Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Asian J Androl ; 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38305695

ABSTRACT

ABSTRACT: Oxidative stress is one of the main mechanisms responsible for male infertility. Various conditions such as varicocele, obesity, advanced age, and lifestyle can lead to an increase in reactive oxygen species, causing an oxidative imbalance in the reproductive environment. Spermatozoa are sensitive to reactive oxygen species and require energy to carry out their main function of fertilizing the egg. Excessive reactive oxygen species can affect sperm metabolism, leading to immobility, impaired acrosome reaction, and cell death, thereby impairing reproductive success. This double-blind randomized study evaluated the effect of supplementation with L-carnitine, acetyl-L-carnitine, vitamins, and other nutrients on semen quality in 104 infertile patients with or without varicocele, while also investigating the impact of factors such as obesity and advanced age on treatment. Sperm concentration significantly increased in the supplemented group compared to the placebo group (P = 0.0186). Total sperm count also significantly increased in the supplemented group (P = 0.0117), as did sperm motility (P = 0.0120). The treatment had a positive effect on patients up to 35 years of age in terms of sperm concentration (P = 0.0352), while a body mass index (BMI) above 25 kg m-2 had a negative effect on sperm concentration (P = 0.0110). Results were not showing a net benefit in stratifying patients in accordance with their BMI since sperm quality increase was not affected by this parameter. In conclusion, antioxidant supplementation may be beneficial for infertile patients and has a more positive effect on younger patients with a normal weight.

2.
Biomedicines ; 11(9)2023 Sep 06.
Article in English | MEDLINE | ID: mdl-37760915

ABSTRACT

Carnitines play a key physiological role in oocyte metabolism and redox homeostasis. In clinical and animal studies, carnitine administration alleviated metabolic and reproductive dysfunction associated with polycystic ovarian syndrome (PCOS). Oxidative stress (OS) at systemic, intraovarian, and intrafollicular levels is one of the main factors involved in the pathogenesis of PCOS. We investigated the ability of different acyl-carnitines to act at the oocyte level by counteracting the effects of OS on carnitine shuttle system and mitochondrial activity in mouse oocytes. Germinal vesicle (GV) oocytes were exposed to hydrogen peroxide and propionyl-l-carnitine (PLC) alone or in association with l-carnitine (LC) and acetyl-l-carnitine (ALC) under different conditions. Expression of carnitine palmitoyltransferase-1 (Cpt1) was monitored by RT-PCR. In in vitro matured oocytes, metaphase II (MII) apparatus was assessed by immunofluorescence. Oocyte mitochondrial respiration was evaluated by Seahorse Cell Mito Stress Test. We found that Cpt1a and Cpt1c isoforms increased under prooxidant conditions. PLC alone significantly improved meiosis completion and oocyte quality with a synergistic effect when combined with LC + ALC. Acyl-carnitines prevented Cpt1c increased expression, modifications of oocyte respiration, and ATP production observed upon OS. Specific effects of PLC on spare respiratory capacity were observed. Therefore, carnitine supplementation modulated the intramitochondrial transfer of fatty acids with positive effects on mitochondrial activity under OS. This knowledge contributes to defining molecular mechanism underlying carnitine efficacy on PCOS.

3.
Antioxidants (Basel) ; 11(4)2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35453430

ABSTRACT

Recently, the importance of bioenergetics in the reproductive process has emerged. For its energetic demand, the oocyte relies on numerous mitochondria, whose activity increases during embryo development under a fine regulation to limit ROS production. Healthy oocyte mitochondria require a balance of pyruvate and fatty acid oxidation. Transport of activated fatty acids into mitochondria requires carnitine. In this regard, the interest in the role of carnitines as mitochondrial modulators in oocyte and embryos is increasing. Carnitine pool includes the un-esterified l-carnitine (LC) and carnitine esters, such as acetyl-l-carnitine (ALC) and propionyl-l-carnitine (PLC). In this review, carnitine medium supplementation for counteracting energetic and redox unbalance during in vitro culture and cryopreservation is reported. Although most studies have focused on LC, there is new evidence that the addition of ALC and/or PLC may boost LC effects. Pathways activated by carnitines include antiapoptotic, antiglycative, antioxidant, and antiinflammatory signaling. Nevertheless, the potential of carnitine to improve energetic metabolism and oocyte and embryo competence remains poorly investigated. The importance of carnitine as a mitochondrial modulator may suggest that this molecule may exert a beneficial role in ovarian disfunctions associated with metabolic and mitochondrial alterations, including PCOS and reproductive aging.

4.
Arch Toxicol ; 95(8): 2769-2784, 2021 08.
Article in English | MEDLINE | ID: mdl-34164711

ABSTRACT

Mitochondrial deregulation has emerged as one of the earliest pathological events in Alzheimer's disease (AD), the most common age-related neurodegenerative disorder. Improvement of mitochondrial function in AD has been considered a relevant therapeutic approach. L-carnitine (LC), an amino acid derivative involved in the transport of long-chain fatty acids into mitochondria, was previously demonstrated to improve mitochondrial function, having beneficial effects in neurological disorders; moreover, acetyl-L-carnitine (ALC) is currently under phase 4 clinical trial for AD (ClinicalTrials.gov NCT01320527). Thus, in the present study, we investigated the impact of different forms of carnitines, namely LC, ALC and propionyl-L-carnitine (PLC) on mitochondrial toxicity induced by amyloid-beta peptide 1-42 oligomers (AßO; 1 µM) in mature rat hippocampal neurons. Our results indicate that 5 mM LC, ALC and PLC totally rescued the mitochondrial membrane potential and alleviated both the decrease in oxygen consumption rates and the increase in mitochondrial fragmentation induced by AßO. These could contribute to the prevention of neuronal death by apoptosis. Moreover, only ALC ameliorated AßO-evoked changes in mitochondrial movement by reducing the number of stationary mitochondria and promoting reversal mitochondrial movement. Data suggest that carnitines (LC, ALC and PLC) may act differentially to counteract changes in mitochondrial function and movement in neurons subjected to AßO, thus counteracting AD-related pathological phenotypes.


Subject(s)
Acetylcarnitine/pharmacology , Alzheimer Disease/drug therapy , Carnitine/analogs & derivatives , Neuroprotective Agents/pharmacology , Alzheimer Disease/physiopathology , Animals , Apoptosis/drug effects , Carnitine/pharmacology , Cells, Cultured , Female , Hippocampus/drug effects , Hippocampus/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/parasitology , Neuroprotective Agents/chemistry , Oxygen Consumption/drug effects , Rats , Rats, Wistar
5.
Clin Endocrinol (Oxf) ; 94(1): 102-110, 2021 01.
Article in English | MEDLINE | ID: mdl-32895999

ABSTRACT

BACKGROUND: Poor sperm function is a major cause of infertility. There is no drug therapy to improve sperm function. Semen oxidative stress is a recently identified pathway for sperm damage. Commercial antioxidants such as L-carnitine and acetyl-L-carnitine (LAL) are commonly self-administered by infertile men. However, concerns have been raised whether inappropriate LAL therapy causes reductive stress-mediated sperm damage. It is imperative to investigate whether: (1) LAL improves sperm function by reducing reactive oxidative species (ROS); (2) LAL has differential effects on sperm function between men with normal and elevated ROS. METHODS: A prospective cohort study of routine clinical practice was performed in infertile men with abnormal sperm quality. Changes in sperm function and semen ROS levels following three months of oral LAL therapy were compared between participants with baseline seminal normal ROS (≤10RLU/SEC/106 sperm; n = 29) and High ROS (>10 RLU/SEC/106 sperm; n = 15) levels measured using an established colorimetric-luminol method. RESULTS: In normal ROS group, sperm function did not change following LAL therapy. In high ROS group, LAL therapy reduced semen ROS fivefold, increased sperm count by 50% (mean count in mill/ml: 21.5 + 7.2, baseline; 32.6 + 9.5, post-treatment, P = .0005), and total and progressive sperm motility each by 30% (mean total sperm motility in % 29.8 + 5.0, baseline: 39.4 + 6.2, post-treatment, P = .004; mean progressive sperm motility in % 23.1 + 4.6, baseline: 30.0 + 5.5, post-treatment, P = .014 vs. baseline). CONCLUSIONS: We report for the first time that LAL only improves sperm quality in infertile men who have baseline high-ROS levels prior to treatment. These data have important potential implications for couples with male infertility and their clinicians.


Subject(s)
Antioxidants , Infertility, Male , Antioxidants/metabolism , Humans , Infertility, Male/drug therapy , Male , Oxidative Stress , Prospective Studies , Reactive Oxygen Species/metabolism , Sperm Count , Sperm Motility , Spermatozoa
6.
Antioxidants (Basel) ; 9(9)2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32942589

ABSTRACT

Polycystic ovary syndrome (PCOS) is a complex metabolic disorder associated with female infertility. Based on energy and antioxidant regulatory functions of carnitines, we investigated whether acyl-L-carnitines improve PCOS phenotype in a mouse model induced by dehydroepiandrosterone (DHEA). CD1 mice received DHEA for 20 days along with two different carnitine formulations: one containing L-carnitine (LC) and acetyl-L-carnitine (ALC), and the other one containing also propionyl-L-carnitine (PLC). We evaluated estrous cyclicity, testosterone level, ovarian follicle health, ovulation rate and oocyte quality, collagen deposition, lipid droplets, and 17ß-HSD IV (17 beta-hydroxysteroid dehydrogenase type IV) expression. Moreover, we analyzed protein expression of SIRT1, SIRT3, SOD2 (superoxide dismutase 2), mitochondrial transcriptional factor A (mtTFA), RAGE (receptor for AGEs), GLO2 (glyoxalase 2) and ovarian accumulation of MG-AGEs (advanced glycation end-products formed by methylglyoxal). Both carnitine formulations ameliorated ovarian PCOS phenotype and positively modulated antioxidant molecular pathways in the ovarian microenvironment. Addition of PLC to LC-ALC formulation mitigated intraovarian MG-AGE accumulation and increased mtTFA expression. In conclusion, our study supports the hypothesis that oral administration of acyl-L-carnitines alleviates ovarian dysfunctions associated with this syndrome and that co-administration of PLC provides better activity. Molecular mechanisms underlying these effects include anti-oxidant/glycative activity and potentiation of mitochondria.

7.
Andrologia ; 52(3): e13523, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32017167

ABSTRACT

Spermatozoa are vulnerable to lack of energy and oxidative stress as a result of elevated levels of reactive oxygen species. Therefore, it is essential that appropriate nutrients are available during maturation. This randomised, double-blind, placebo-controlled trial investigated the effect of 6-month supplementation with carnitines and other micronutrients on sperm quality in 104 subjects with oligo- and/or astheno- and/or teratozoospermia with or without varicocele. Semen analyses were done at the beginning and end of the treatment. In addition to main analyses, post hoc analyses for age and body mass index (BMI) were carried out. Results were interpreted by dividing the population into two age and BMI classes. In 94 patients who completed the study, all sperm parameters increased in supplemented patients compared to the placebo group. A significant (p = .0272) difference in supplementation efficacy was observed for total motility on patients with varicocele and BMI < 25. In the same group, also the progressive motility was significantly superior (p = .0159). For Responder analysis, total motility results were confirmed in both the cited group (p = .0066) and in the varicocele group with BMI < 25 and age < 35 (p = .0078). This study suggests that supplementation is more effective in subjects with varicocele younger than 35 years with BMI < 25.


Subject(s)
Antioxidants/administration & dosage , Body Mass Index , Dietary Supplements , Infertility, Male/diet therapy , Micronutrients/administration & dosage , Varicocele/diet therapy , Adolescent , Adult , Age Factors , Double-Blind Method , Humans , Infertility, Male/etiology , Infertility, Male/pathology , Male , Middle Aged , Oxidative Stress/drug effects , Placebos/administration & dosage , Reactive Oxygen Species/metabolism , Sperm Count , Sperm Motility/drug effects , Sperm Motility/physiology , Treatment Outcome , Varicocele/complications , Varicocele/pathology , Young Adult
8.
Andrologia ; 51(6): e13267, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30873633

ABSTRACT

Carnitine is essential for energy metabolism and spermatozoa maturation. Combining L-carnitine and L-acetylcarnitine with micronutrients has been investigated as a treatment for infertility in men. We evaluated the effects of a therapeutic formulation, Proxeed Plus, on sperm parameters in oligoasthenozoospermic men. This prospective, randomised, double-blind, placebo-controlled clinical trial involved 175 males (19-44 years) with idiopathic oligoasthenozoospermia who failed to impregnate their partners (12 months). Males received Proxeed Plus or placebo for 3 and 6 months. Sperm volume, progressive motility and vitality significantly (p < 0.001) improved after 6 months compared to baseline. Sperm DNA fragmentation index significantly decreased compared to baseline (p < 0.001) and the 3-month therapy (p = 0.014) in treated men. Increased seminal carnitine and α-glucosidase concentration also positively correlated with improved progressive motility. Decreased DNA fragmentation index was the good predictor of progressive sperm motility >10%, and simultaneous measurement of changes in sperm vitality and DNA fragmentation index gave the highest probability of sperm motility 10% (AUC = 0.924; 95% CI = 0.852-0.996; p < 0.001). Logistic regression analyses revealed DNA fragmentation index decrease as the only independent predictor of sperm motility 10% (OR = 1.106; p = 0.034). We have demonstrated the beneficial effects of carnitine derivatives on progressive motility, vitality and sperm DNA fragmentation. Combining metabolic and micronutritive factors is beneficial for male infertility.


Subject(s)
Acetylcarnitine/administration & dosage , Carnitine/administration & dosage , Micronutrients/administration & dosage , Oligospermia/drug therapy , Spermatozoa/drug effects , Adult , DNA Fragmentation/drug effects , Double-Blind Method , Drug Combinations , Humans , Male , Placebos/administration & dosage , Prospective Studies , Sperm Count , Sperm Maturation/drug effects , Sperm Motility/drug effects , Treatment Outcome
9.
Mol Neurobiol ; 52(2): 826-36, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26041663

ABSTRACT

The carnitine palmitoyl transferase (CPT) system is a multiprotein complex with catalytic activity localized within a core represented by CPT1 and CPT2 in the outer and inner membrane of the mitochondria, respectively. Two proteins, the acyl-CoA synthase and a translocase also form part of this system. This system is crucial for the mitochondrial beta-oxidation of long-chain fatty acids. CPT1 has two well-known isoforms, CPT1a and CPT1b. CPT1a is the hepatic isoform and CPT1b is typically muscular; both are normally utilized by the organism for metabolic processes throughout the body. There is a strong evidence for their involvement in various disease states, e.g., metabolic syndrome, cardiovascular diseases, and in diabetes mellitus type 2. Recently, a new, third isoform of CPT was described, CPT1c. This is a neuronal isoform and is prevalently localized in brain regions such as hypothalamus, amygdala, and hippocampus. These brain regions play an important role in control of food intake and neuropsychiatric and neurological diseases. CPT activity has been implicated in several neurological and social diseases mainly related to the alteration of insulin equilibrium in the brain. These pathologies include Parkinson's disease, Alzheimer's disease, and schizophrenia. Evolution of both Parkinson's disease and Alzheimer's disease is in some way linked to brain insulin and related metabolic dysfunctions with putative links also with the diabetes type 2. Studies show that in the CNS, CPT1c affects ceramide levels, endocannabionoids, and oxidative processes and may play an important role in various brain functions such as learning.


Subject(s)
Carnitine O-Palmitoyltransferase/physiology , Mitochondria/enzymology , Neurodegenerative Diseases/enzymology , Animals , Brain/enzymology , Cardiovascular Diseases/enzymology , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/genetics , Ceramides/metabolism , Diabetes Mellitus, Type 2/enzymology , Disease Progression , Eating/physiology , Endocannabinoids/metabolism , Energy Metabolism/physiology , Fatty Acids/metabolism , Humans , Hypoglycemia , Insulin/metabolism , Learning/physiology , Lipid Metabolism, Inborn Errors , Malonyl Coenzyme A/metabolism , Metabolic Syndrome/enzymology , Mitochondria, Liver/enzymology , Mitochondria, Muscle/enzymology , Multienzyme Complexes/physiology , Oxidation-Reduction , Protein Isoforms
10.
Mech Ageing Dev ; 145: 39-50, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25660059

ABSTRACT

Previously we showed that in vivo treatment of elderly Fisher 344 rats with acetylcarnitine abolished the age-associated defect in respiratory chain complex III in interfibrillar mitochondria and improved the functional recovery of the ischemic/reperfused heart. Herein, we explored mitochondrial protein acetylation as a possible mechanism for acetylcarnitine's effect. In vivo treatment of elderly rats with acetylcarnitine restored cardiac acetylcarnitine content and increased mitochondrial protein lysine acetylation and increased the number of lysine-acetylated proteins in cardiac subsarcolemmal and interfibrillar mitochondria. Enzymes of the tricarboxylic acid cycle, mitochondrial ß-oxidation, and ATP synthase of the respiratory chain showed the greatest acetylation. Acetylation of isocitrate dehydrogenase, long-chain acyl-CoA dehydrogenase, complex V, and aspartate aminotransferase was accompanied by decreased catalytic activity. Several proteins were found to be acetylated only after treatment with acetylcarnitine, suggesting that exogenous acetylcarnitine served as the acetyl-donor. Two-dimensional fluorescence difference gel electrophoresis analysis revealed that acetylcarnitine treatment also induced changes in mitochondrial protein amount; a two-fold or greater increase/decrease in abundance was observed for thirty one proteins. Collectively, our data provide evidence for the first time that in the aged rat heart in vivo administration of acetylcarnitine provides acetyl groups for protein acetylation and affects the amount of mitochondrial proteins.


Subject(s)
Acetylcarnitine/metabolism , Aging/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Myocardium/metabolism , Acetylation , Animals , Rats , Rats, Inbred F344
11.
Mol Neurobiol ; 48(2): 353-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23813102

ABSTRACT

Diet in human health is no longer simple nutrition, but in light of recent research, especially nutrigenomics, it is linked via evolution and genetics to cell health status capable of modulating apoptosis, detoxification, and appropriate gene response. Nutritional deficiency and disease especially lack of vitamins and minerals is well known, but more recently, epidemiological studies suggest a role of fruits and vegetables, as well as essential fatty acids and even red wine (French paradox), in protection against disease. In the early 1990s, various research groups started considering the use of antioxidants (e.g., melatonin, resveratrol, green tea, lipoic acid) and metabolic compounds (e.g., nicotinamide, acetyl-L-carnitine, creatine, coenzyme Q10) as possible candidates in neuroprotection. They were of course considered on par with snake oil salesman (women) at the time. The positive actions of nutritional supplements, minerals, and plant extracts in disease prevention are now mainstream and commercial health claims being made are subject to regulation in most countries. Apart from efficacy and finding, the right dosages, the safety, and especially the level of purification and lack of contamination are all issues that are important as their use becomes widespread. From the mechanistic point of view, most of the time these substances replenish the body's deficiency and restore normal function. However, they also exert actions that are not sensu stricto nutritive and could be considered pharmacological especially that, at times, higher intake than recommended (RDA) is needed to see these effects. Free radicals and neuroinflammation processes underlie many neurodegenerative conditions, even Parkinson's disease and Alzheimer's disease. Curcumin, carotenoids, acetyl-L-carnitine, coenzyme Q10, vitamin D, and polyphenols and other nutraceuticals have the potential to target multiple pathways in these conditions. In summary, augmenting neuroprotective pathways using diet and finding new natural substances that can be more efficacious, i.e., induction of health-promoting genes and reduction of the expression of disease-promoting genes, could be incorporated into neuroprotective strategies of the future.


Subject(s)
Food , Nerve Degeneration/drug therapy , Neuroprotective Agents/therapeutic use , Nutrigenomics , Aging/drug effects , Aging/genetics , Animals , Humans , Nerve Degeneration/pathology , Neuroprotective Agents/pharmacology , Systems Biology
12.
J Cell Biochem ; 114(7): 1665-73, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23386399

ABSTRACT

Dietary agents are receiving much attention for the chemoprevention of cancer. While curcumin is known to influence several pathways and affect tumor growth in vivo, carnitin and its congeners play a variety of important metabolic functions: are involved in the oxydation of long-chain fatty acids, regulate acyl-CoA levels and influence protein activity and stability by modifying the extent of protein acetylation. In this study we evaluated the efficacy of carnitines in the prevention of cancer development using the 1,2,-dimethylhydrazine (DMH)-induced colon carcinogenesis model. We also assessed whether their combination was able to give rise to increased protection from cancer development. Mice treated with DMH were dosed orally with curcumin and/or carnitine and acylcarnitines for 20 weeks. At the end of the treatment colon samples were collected, and scored for multiple ACF and adenomas. We observed that carnitine and acyl-carnitines had same, if not higher, efficacy than curcumin alone in inhibiting the formation of neoplastic lesions induced by DMH treatment. Interestingly, the combination of curcumin and acetyl-L-carnitine was able to fully inhibit the development of advanced adenoma lesions. Our data unveil the antitumor effects of carnitines and warrant additional studies to further support the adoption of carnitines as cancer chemopreventative agents.


Subject(s)
1,2-Dimethylhydrazine/toxicity , Carnitine/therapeutic use , Colonic Neoplasms/drug therapy , Acetylcarnitine/therapeutic use , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/chemistry , Colonic Neoplasms/metabolism , Curcumin/therapeutic use , HT29 Cells , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C
13.
Neurotoxicol Teratol ; 33(2): 288-96, 2011.
Article in English | MEDLINE | ID: mdl-21216281

ABSTRACT

Amyloid-beta peptide (Aß) deposition is assumed to play a pathogenic role in the brain of Alzheimer's disease patients. To date, the precise mechanisms underlying Aß toxicity are not fully understood. A recent hypothesis suggesting that the Receptor-for-Advanced-Glycation-End-Products (RAGE)-a trans-membrane protein signaling for oxidative stress-is involved in Aß toxicity is gaining attention. Early Aß toxicity could indeed help to explain the deleterious events further produced by this molecule in the brain. In this work, we evaluated the pattern of early expression of RAGE in the toxic model induced by Aß25₋35 in rat CA1 region. Intrahippocampal injections of Aß25₋35 in rats increased the RAGE expression at 24 h post-injection; this event was accompanied by increased components of RAGE downstream signaling in hippocampal cells, such as enhanced expression of the pro-apoptotic factor NF-κB, increased nitric oxide production, LDH leakage, mitochondrial dysfunction, increased TNF-α expression, antioxidant genes down-regulation, and augmented neurodegeneration. Our findings support an active role of RAGE during the early stages of Aß25₋35 toxicity in the hippocampus.


Subject(s)
Amyloid beta-Peptides/toxicity , CA1 Region, Hippocampal/drug effects , Gene Expression/drug effects , Peptide Fragments/toxicity , Receptors, Immunologic/genetics , Animals , Antioxidants/metabolism , Blotting, Western , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Cell Death/drug effects , Male , Microinjections , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/physiology , Nitric Oxide/biosynthesis , Oxidative Stress/drug effects , Oxidative Stress/genetics , Rats , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products , Reverse Transcriptase Polymerase Chain Reaction
14.
Ann N Y Acad Sci ; 1199: 52-68, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20633109

ABSTRACT

Encephalopathy is evidenced as an altered mental state with various neurological symptoms, such as memory and cognitive problems. The type of a substance-evoked encephalopathy will depend on the drug, substance, or combination being abused. The categories into which we could place the various abused substances could be tentatively divided into stimulants, amphetamines, hallucinogens, narcotics, inhalants, anesthetics, anabolic steroids, and antipsychotics/antidepressants. Other factors that may underlie encephalopathy, such as infectious agents, environmental, and other factors have also to be taken into account. Drugs of abuse can be highly toxic to the CNS following acute, but more so in chronic exposure, and can produce significant damage to other organs, such as the heart, lungs, liver, and kidneys. The damage to these organs may be at least partially reversible when drug abuse is stopped but CNS damage from repeated or prolonged abuse is often irreversible. The major pathways for the organ and CNS toxicity could be related to ischemic events as well as increased cell damage due to metabolic or mitochondrial dysfunction resulting in increased excitotoxicity, reduced energy production, and lowered antioxidant potential. These susceptibilities could be strengthened by the use of antioxidants to combat free radicals (e.g., vitamin E, lipoic acid); trying to improve energy generation by using mitochondriotropic/metabolic compounds (e.g., thiamine, coenzyme Q10, carnitine, riboflavin); by reducing excitotoxicity (e.g., glutamate antagonists) and other possible strategies, such as robust gene response, need to be investigated further. The drug-abuse-evoked encephalopathy still needs to be studied further to enable better preventative and protective strategies.


Subject(s)
Brain Diseases/etiology , Neuroprotective Agents/therapeutic use , Substance-Related Disorders/complications , Brain Diseases/prevention & control , Humans
15.
Neurosci Lett ; 480(2): 117-21, 2010 Aug 16.
Article in English | MEDLINE | ID: mdl-20542088

ABSTRACT

Oxidative stress and secondary excitotoxicity, due to cellular energy deficit, are major factors playing roles in 3-nitropropionic acid (3-NPA) induced mitochondrial dysfunction. Acute or chronic exposure to 3-NPA also leads to neuronal degeneration in different brain regions. The present study quantitatively assessed peripheral neuropathy induced by chronic exposure to 3-NPA in rats. The neuroprotective abilities of two antioxidants, acetyl-l-carnitine and resveratrol, were investigated as well. Rats were exposed for up to four weeks to 3-NPA alone or 3-NPA combined with acetyl-l-carnitine or resveratrol, administered peripherally. The experimental outcome was evaluated by neurophysiological, histological, and morphometric analyses. Rats exposed to 3-NPA developed hind limb paresis. Furthermore, a significant decrease in motor nerve conduction velocity (MCV) was detected in tail nerves and axonal degeneration in sciatic nerves (p<0.05). Treatment with resveratrol prevented the functional effects of 3-NPA exposure, whereas treatment with acetyl-l-carnitine, preventing paresis, was not effective to MCV and morphological changes. These data suggest that resveratrol is a good candidate for treatment of metabolic neuropathy. The experimental outcome of this study shows that chronic treatment with 3-NPA in rats is relevant in development of an experimental model of toxic neuropathy.


Subject(s)
Acetylcarnitine/pharmacology , Antioxidants/pharmacology , Environmental Pollutants/toxicity , Neuroprotective Agents/pharmacology , Nitro Compounds/toxicity , Peripheral Nervous System Diseases/drug therapy , Propionates/toxicity , Stilbenes/pharmacology , Acetylcarnitine/therapeutic use , Animals , Antioxidants/therapeutic use , Axons/drug effects , Axons/pathology , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neural Conduction/drug effects , Neuroprotective Agents/therapeutic use , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/pathology , Rats , Rats, Sprague-Dawley , Resveratrol , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Stilbenes/therapeutic use
16.
Ann Vasc Surg ; 22(4): 552-8, 2008.
Article in English | MEDLINE | ID: mdl-18502605

ABSTRACT

Peripheral arterial obstructive disease (PAOD) of the lower limbs affects 5% of the adult population. Uncontrolled arteriopathy is established due to a microcirculatory deficit, which may be present despite a good Winsor index and which leads to exhaustion of the functional microcirculatory reserve. The target of this study was to examine possible improvements in microvascular and tissue homeostasis by the administration of propionyl-L-carnitine (PLC). A total of 26 patients were enrolled in this study, aged 65 +/- 15 years; two males were diagnosed at stage IIA and 17 males and seven females at stage IIB PAOD. The main criterion of inclusion was the worsening of walking distance during the last month. In this study the duration of therapy was 33 days. PLC was administered in three flasks, each containing 300 mg in 250 cc saline by continuous infusion. The following parameters were measured before and after treatment: pain-free and maximum walking distance (measured on a treadmill at 3.2 km/hr with a gradient of 12%), recovery time from pain after maximum walking distance, ankle-brachial index by means of the Doppler apparatus, and evaluation of the microcirculation using capillaroscopy. The results showed that therapy with PLC was effective at restoring activity of skeletal muscle in ischemic conditions. In particular, capillaroscopy showed improvement in the angioarchitecture in the microcirculation fields, expressed as increased numbers of visible capillaries and diminution in the time of loss of sodium fluorescein marker. The clinical data showed increased walking distance and diminished time to recover from pain, and the clinical improvement correlated with improved microcirculatory function. From these preliminary data has emerged an indication of therapy with PLC for chronic obstructive arteriopathy of the lower limbs at stage II. Further studies with higher numbers of patients and more controlled variables are planned.


Subject(s)
Arterial Occlusive Diseases/drug therapy , Carnitine/analogs & derivatives , Leg/blood supply , Muscle, Skeletal/blood supply , Peripheral Vascular Diseases/drug therapy , Aged , Arterial Occlusive Diseases/physiopathology , Blood Flow Velocity , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/therapeutic use , Carnitine/administration & dosage , Carnitine/therapeutic use , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Male , Microcirculation , Microscopic Angioscopy , Peripheral Vascular Diseases/physiopathology , Walking
17.
Ann N Y Acad Sci ; 1122: 50-68, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18077564

ABSTRACT

Drug abuse is associated with significant health risk. Whether drug abusers are at a higher risk of suffering the metabolic syndrome is not widely known. The metabolic syndrome is a cluster of metabolic abnormalities, including hyperinsulinemia, hypertension, dyslipidemia, and abdominal obesity, and is probably triggered by initial imbalances at the cellular level in various critical metabolic pathways. These initially small metabolic imbalances are believed to cascade with time and lead to larger problems. Some indications that drug abuse may increase the risk of the metabolic syndrome include the following: Drug-abusing patients have higher rates of diabetes complications. Substance abuse is a significant contributing factor for treatment noncompliance in diabetes. Nutrition education can enhance substance abuse treatment outcomes. Each type of drug/substance abuse has a unique profile of toxicity. For example, the amphetamines generally affect the cardiovascular and neurological systems, worsening the risk factors for the metabolic syndrome. Methamphetamine (meth) abusers suffer cognitive deficits and abnormal metabolic activity, which affect nutritional status. This condition is further worsened by a drastic reduction in oral health in meth abusers, resulting in improper chewing and, therefore, digestion. Nutritional deficiency in combination with drug abuse would increase the risk of developing the metabolic syndrome by increasing cell damage, augmenting excitotoxicity, reducing energy production, and lowering the antioxidant potential of the cells. Another potential risk factor in the development of the metabolic syndrome is genetic vulnerability, especially in combination with drug abuse and nutritional deficiencies. The strategies available to treat this problem include pharmacological agents as well as dietary antioxidants. Such measures may be useful in reducing drug abuse-related toxicity that may lead to the metabolic syndrome.


Subject(s)
Metabolic Diseases/etiology , Substance-Related Disorders/complications , Animals , Humans , Metabolic Diseases/therapy , Risk Factors
18.
Ann N Y Acad Sci ; 1122: 260-75, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18077579

ABSTRACT

This study deals with the possible inhibitory role played by acetyl-l-carnitine (ALC) against methamphetamine (METH)-induced behavioral sensitization. Because valproate (VAL) inhibits the behavioral sensitization exerted by different psychostimulants, we investigated ALC's potential to prevent the amplification of METH-mediated psychomotor effects. We therefore evaluated the locomotor effects of VAL or ALC alone or in combination with METH after acute (day 1) as well as repeated (day 7) drug challenge. Finally, to assess the induction of behavioral sensitization, we also recorded the METH-mediated locomotor response after 7 days of drug suspension (day 15). Results showed that both VAL and ALC prevented the METH-induced sensitization. Another interesting observation was the significantly higher METH-induced hyperactivity at day 15 (after a 7-day drug-free period), indicating that behavioral sensitization developed during the washout period. Results also showed that both the acute and repeated coadministration of METH with either VAL or ALC inhibited METH-induced hyperactivity. We present different hypotheses concerning similar but also peculiar mechanisms that might underlie the preventive action of VAL and ALC. These data add to a growing body of literature that illustrates the potential of ALC in protecting against the insult of dysfunctional mitochondrial metabolism and psychostimulant-mediated neurotoxicity. By demonstrating an in vivo action against one of the most abused drugs, these results raise the possibility of beneficial effects of ALC in abuse behavior.


Subject(s)
Acetylcarnitine/therapeutic use , Methamphetamine/adverse effects , Neuroprotective Agents/therapeutic use , Psychomotor Disorders/chemically induced , Psychomotor Disorders/prevention & control , Valproic Acid/therapeutic use , Analysis of Variance , Animals , Behavior, Animal/drug effects , Drug Administration Schedule , Drug Interactions , Male , Mice , Motor Activity/drug effects , Time Factors
19.
Ann N Y Acad Sci ; 1074: 74-83, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17105904

ABSTRACT

Adult, male Sprague-Dawley rats were injected with 3-ni-tropropionic acid (3-NPA) at 30 mg/kg or methamphetamine (METH) at 20 mg/kg alone or following pretreatment with L-cartnitine (LC) at 100 mg/kg. Rectal temperature was measured before and 4 h following treatment. Animals were sacrificed at 4 h posttreatment. Monoamine neurotransmitters, dopamine (DA) and serotonin (5-HT), and their metabolites were analyzed in the striatum using high-performance liquid chromatography method coupled with electrochemical detection (HPLC/ED). Transcripts of several genes related to DA metabolism were quantified using real time reverse transciption polymerase chain reaction (RT-PCR). Core temperature decreased significantly after 3-NPA acid and increased in METH-treated rats (P < 0.05). Temperature change at 4 h exhibited a significant LC effect for 3-NPA, preventing hypothermia (P < 0.05) and no effect for METH. Concentration of DA and 5-HT, and their metabolites, 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), and 5-hydroxyindoleacetic acid (5-HIAA), increased significantly in 3-NPA and decreased in METH-treated rats. An increase in DOPAC/DA turnover and serotonin observed after 3-NPA was abolished in LC-/3-NPA-treated rats. In both 3-NPA- and METH-treated rats, LC prevented an increase in DA receptor D(1) gene expression. It appears that carnitine effect preventing hypothermia after 3-NPA treatments may be related not only to its mitochondriotropic actions but also to inhibitory effect on the DA and 5-HT systems activated after the exposure to 3-NPA. The same effect observed at the transcriptional level, at least for the DA receptor D(1), may account for protection against METH toxicity.


Subject(s)
Antihypertensive Agents/pharmacology , Carnitine/pharmacology , Dopamine Agents/pharmacology , Methamphetamine/pharmacology , Neurotoxicity Syndromes/therapy , Nitro Compounds/pharmacology , Propionates/pharmacology , Vitamin B Complex/pharmacology , Animals , Male , Rats , Rats, Sprague-Dawley
20.
Ann N Y Acad Sci ; 1074: 303-14, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17105926

ABSTRACT

Nutritional deficiency in combination with drug abuse may increase risk of developing the metabolic syndrome by augmenting cell damage, excitotoxicity, reducing energy production, and lowering the antioxidant potential of the cells. We have reviewed here the following points: effects of drugs of abuse on nutrition and brain metabolism; effects of nutrition on actions of the drugs of abuse; drug abuse and probability of developing metabolic syndrome; role of genetic vulnerability in nutrition/drug abuse and brain damage; and the role of neuroprotective supplements in drug abuse. Nutrition education is an essential component of substance abuse treatment programs and can enhance substance abuse treatment outcomes. The strategies available, in particular the nutritional approach to protect the drug abusers from the metabolic syndrome and other diseases are discussed.


Subject(s)
Brain/metabolism , Metabolic Syndrome/etiology , Neuroprotective Agents/pharmacology , Nutritional Status/drug effects , Substance-Related Disorders/complications , Animals , Antioxidants/pharmacology , Dietary Supplements , Humans , Mice , Rats , Selenium/pharmacology , Substance-Related Disorders/etiology , Substance-Related Disorders/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...