Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
1.
Pharmacol Biochem Behav ; 235: 173702, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38154590

ABSTRACT

Smoking is the leading cause of preventable death worldwide, with <7 % of smoking cessation attempts being met with success. Nicotine, the main addictive agent in cigarettes, enhances the reinforcing value of other environmental rewards. Under some circumstances, this reward enhancement maintains nicotine consumption. Varenicline (i.e., cessation aid Chantix™) also has reward-enhancement effects via nicotinic acetylcholine receptor agonism (nAChRs) - albeit less robust than nicotine. Cotinine is the major metabolite of nicotine. Recent studies suggest that cotinine is a positive allosteric modulator (PAM) and/or a weak agonist at nAChRs. Thus, cotinine may enhance the behavioral effects of nAChR compounds such as varenicline and/or exert some behavioral effects alone. We used 20 (10M, 10F) Sprague-Dawley rats to assess reward-enhancement within-subjects by examining responding maintained by a reinforcing visual stimulus on a Variable Ratio 2 schedule of reinforcement. To assess the reward-enhancing effects of cotinine, rats received one injection of cotinine (saline, 0.1, 0.3, 1.0, 3.0, 6.0 mg/kg) before each 1 h session. To assess cotinine and varenicline interactions, rats received an injection of cotinine (saline, 0.1, 1.0, or 6.0 mg/kg) and of varenicline (saline, 0.1, 0.3, 1.0, or 3.0 mg/kg) before the session. While we replicated prior work identifying reward-enhancement by 0.1, 0.3, and 1.0 mg/kg varenicline, cotinine alone did not produce reward-enhancement nor augment the reward-enhancing effects of varenicline. Future studies may consider examining the reward-enhancing effects of cotinine with other reinforcers or co-administered with other smoking cessation aids such as bupropion.


Subject(s)
Nicotine , Receptors, Nicotinic , Humans , Rats , Animals , Varenicline/pharmacology , Nicotine/pharmacology , Cotinine/pharmacology , Rats, Sprague-Dawley , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Benzazepines/pharmacology , Quinoxalines/pharmacology
2.
Neuropharmacology ; 230: 109495, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36914092

ABSTRACT

Previous studies indicated that cotinine, the major metabolite of nicotine, supported intravenous self-administration and exhibited relapse-like drug-seeking behaviors in rats. Subsequent studies started to reveal an important role of the mesolimbic dopamine system in cotinine's effects. Passive administration of cotinine elevated extracellular dopamine levels in the nucleus accumbens (NAC) and the D1 receptor antagonist SCH23390 attenuated cotinine self-administration. The objective of the current study was to further investigate the role of mesolimbic dopamine system in mediating cotinine's effects in male rats. Conventional microdialysis was conducted to examine NAC dopamine changes during active self-administration. Quantitative microdialysis and Western blot were used to determine cotinine-induced neuroadaptations within the NAC. Behavioral pharmacology was performed to investigate potential involvement of D2-like receptors in cotinine self-administration and relapse-like behaviors. NAC extracellular dopamine levels increased during active self-administration of cotinine and nicotine with less robust increase during cotinine self-administration. Repeated subcutaneous injections of cotinine reduced basal extracellular dopamine concentrations without altering dopamine reuptake in the NAC. Chronic self-administration of cotinine led to reduced protein expression of D2 receptors within the core but not shell subregion of the NAC, but did not change either D1 receptors or tyrosine hydroxylase in either subregion. On the other hand, chronic nicotine self-administration had no significant effect on any of these proteins. Systemic administration of eticlopride, a D2-like receptor antagonist attenuated both cotinine self-administration and cue-induced reinstatement of cotinine seeking. These results further support the hypothesis that the mesolimbic dopamine transmission plays a critical role in mediating reinforcing effects of cotinine.


Subject(s)
Dopamine Antagonists , Dopamine , Rats , Male , Animals , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Cotinine/pharmacology , Nicotine/pharmacology , Nicotine/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D1/metabolism , Nucleus Accumbens , Self Administration
3.
Compr Psychiatry ; 123: 152381, 2023 05.
Article in English | MEDLINE | ID: mdl-36905856

ABSTRACT

BACKGROUND: Of interest to women's mental health, a wealth of studies suggests sex differences in nicotine addiction and treatment response, but their psychoneuroendocrine underpinnings remain largely unknown. A pathway involving sex steroids could indeed be involved in the behavioural effects of nicotine, as it was found to inhibit aromatase in vitro and in vivo in rodents and non-human primates, respectively. Aromatase regulates the synthesis of oestrogens and, of relevance to addiction, is highly expressed in the limbic brain. METHODS: The present study sought to investigate in vivo aromatase availability in relation to exposure to nicotine in healthy women. Structural magnetic resonance imaging and two [11C]cetrozole positron emission tomography (PET) scans were performed to assess the availability of aromatase before and after administration of nicotine. Gonadal hormones and cotinine levels were measured. Given the region-specific expression of aromatase, a ROI-based approach was employed to assess changes in [11C]cetrozole non-displaceable binding potential. RESULTS: The highest availability of aromatase was found in the right and left thalamus. Upon nicotine exposure, [11C]cetrozole binding in the thalamus was acutely decreased bilaterally (Cohen's d = -0.99). In line, cotinine levels were negatively associated with aromatase availability in the thalamus, although as non-significant trend. CONCLUSIONS: These findings indicate acute blocking of aromatase availability by nicotine in the thalamic area. This suggests a new putative mechanism mediating the effects of nicotine on human behaviour, particularly relevant to sex differences in nicotine addiction.


Subject(s)
Nicotine , Tobacco Use Disorder , Animals , Humans , Female , Male , Nicotine/adverse effects , Nicotine/metabolism , Aromatase/metabolism , Aromatase/pharmacology , Cotinine/metabolism , Cotinine/pharmacology , Brain/diagnostic imaging , Positron-Emission Tomography
4.
Behav Pharmacol ; 33(7): 482-491, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36148836

ABSTRACT

Relapse is a defining feature of smoking and a significant challenge in cessation management. Elucidation of novel factors underlying relapse may inform future treatments. Cotinine, the major metabolite of nicotine, has been shown to support intravenous self-administration in rats, implicating it as one potential factor contributing to nicotine reinforcement. However, it remains unknown whether cotinine would induce relapse-like behaviors. The current study investigated relapse to cotinine seeking in two relapse models, the reinstatement of drug seeking and incubation of drug craving models. In the reinstatement model, rats were trained to self-administer cotinine, underwent extinction of cotinine-associated responses, and were tested for cue-, drug-, or stress-induced reinstatement. Conditioned cues associated with cotinine self-administration, cotinine (1-2 mg/kg), or the pharmacological stressor yohimbine (1.25-2.5 mg/kg) induced reinstatement of cotinine seeking. Female rats displayed more pronounced cue-induced, but not drug- or stress-induced reinstatement than male rats. In the incubation of the craving model, rats were trained to self-administer cotinine and underwent forced withdrawal in home cages. Rats were tested for cue-induced cotinine-seeking on both withdrawal day 1 and withdrawal day 18. Rats exhibited greater cue-induced cotinine-seeking on withdrawal day 18 compared to withdrawal day 1, with no difference between male and female rats. These findings indicate that cotinine induces sex-specific relapse to drug seeking in rats, suggesting that cotinine may contribute to relapse.


Subject(s)
Cotinine , Nicotine , Animals , Conditioning, Operant , Cotinine/pharmacology , Cues , Extinction, Psychological , Female , Male , Nicotine/pharmacology , Rats , Rats, Sprague-Dawley , Recurrence , Self Administration , Yohimbine/pharmacology
5.
Biol Reprod ; 107(2): 635-649, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35191979

ABSTRACT

Levels of cotinine, a major metabolite of nicotine, have been positively correlated with risks of cigarette smoking-related diseases. Melatonin is synthesized by the pineal gland and has been demonstrated to be beneficial to oocyte maturation due to its antioxidative activity. In this study, we investigated the effects of cotinine on mouse oocyte meiosis and the protective roles of melatonin in vitro and in vivo. The results showed that cotinine exposure caused defects in the first polar body extrusion and reduced parthenogenetic activation in in vitro-matured oocytes. Additionally, cotinine exposure increased the level of oxidative stress, which resulted in aberrant actin distribution, abnormal spindle morphology, chromosome misalignment, and even oocyte aneuploidy. Simultaneously, cotinine exposure decreased the mitochondrial membrane potential and antioxidant gene expression and increased apoptosis-related gene expression. However, all these toxic effects of cotinine could be reversed after the addition of melatonin, and the mechanism may be a decrease in reactive oxygen species production. In conclusion, cotinine causes poor oocyte quality, which could be rescued by melatonin supplementation during meiotic maturation in mouse oocytes.


Subject(s)
Melatonin , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Cotinine/metabolism , Cotinine/pharmacology , Meiosis , Melatonin/metabolism , Melatonin/pharmacology , Mice , Oocytes/metabolism , Oogenesis , Oxidative Stress , Reactive Oxygen Species/metabolism
6.
Pharmacol Biochem Behav ; 207: 173223, 2021 08.
Article in English | MEDLINE | ID: mdl-34197843

ABSTRACT

Zebrafish provide a valuable emerging complementary model for neurobehavioral research. They offer a powerful way to screen for the potential therapeutic effects of neuroactive drugs. A variety of behavioral tests for zebrafish have been developed and validated for assessing neurobehavioral function. The novel tank diving test is a straightforward, reproducible way of measuring anxiety-like behavior in zebrafish. When introduced into a novel tank, zebrafish normally dive to the bottom of the tank and then gradually explore the higher levels of the water column as time progresses. Buspirone is an effective anxiolytic drug in humans, which has been found, with acute administration, to reduce this anxiety-like response in zebrafish. The current study used the zebrafish model to evaluate the potential anxiolytic effects of alkaloids, commonly found in Solanaceae plants, with known neuropharmacology relevant to mood regulation. In line with previous findings, acute treatment with anxiolytic positive controls buspirone and the plant alkaloid nicotine reduced the anxiety-like diving response in the zebrafish novel tank diving test. Further, both buspirone and nicotine continued to produce anxiolytic-like effects in zebrafish after 5 days of exposure. In the same treatment paradigm, the effects of five other alkaloids-cotinine, anatabine, anabasine, harmane, and norharmane-were investigated. Cotinine, the major metabolite of nicotine, also caused anxiolytic-like effects, albeit at a dose higher than the effective dose of nicotine. Nicotine's anxiolytic-like effect was not shared by the other nicotinic alkaloids, anabasine and anatabine, or by the naturally present monoamine oxidase inhibitors harmane and norharmane. We conclude that nicotine uniquely induces anxiolytic-like effects after acute and subchronic treatment in zebrafish. The zebrafish model with the novel tank diving test could be a useful complement to rodent models for screening candidate compounds for anxiolytic effects in nonclinical studies.


Subject(s)
Alkaloids/pharmacology , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Solanaceae/chemistry , Anabasine/pharmacology , Animals , Anxiety/metabolism , Behavior, Animal/drug effects , Buspirone/pharmacology , Carbolines/pharmacology , Cotinine/pharmacology , Disease Models, Animal , Female , Harmine/analogs & derivatives , Harmine/pharmacology , Humans , Male , Nicotine/pharmacology , Pyridines/pharmacology , Zebrafish
7.
J Pharmacol Exp Ther ; 376(3): 338-347, 2021 03.
Article in English | MEDLINE | ID: mdl-33361363

ABSTRACT

Nicotine is the major addictive component in tobacco. Cotinine is the major metabolite of nicotine and a weak agonist for nicotinic acetylcholine receptors (nAChRs). Nicotine supports self-administration in rodents. However, it remains undetermined whether cotinine can be self-administered. This study aimed to characterize cotinine self-administration in rats, to compare effects of cotinine to those of nicotine, and to determine potential involvement of nAChRs in cotinine's effects. Adult Wistar rats were trained to self-administer cotinine or nicotine (0.0075, 0.015, 0.03, or 0.06 mg/kg per infusion) under fixed-ratio (FR) and progressive-ratio (PR) schedules. Blood nicotine and cotinine levels were determined after the last FR session. Effects of mecamylamine, a nonselective nAChR antagonist, and varenicline, a partial agonist for α4ß2* nAChRs, on cotinine and nicotine self-administration were determined. Rats readily acquired cotinine self-administration, responded more on active lever, and increased motivation to self-administer cotinine when the reinforcement requirement increased. Blood cotinine levels ranged from 77 to 792 ng/ml. Nicotine induced more infusions at lower doses during FR schedules and greater breakpoints at higher doses during the PR schedule than cotinine. There was no difference in cotinine self-administration between male and female rats. Mecamylamine and varenicline attenuated nicotine but not cotinine self-administration. These results indicate that cotinine was self-administered by rats. These effects of cotinine were less robust than nicotine and exhibited no sex difference. nAChRs appeared to be differentially involved in self-administration of nicotine and cotinine. These results suggest cotinine may play a role in the development of nicotine use and misuse. SIGNIFICANCE STATEMENT: Nicotine addiction is a serious public health problem. Cotinine is the major metabolite of nicotine, but its involvement in nicotine reinforcement remains elusive. Our findings indicate that cotinine, at doses producing clinically relevant blood cotinine levels, supported intravenous self-administration in rats. Cotinine self-administration was less robust than nicotine. Mecamylamine and varenicline attenuated nicotine but not cotinine self-administration. These results suggest cotinine may play a role in the development of nicotine use and misuse.


Subject(s)
Cotinine/administration & dosage , Cotinine/pharmacology , Nicotine/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Interactions , Female , Male , Mecamylamine/pharmacology , Nicotine/administration & dosage , Rats , Rats, Wistar , Receptors, Nicotinic/metabolism , Self Administration , Varenicline/pharmacology
8.
Int J Dev Biol ; 64(7-8-9): 445-452, 2020.
Article in English | MEDLINE | ID: mdl-32930354

ABSTRACT

Planarians are traditional model invertebrates in regeneration and developmental biology research that also display a variety of quantifiable behaviors useful to screen for pharmacologically active compounds. One such behavior is the expression of seizure-like movements (pSLMs) induced by a variety of substances. Previous work from our laboratory showed that cocaine, but not nicotine, induced pSLMs in intact but not decapitated planarians. Interestingly, as decapitated planarians regenerated their heads, they gradually recovered their sensitivity to cocaine. These results suggested a method to assess planarian brain regeneration and a possible way of identifying compounds that could enhance or hold back brain regeneration. In the present work, we demonstrate that the cholinergic agent cytisine is a suitable reference compound to apply our method. Cytisine induces pSLMs in a concentration-dependent manner in intact (but not decapitated) planarians of the species Girardia tigrina. Based on our data, we developed a behavioral protocol to assess planarian brain regeneration over time. We tested this method to measure the effect of ethanol on G. tigrina's brain regeneration. We found that ethanol slows down the rate of planarian brain regeneration in a concentration-dependent manner, consistently with data from other research groups that tested ethanol effects on planarian brain regeneration using different behavioral protocols. Thus, here we establish a general method using cytisine-induced pSLMs as an indicator of brain regeneration in planarians, a method that shows potential for assessing the effect of pharmacologically active compounds in this process.


Subject(s)
Alkaloids/pharmacology , Brain/drug effects , Cholinergic Agents/pharmacology , Planarians/physiology , Regeneration/drug effects , Anesthetics, Local/pharmacology , Animals , Azocines/pharmacology , Brain/physiology , Carbachol/pharmacology , Cocaine/pharmacology , Cotinine/pharmacology , Dose-Response Relationship, Drug , Ethanol/pharmacology , Nicotine/pharmacology , Quinolizines/pharmacology , Regeneration/physiology , Seizures/physiopathology
9.
Brain Res Bull ; 164: 65-74, 2020 11.
Article in English | MEDLINE | ID: mdl-32818583

ABSTRACT

This study aimed to assess the effects of cotinine on age-induced memory and learning impairment and related downstream pathways in mice. Thirty aged (18-month old) and 10 young mice (8-week old) were randomly divided into 4 groups (n = 10 each) and subjected to cotinine at 5 mg/kg dose and/or methyllycaconitine (MLA) at 1 mg/kg, i.p. dose (α7 nAChRs antagonist) for 4 weeks. Morris water maze (MWM) and novel object recognition (NOR) tasks were used to assess spatial and recognition learning and memories of the mice, respectively. Levels of oxidative stress, apoptosis, neuroinflammation, and structural synaptic plasticity, and also neurotrophic factors and α7 nAChRs were assessed in the hippocampus using either ELISA or Western blotting. Aging was associated with learning and memory disabilities and dysregulation of the assessed pathways in the hippocampus of mice. Chronic cotinine treatment improved learning and memory in aged animals, indicated by decreased latency time, and increased time spent in the target quadrant and discrimination index (DI) in the MWM and NOR tasks. Also, chronic cotinine injection increased total antioxidant capacity (TAC), SOD and GSH-px activity, PSD-95, GAP-43, SYN, brain-derived neurotrophic factor, and neural growth factor levels and decreased malondialdehyde, TNF-α, and IL-1ß in the hippocampus of aged mice. Conversely, MLA treatment reversed most of the mentioned effects via the blockade of α7 nAChRs. Cotinine improves age-induced memory and learning impairment via its modulatory effects on α7 nAChRs and subsequent activation/deactivation of the mentioned pathways in the hippocampus of aged mice.


Subject(s)
Aging/metabolism , Cognitive Dysfunction/drug therapy , Cotinine/pharmacology , Maze Learning/drug effects , Memory/drug effects , Animals , Apoptosis/drug effects , Cotinine/therapeutic use , Hippocampus/drug effects , Hippocampus/metabolism , Male , Malondialdehyde/metabolism , Mice , Oxidative Stress/drug effects , Receptors, Nicotinic/metabolism , Superoxide Dismutase/metabolism
10.
Biosci Rep ; 40(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32441737

ABSTRACT

BACKGROUND: The present study was designed to explore the regulatory mechanisms and influences of cotinine on deep vein thrombosis (DVT) in rats via the toll-like receptor 4/nuclear factor κ binding (TLR-4/NF-κB) pathway. METHODS: In this experimental study, 30 SD rats were randomly assigned to control group, sham operation group, model group, cotinine (10 µg/kg) group, and model + cotinine (10 µg/kg) group. The thromboxane B2 (TXB2), 6-keto-PGF1α, plasminogen activator inhibitor (PAI), tissue plasminogen activator (t-PA), TLR4, NF-κB, and p65 mRNA and protein expression and tissue changes were analyzed by ELISA, Hematoxylin-Eosin (HE) staining, RT-PCR, and Western blot. RESULTS: There was no significant difference between the control and sham operation groups (P>0.05). The model and cotinine groups showed significantly higher mRNA and protein levels of TXB2, interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), PAI, TLR-4, and NF-κB, and significantly lower levels of 6-keto-PGF1α and t-PA than the control and sham operation groups (P<0.05), and the model + cotinine group showed significantly higher mRNA and protein levels of TXB2, IL-6 and TNF-α, PAI, TLR-4, and NF-κB and significantly lower levels of 6-keto-PGF1α and t-PA than the model group (P<0.05). CONCLUSION: Cotinine can aggravate thrombus and inflammation in rats with DVT, and the mechanism may be associated with the activation of the TLR-4/NF-κB inflammatory signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cotinine/pharmacology , Fibrinolytic Agents/pharmacology , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Venous Thrombosis/drug therapy , 6-Ketoprostaglandin F1 alpha/blood , Animals , Disease Models, Animal , Male , NF-kappa B/genetics , Rats, Sprague-Dawley , Signal Transduction , Thromboxane B2/blood , Toll-Like Receptor 4/genetics , Venous Thrombosis/genetics , Venous Thrombosis/metabolism
11.
Arch Toxicol ; 93(11): 3141-3152, 2019 11.
Article in English | MEDLINE | ID: mdl-31515601

ABSTRACT

The chemical warfare agent sulfur mustard (SM) alkylates a multitude of biomacromolecules including DNA and proteins. Cysteine residues and nucleophilic nitrogen atoms in purine DNA bases are typical targets of SM but potentially every nucleophilic structure may be alkylated by SM. In the present study, we analyzed potential SM-induced alkylation of glucocorticoid (GC) hormones and functional consequences thereof. Hydrocortisone (HC), the synthetic betamethasone (BM) and dexamethasone (DEX) were chosen as representative GCs. Structural modifications were assessed by matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry and nuclear magnetic resonance (NMR) spectroscopy. The hypothesized alkylation was verified and structurally allocated to the OH-group of the C21 atom. The biological function of SM-alkylated GCs was investigated using GC-regulated dual-luciferase reporter gene assays and an ex vivo GC responsiveness assay coupled with real-time quantitative polymerase chain reaction (RT-qPCR). For the reporter gene assays, HEK293-cells were transiently transfected with a dual-luciferase reporter gene that is transcriptional regulated by a GC-response element. These cells were then incubated either with untreated or SM-derivatized HC, BM or DEX. Firefly-luciferase (Fluc) activity was determined 24 h after stimulation. Fluc-activity significantly decreased after stimulation with SM-pre-exposed GC dependent on the SM concentration. The ex vivo RT-qPCR-based assay for human peripheral leukocyte responsiveness to DEX revealed a transcriptional dysregulation of GC-regulated genes (FKBP5, IL1R2, and GILZ) after stimulation with SM-alkylated DEX. Our results present GCs as new biological targets of SM associated with a disturbance of hormone function.


Subject(s)
Alkylating Agents/toxicity , Chemical Warfare Agents/toxicity , Gene Expression Regulation/drug effects , Glucocorticoids/metabolism , Mustard Gas/toxicity , Animals , Betamethasone/pharmacology , Cotinine/analogs & derivatives , Cotinine/pharmacology , Dexamethasone/pharmacology , Genes, Reporter , Glucocorticoids/genetics , HEK293 Cells , Humans , Luciferases/genetics , Renilla , Transfection
12.
J Cell Biochem ; 120(3): 4203-4213, 2019 03.
Article in English | MEDLINE | ID: mdl-30260034

ABSTRACT

Pterygium is a triangular-shaped hyperplastic growth, characterized by conjunctivalization, inflammation, and connective tissue remodeling. Our previous meta-analysis found that cigarette smoking is associated with a reduced risk of pterygium. Yet, the biological effect of cigarette smoke components on pterygium has not been studied. Here we reported the proliferation and migration properties of human primary pterygium cells with continuous exposure to nicotine and cotinine. Human primary pterygium cells predominantly expressed the α5, ß1, and γ subunits of the nicotinic acetylcholine receptor. Continuous exposure to the mixture of 0.15 µM nicotine and 2 µM cotinine retarded pterygium cell proliferation by 16.04% (P = 0.009) and hindered their migration by 11.93% ( P = 0.039), without affecting cell apoptosis. SNAIL and α-smooth muscle actin protein expression was significantly downregulated in pterygium cells treated with 0.15 µM nicotine-2 µM cotinine mixture by 1.33- ( P = 0.036) and 1.31-fold ( P = 0.001), respectively. Besides, the 0.15 µM nicotine-2 µM cotinine mixture also reduced matrix metalloproteinase (MMP)-1 and MMP-9 expressions in pterygium cells by 1.56- ( P = 0.043) and 1.27-fold ( P = 0.012), respectively. In summary, this study revealed that continuous exposure of nicotine and cotinine inhibited human primary pterygium cell proliferation and migration in vitro by reducing epithelial-to-mesenchymal transition and MMP protein expression, partially explaining the lower incidence of pterygium in cigarette smokers.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Cotinine/pharmacology , Nicotine/pharmacology , Pterygium/metabolism , Actins/metabolism , Apoptosis/drug effects , Cells, Cultured , Epithelial-Mesenchymal Transition/drug effects , Humans , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 9/metabolism , Pterygium/pathology , Receptors, Nicotinic/metabolism , Signal Transduction/drug effects , Smoking/metabolism , Snail Family Transcription Factors/metabolism
13.
Neurosci Lett ; 696: 146-150, 2019 03 23.
Article in English | MEDLINE | ID: mdl-30579994

ABSTRACT

Caffeine and tobacco smoke are among the most frequently self-administered licit psychoactive drugs in the world. Both drugs affect anxiety levels, however, little is known on the impact of the dual exposure in the adolescent brain, the period during which smoking begins. Considering that anxiety is a relevant factor for smoking maintenance and relapse, we investigated the effects of lifelong exposure to caffeine on anxiety levels of Swiss mice exposed to tobacco smoke during adolescence. Caffeine was administrated during all prenatal and postnatal life (CAF, 0.1 g/l to drink). From postnatal day 30-45, animals were exposed to tobacco smoke (SMK, whole body exposure, 8 h/day) generated from research cigarettes type 3R4F (nicotine = 0.73 mg/per cigarette). Four groups were analyzed: (1) CAF + SMK exposure; (2) SMK exposure; (3) CAF exposure; (4) Control. Anxiety levels were assessed in the elevated plus maze at the end of smoke exposure (PN45), at short- (PN55) and long-term (PN75) withdrawal. Caffeine exposure reduced decision making time (time in center of maze) during adolescence (PN45 and PN55). In addition, caffeine increased anxiety-like behavior during long-term tobacco smoke withdrawal. The present study provides experimental evidence that caffeine and tobacco smoke during adolescence interact resulting in emotional dysregulation during tobacco smoke withdrawal. Particularly, increased anxiety-like behavior during long-term withdrawal in CAF + SMK animals demonstrates late-emergent effects. In this sense, our data suggest that lifelong caffeine exposure may be an important factor in tobacco relapse.


Subject(s)
Anxiety/chemically induced , Behavior, Animal/drug effects , Caffeine/pharmacology , Nicotiana/adverse effects , Aging , Animals , Anxiety/psychology , Cotinine/pharmacology , Exploratory Behavior/drug effects , Female , Male , Mice , Nicotine/pharmacology , Substance Withdrawal Syndrome/psychology
14.
Oxid Med Cell Longev ; 2018: 3172480, 2018.
Article in English | MEDLINE | ID: mdl-30533170

ABSTRACT

Cigarette smoking has been identified as a major risk factor for osteoporosis decades ago. Several studies have shown a direct relationship between cigarette smoking, decreased bone mineral density, and impaired fracture healing. However, the mechanisms behind impaired fracture healing and cigarette smoking are yet to be elucidated. Migration and osteogenesis of mesenchymal stem/stromal cells (MSCs) into the fracture site play a vital role in the process of fracture healing. In human nicotine, the most pharmacologically active and major addictive component present in tobacco gets rapidly metabolized to the more stable cotinine. This study demonstrates that physiological concentrations of both nicotine and cotinine do not affect the osteogenic differentiation of MSCs. However, cigarette smoke exposure induces oxidative stress by increasing superoxide radicals and reducing intracellular glutathione in MSCs, negatively affecting osteogenic differentiation. Although, not actively producing reactive oxygen species (ROS) nicotine and cotinine inhibit catalase and glutathione reductase activity, contributing to an accumulation of ROS by cigarette smoke exposure. Coincubation with N-acetylcysteine or L-ascorbate improves impaired osteogenesis caused by cigarette smoke exposure by both activation of nuclear factor erythroid 2-related factor 2 (Nrf2) signaling and scavenging of ROS, which thus might represent therapeutic targets to support fracture healing in smokers.


Subject(s)
Antioxidants/metabolism , Cotinine/pharmacology , Nicotine/pharmacology , Osteogenesis/drug effects , Smoke/adverse effects , Tobacco Products/toxicity , Catalase/metabolism , Cell Differentiation/drug effects , Cell Line , Glutathione Reductase/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Reactive Oxygen Species/metabolism , Tobacco Products/adverse effects
15.
Exp Mol Med ; 50(5): 1-14, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29795377

ABSTRACT

Antibody-drug conjugates (ADCs) can selectively deliver cytotoxic agents to tumor cells and are frequently more potent than naked antibodies. However, optimization of the conjugation process between antibodies and cytotoxic agents and characterization of ADCs are laborious and time-consuming processes. Here, we describe a novel ADC platform using a tetravalent bispecific antibody that simultaneously binds to the tumor-associated antigen and a hapten conjugated to a cytotoxic agent. We selected cotinine as the hapten because it is not present in biological systems and is inert and nontoxic. We prepared an anti-epidermal growth factor receptor (EGFR) × cotinine bispecific antibody and mixed it with an equimolar amount of cotinine-conjugated duocarmycin to form the ADC. This ADC showed significant in vitro and in vivo antitumor activity against EGFR-positive, cetuximab-refractory lung adenocarcinoma cells with KRAS mutations.


Subject(s)
Antibodies, Bispecific/pharmacology , Cotinine/pharmacology , Indoles/pharmacology , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/pharmacokinetics , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab/pharmacology , Cotinine/chemistry , Cotinine/pharmacokinetics , Disease Models, Animal , ErbB Receptors/metabolism , Female , Humans , Male , Mice, Inbred BALB C , Single-Chain Antibodies/metabolism , Tissue Distribution , Xenograft Model Antitumor Assays
16.
Mol Neurobiol ; 55(10): 7949-7960, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29488138

ABSTRACT

Failure in fear extinction is one of the more troublesome characteristics of posttraumatic stress disorder (PTSD). Cotinine facilitates fear memory extinction and reduces depressive-like behavior when administered 24 h after fear conditioning in mice. In this study, it was investigated the behavioral and molecular effects of cotinine, and other antidepressant preparations infused intranasally. Intranasal (IN) cotinine, IN krill oil, IN cotinine plus krill oil, and oral sertraline were evaluated on depressive-like behavior and fear retention and extinction after fear conditioning in C57BL/6 mice. Since calcineurin A has been involved in facilitating fear extinction in rodents, we also investigated changes of calcineurin in the hippocampus, a region key on contextual fear extinction. Short-term treatment with cotinine formulations was superior to krill oil and oral sertraline in reducing depressive-like behavior and fear consolidation and enhancing contextual fear memory extinction in mice. IN krill oil slowed the extinction of fear. IN cotinine preparations increased the levels of calcineurin A in the hippocampus of conditioned mice. In the light of the results, the future investigation of the use of IN cotinine preparations for the extinction of contextual fear memory and treatment of treatment-resistant depression (TRD) in PTSD is discussed.


Subject(s)
Calcineurin/metabolism , Cotinine/pharmacology , Depression/drug therapy , Depression/psychology , Extinction, Psychological/drug effects , Fear/drug effects , Hippocampus/metabolism , Oils/pharmacology , Administration, Intranasal , Animals , Behavior, Animal , Conditioning, Psychological , Cotinine/administration & dosage , Cotinine/therapeutic use , Euphausiacea/chemistry , Mice, Inbred C57BL , Models, Biological , Oils/administration & dosage , Sertraline/pharmacology
17.
Mol Neurobiol ; 55(8): 6700-6711, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29335846

ABSTRACT

Post-traumatic stress disorder (PTSD) is a mental disorder that may develop after exposure to exceptionally threatening or unescapable horrifying events. Actual therapies fail to alleviate the emotional suffering and cognitive impairment associated with this disorder, mostly because they are ineffective in treating the failure to extinguish trauma memories in a great percentage of those affected. In this review, current behavioral, cellular, and molecular evidence supporting the use of cotinine for treating PTSD are reviewed. The role of the positive modulation by cotinine of the nicotinic acetylcholine receptors (nAChRs) and their downstream effectors, the protection of astroglia, and the inhibition of microglia in the PTSD brain are also discussed.


Subject(s)
Cotinine/pharmacology , Extinction, Psychological/drug effects , Memory/drug effects , Stress Disorders, Post-Traumatic/psychology , Stress Disorders, Post-Traumatic/therapy , Animals , Humans , Neuronal Plasticity/drug effects , Receptors, Nicotinic/metabolism
18.
Sci Rep ; 7(1): 3139, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28600524

ABSTRACT

Cigarette smoking is a major risk factor for age-related macular degeneration (AMD), in which progressive retinal pigment epithelial (RPE) cell degeneration is a major pathological change. Nicotine is a major biologically active component in cigarette smoke. It is continuously catabolized into cotinine, which has longer half-life and higher concentration in tissue cells and fluids. Here we hypothesized that continuous exposure of cotinine has more potent effects on human RPE cell properties than nicotine. Human RPE cell line (ARPE-19) was treated continuously with 1-2 µM of nicotine and/or cotinine for 7 days. RPE cells treated with 2 µM cotinine and nicotine-cotinine mixture has lower MTT signals without significant changes in cell apoptosis or integrity. Moreover, RPE cell migration was retarded under cotinine treatments, but not nicotine. Both nicotine and cotinine treatments attenuated the phagocytotic activity of RPE cells. In addition, cotinine and nicotine-cotinine mixture suppressed VEGF and IL-8 expression and upregulated TIMP-2 expression. Expressions of autophagy genes were upregulated by the cotinine treatment, whereas expressions of epithelial-to-mesenchymal transition markers were downregulated. In conclusion, our study, for the first time, demonstrated that cotinine, rather than nicotine, affects the properties of RPE cells in vitro, which could explain the smoking-induced RPE pathology.


Subject(s)
Cotinine/pharmacology , Nicotine/pharmacology , Retinal Pigment Epithelium/drug effects , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Humans , Interleukin-8/metabolism , Phagocytosis/drug effects , Rats , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Vascular Endothelial Growth Factor A/metabolism
19.
Article in English | MEDLINE | ID: mdl-28536070

ABSTRACT

Alzheimer's disease (AD) is associated with the progressive aggregation of hyperphosphorylated forms of the microtubule associated protein Tau in the central nervous system. Cotinine, the main metabolite of nicotine, reduced working memory deficits, synaptic loss, and amyloid ß peptide aggregation into oligomers and plaques as well as inhibited the cerebral Tau kinase, glycogen synthase 3ß (GSK3ß) in the transgenic (Tg)6799 (5XFAD) mice. In this study, the effect of cotinine on visual recognition memory and cortical Tau phosphorylation at the GSK3ß sites Serine (Ser)-396/Ser-404 and phospho-CREB were investigated in the Tg6799 and non-transgenic (NT) littermate mice. Tg mice showed short-term visual recognition memory impairment in the novel object recognition test, and higher levels of Tau phosphorylation when compared to NT mice. Cotinine significantly improved visual recognition memory performance increased CREB phosphorylation and reduced cortical Tau phosphorylation. Potential mechanisms underlying theses beneficial effects are discussed.


Subject(s)
Cerebral Cortex/metabolism , Cotinine/pharmacology , Memory/drug effects , Recognition, Psychology/drug effects , Visual Perception/drug effects , tau Proteins/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Mice , Mice, Transgenic , Phosphorylation/drug effects
20.
Curr Alzheimer Res ; 14(11): 1155-1163, 2017.
Article in English | MEDLINE | ID: mdl-28356047

ABSTRACT

BACKGROUND: The vascular endothelial growth factor (VEGF) is a neuroprotective cytokine that promotes neurogenesis and angiogenesis in the brain. In animal models, it has been shown that environmental enrichment and exercise, two non-pharmacological interventions that are beneficial decreasing the progression of Alzheimer disease (AD) and depressive-like behavior, enhance hippocampal VEGF expression and neurogenesis. Furthermore, the stimulation of VEGF expression promotes neurotransmission and synaptic plasticity processes such as neurogenesis. It is thought that these VEGF actions in the brain, may underly its beneficial therapeutic effects against psychiatric and other neurological conditions. CONCLUSION: In this review, evidence linking VEGF deficit with the development of AD as well as the potential role of VEGF signaling as a therapeutic target for cotinine and other interventions in neurodegenerative conditions are discussed.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Cotinine/therapeutic use , Neuroprotective Agents/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Animals , Cotinine/pharmacology , Humans , Neuroprotective Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...