult stage), although cold acclimation further improves this capacity (62% survival). Profiling of 61 different metabolites identified proline as a prominent compound whose concentration increased from 20 to 147 mM during diapause transition and subsequent cold acclimation. This study provides direct evidence for the essential role of proline in high freeze tolerance. We increased the levels of proline in the larval tissues by feeding larvae proline-augmented diets and found that this simple treatment dramatically improved their freeze tolerance. Cell and tissue survival following exposure to liquid nitrogen was evident in proline-fed nondiapause larvae, and survival to adult stage increased from 0% to 36% in proline-fed diapause-destined larvae. A significant statistical correlation was found between the whole-body concentration of proline, either natural or artificial, and survival to the adult stage in liquid nitrogen for diapause larvae. Differential scanning calorimetry analysis suggested that high proline levels, in combination with a relatively low content of osmotically active water and freeze dehydration, increased the propensity of the remaining unfrozen water to undergo a glass-like transition (vitrification) and thus facilitated the prevention of cryoinjury.


Subject(s)
Adaptation, Physiological/drug effects , Amino Acid Metabolism, Inborn Errors/veterinary , Cryopreservation , Drosophilidae/drug effects , Drosophilidae/physiology , Nitrogen/pharmacology , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Acclimatization/drug effects , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Body Water/drug effects , Calorimetry, Differential Scanning , Diet , Feeding Behavior/drug effects , Freezing , Glass , Larva/drug effects , Larva/physiology , Osmosis/drug effects , Principal Component Analysis , Proline/metabolism , Proline Oxidase/deficiency , Survival Analysis
17.
Metab Brain Dis ; 26(3): 159-72, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21643764

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.


Subject(s)
Antioxidants , Brain Diseases, Metabolic , Energy Metabolism/physiology , Free Radicals/adverse effects , Memory Disorders , Oxidative Stress/physiology , Proline , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Antioxidants/metabolism , Antioxidants/therapeutic use , Ascorbic Acid/therapeutic use , Brain/metabolism , Brain Diseases, Metabolic/chemically induced , Brain Diseases, Metabolic/metabolism , Creatine Kinase/metabolism , Glycine/metabolism , Glycine/urine , Gyrate Atrophy/metabolism , Humans , Memory Disorders/chemically induced , Memory Disorders/metabolism , Mice , Proline/adverse effects , Proline/metabolism , Proline Oxidase/deficiency , Proline Oxidase/metabolism , Rats , Receptors, Cholinergic/metabolism , Receptors, Purinergic/metabolism , Renal Tubular Transport, Inborn Errors/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Vitamin E/therapeutic use
18.
Schizophr Res ; 131(1-3): 139-45, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21645996

ABSTRACT

There are multiple genetic links between schizophrenia and a deficit of proline dehydrogenase (PRODH) enzyme activity. However, reports testing for an association of schizophrenia with the resulting proline elevation have been conflicting. The objectives of this study were to investigate whether hyperprolinemia is associated with schizophrenia, and to measure the relationship between plasma proline, and clinical features and symptoms of schizophrenia. We performed a cross-sectional case-control study, comparing fasting plasma proline in 90 control subjects and 64 schizophrenic patients and testing for association of mild to moderate hyperprolinemia with schizophrenia. As secondary analyses, the relationship between hyperprolinemia and five measures of clinical onset, symptoms and outcome were investigated. Patients had significantly higher plasma proline than matched controls (p<0.0001), and categorical analysis of gender adjusted hyperprolinemia showed a significant association with schizophrenia (OR 6.15, p=0.0003). Hyperprolinemic patients were significantly older at their first hospitalization (p=0.015 following correction for multiple testing). While plasma proline level was not related to total, positive or negative symptoms, hyperprolinemic status had a significant effect on length of hospital stay (p=0.005), following adjustment for race, BPRS score, and cross-sectional time from admission to proline measurement. Mild to moderate hyperprolinemia is a significant risk factor for schizophrenia, and may represent an intermediate phenotype in the disease. Hyperprolinemic patients have a significantly later age of first psychiatric hospitalization, suggestive of later onset, and hospital stays 46% longer than non-hyperprolinemic subjects. These findings have implications in the etiology of schizophrenia, and for the clinical management of these patients.


Subject(s)
Amino Acid Metabolism, Inborn Errors/blood , Amino Acid Metabolism, Inborn Errors/complications , Proline/blood , Schizophrenia/blood , Schizophrenia/complications , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Adolescent , Adult , Aged , Amino Acid Metabolism, Inborn Errors/drug therapy , Analysis of Variance , Brief Psychiatric Rating Scale , Case-Control Studies , Chi-Square Distribution , Cross-Sectional Studies , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Middle Aged , Proline Oxidase/blood , Proline Oxidase/deficiency , Schizophrenia/drug therapy , Valproic Acid/therapeutic use , Young Adult
19.
Metab Brain Dis ; 26(2): 141-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21509571

ABSTRACT

Considering that Na(+),K(+)-ATPase is an embedded-membrane enzyme and that experimental chronic hyperprolinemia decreases the activity of this enzyme in brain synaptic plasma membranes, the present study investigated the effect of chronic proline administration on thiobarbituric acid-reactive substances, as well as the influence of antioxidant vitamins E plus C on the effects mediated by proline on Na(+),K(+)-ATPase activity in cerebral cortex of rats. The expression of Na(+),K(+)-ATPase catalytic subunits was also evaluated. Results showed that proline increased thiobarbituric acid-reactive substances, suggesting an increase of lipid peroxidation. Furthermore, concomitant administration of vitamins E plus C significantly prevented the increase of lipid peroxidation, as well as the inhibition of Na(+),K(+)-ATPase activity caused by proline. We did not observe any change in levels of Na(+),K(+)-ATPase mRNA transcripts after chronic exposure to proline and vitamins E plus C. These findings provide insights into the mechanisms through which proline exerts its effects on brain function and suggest that treatment with antioxidants may be beneficial to treat neurological dysfunctions present in hyperprolinemic patients.


Subject(s)
Antioxidants , Ascorbic Acid , Cerebral Cortex/enzymology , Lipid Peroxidation/drug effects , Sodium-Potassium-Exchanging ATPase/drug effects , Vitamin E , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/chemically induced , Amino Acid Metabolism, Inborn Errors/metabolism , Analysis of Variance , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Ascorbic Acid/metabolism , Ascorbic Acid/pharmacology , Cerebral Cortex/drug effects , Disease Models, Animal , Drug Synergism , Gene Expression/drug effects , Humans , Oxidative Stress/drug effects , Proline/administration & dosage , Proline/adverse effects , Proline Oxidase/deficiency , Proline Oxidase/metabolism , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Synaptic Membranes/drug effects , Thiobarbituric Acid Reactive Substances/metabolism , Vitamin E/metabolism , Vitamin E/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL