Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Addict Biol ; 29(3): e13382, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38488467

RESUMO

Methamphetamine (METH) is a highly addictive psycho-stimulant that induces addictive behaviour by stimulating increased dopamine release in the nucleus accumbens (NAc). The sarco/endoplasmic reticulum calcium ion transport ATPases (SERCA or ATP2A) is a calcium ion (Ca2+) pump in the endoplasmic reticulum (ER) membrane. SERCA2b is a SERCA subtype mainly distributed in the central nervous system. This study used conditioned place preference (CPP), a translational drug reward model, to observe the effects of SERCA and SERCA2b on METH-CPP in mice. Result suggested that the activity of SERCA was significantly decreased in NAc after METH-CPP. Intraperitoneal SERCA agonist CDN1163 injection or bilateral CDN1163 microinjection in the NAc inhibited METH-CPP formation. SERCA2b overexpression by the Adeno-associated virus can reduce the DA release of NAc and inhibit METH-CPP formation. Although microinjection of SERCA inhibitor thapsigargin in the bilateral NAc did not significantly aggravate METH-CPP, interference with SERCA2b expression in NAc by adeno-associated virus increased DA release and promoted METH-CPP formation. METH reduced the SERCA ability to transport Ca2+ into the ER in SHSY5Y cells in vitro, which was reversed by CDN1163. This study revealed that METH dysregulates intracellular calcium balance by downregulating SERCA2b function, increasing DA release in NAc and inducing METH-CPP formation. Drugs that target SERCA2b may have the potential to treat METH addiction.


Assuntos
Benzamidas , Estimulantes do Sistema Nervoso Central , Metanfetamina , Camundongos , Animais , Metanfetamina/farmacologia , Metanfetamina/metabolismo , Núcleo Accumbens , Cálcio/metabolismo , Aminoquinolinas/metabolismo , Aminoquinolinas/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Estimulantes do Sistema Nervoso Central/metabolismo
2.
Cell Biol Toxicol ; 40(1): 13, 2024 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-38347241

RESUMO

AIMS: Nuclear protein 1 (Nupr1) is a multifunctional stress-induced protein involved in the regulation of tumorigenesis, apoptosis, and autophagy. However, its role in pulmonary hypertension (PH) after METH exposure remains unexplored. In this study, we aimed to investigate whether METH can induce PH and describe the role and mechanism of Nupr1 in the development of PH. METHODS AND RESULTS: Mice were made to induce pulmonary hypertension (PH) upon chronic intermittent treatment with METH. Their right ventricular systolic pressure (RVSP) was measured to assess pulmonary artery pressure. Pulmonary artery morphometry was determined by H&E staining and Masson staining. Nupr1 expression and function were detected in human lungs, mice lungs exposed to METH, and cultured pulmonary arterial smooth muscle cells (PASMCs) with METH treatment. Our results showed that chronic intermittent METH treatment successfully induced PH in mice. Nupr1 expression was increased in the cultured PASMCs, pulmonary arterial media from METH-exposed mice, and METH-ingested human specimens compared with control. Elevated Nupr1 expression promoted PASMC phenotype change from contractile to synthetic, which triggered pulmonary artery remodeling and resulted in PH formation. Mechanistically, Nupr1 mediated the opening of store-operated calcium entry (SOCE) by activating the expression of STIM1, thereby promoting Ca2+ influx and inducing phenotypic conversion of PASMCs. CONCLUSIONS: Nupr1 activation could promote Ca2+ influx through STIM1-mediated SOCE opening, which promoted METH-induced pulmonary artery remodeling and led to PH formation. These results suggested that Nupr1 played an important role in METH-induced PH and might be a potential target for METH-related PH therapy.


Assuntos
Hipertensão Pulmonar , Metanfetamina , Camundongos , Humanos , Animais , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Metanfetamina/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas Nucleares/metabolismo , Células Cultivadas , Artéria Pulmonar/metabolismo , Miócitos de Músculo Liso/metabolismo , Proliferação de Células
3.
Nature ; 624(7992): 663-671, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37935377

RESUMO

Trace amine-associated receptor 1 (TAAR1), the founding member of a nine-member family of trace amine receptors, is responsible for recognizing a range of biogenic amines in the brain, including the endogenous ß-phenylethylamine (ß-PEA)1 as well as methamphetamine2, an abused substance that has posed a severe threat to human health and society3. Given its unique physiological role in the brain, TAAR1 is also an emerging target for a range of neurological disorders including schizophrenia, depression and drug addiction2,4,5. Here we report structures of human TAAR1-G-protein complexes bound to methamphetamine and ß-PEA as well as complexes bound to RO5256390, a TAAR1-selective agonist, and SEP-363856, a clinical-stage dual agonist for TAAR1 and serotonin receptor 5-HT1AR (refs. 6,7). Together with systematic mutagenesis and functional studies, the structures reveal the molecular basis of methamphetamine recognition and underlying mechanisms of ligand selectivity and polypharmacology between TAAR1 and other monoamine receptors. We identify a lid-like extracellular loop 2 helix/loop structure and a hydrogen-bonding network in the ligand-binding pockets, which may contribute to the ligand recognition in TAAR1. These findings shed light on the ligand recognition mode and activation mechanism for TAAR1 and should guide the development of next-generation therapeutics for drug addiction and various neurological disorders.


Assuntos
Metanfetamina , Fenetilaminas , Receptores Acoplados a Proteínas G , Humanos , Ligantes , Metanfetamina/metabolismo , Doenças do Sistema Nervoso/metabolismo , Fenetilaminas/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Polifarmacologia , Ligação de Hidrogênio
4.
BMC Pharmacol Toxicol ; 24(1): 33, 2023 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-37208773

RESUMO

BACKGROUND: Methamphetamine is widely abused in all parts of the world. It has been reported that short-term and long-term methamphetamine exposure could damage the dopaminergic system and induce cardiomyopathy and cardiotoxicity via mitochondrial dysfunction and oxidative stress. Vanillic acid (VA), a phenolic acid compound derived from plants, is known for its antioxidant and mitochondrial protection properties. METHODS: In the current study we used VA for attenuating of Methamphetamine-induced mitochondrial toxicity in cardiac mitochondria. Isolated mitochondria obtained from rat heart were grouped as: control, methamphetamine (250 µM), VA (10, 50 and 100 µM) was cotreated with methamphetamine (250 µM) and VA (100 µM) alone. After 60 min, mitochondrial fraction including: succinate dehydrogenases (SDH) activity, mitochondrial membrane potential (MMP), mitochondrial swelling, mitochondrial glutathione (GSH), reactive oxygen species (ROS) and lipid peroxidation (LPO) were evaluated. RESULTS: Methamphetamine exposure significantly disrupted mitochondrial function and induced ROS formation, lipid peroxidation, GSH depletion, MMP collapse and mitochondrial swelling, while VA significantly increased SDH activity as indicator of mitochondrial toxicity and dysfunction. VA also significantly decreased ROS formation, lipid peroxidation, mitochondrial swelling, MMP collapse and depletion of GSH in cardiac mitochondria in the presence of methamphetamine. CONCLUSION: These findings suggested that VA is able to reduce methamphetamine-induced mitochondrial dysfunction and oxidative stress. Our results demonstrate that VA could potentially serve as a promising and accessible cardioprotective agent against methamphetamine-induced cardiotoxicity, via antioxidant and mitochondrial protection properties.


Assuntos
Antioxidantes , Metanfetamina , Ratos , Animais , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Metanfetamina/toxicidade , Metanfetamina/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ácido Vanílico/farmacologia , Ácido Vanílico/metabolismo , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/metabolismo , Estresse Oxidativo , Mitocôndrias/metabolismo , Glutationa/metabolismo , Peroxidação de Lipídeos , Potencial da Membrana Mitocondrial
5.
Int J Mol Sci ; 24(6)2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36982742

RESUMO

Methamphetamine, a highly addictive central nervous system (CNS) stimulant, is used worldwide as an anorexiant and attention enhancer. Methamphetamine use during pregnancy, even at therapeutic doses, may harm fetal development. Here, we examined whether exposure to methamphetamine affects the morphogenesis and diversity of ventral midbrain dopaminergic neurons (VMDNs). The effects of methamphetamine on morphogenesis, viability, the release of mediator chemicals (such as ATP), and the expression of genes involved in neurogenesis were evaluated using VMDNs isolated from the embryos of timed-mated mice on embryonic day 12.5. We demonstrated that methamphetamine (10 µM; equivalent to its therapeutic dose) did not affect the viability and morphogenesis of VMDNs, but it reduced the ATP release negligibly. It significantly downregulated Lmx1a, En1, Pitx3, Th, Chl1, Dat, and Drd1 but did not affect Nurr1 or Bdnf expression. Our results illustrate that methamphetamine could impair VMDN differentiation by altering the expression of important neurogenesis-related genes. Overall, this study suggests that methamphetamine use may impair VMDNs in the fetus if taken during pregnancy. Therefore, it is essential to exercise strict caution for its use in expectant mothers.


Assuntos
Estimulantes do Sistema Nervoso Central , Metanfetamina , Efeitos Tardios da Exposição Pré-Natal , Humanos , Feminino , Camundongos , Animais , Neurônios Dopaminérgicos/metabolismo , Metanfetamina/toxicidade , Metanfetamina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Mesencéfalo/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Trifosfato de Adenosina/metabolismo , Diferenciação Celular
6.
BMC Neurosci ; 24(1): 2, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36631757

RESUMO

BACKGROUND: The head-twitch response (HTR) in mice is considered a behavioral model for hallucinogens and serotonin 5-HT2A receptor function, as well as Tourette syndrome in humans. It is mediated by 5-HT2A receptor agonists such as ( ±)- 2,5-dimethoxy-4-iodoamphetamine (DOI) in the prefrontal cortex (PFC). The 5-HT2A antagonist EMD 281014, can prevent both DOI-induced HTR during ageing and c-fos expression in different regions of PFC. Moreover, the nonselective monoamine releaser methamphetamine (MA) suppressed DOI-induced HTR through ageing via concomitant activation of inhibitory 5-HT1A receptors, but enhanced DOI-evoked c-fos expression. d-Fenfluramine is a selective 5-HT releaser and induces HTR in mice, whereas MA does not. Currently, we investigated whether EMD 281014 or MA would alter: (1) d-fenfluramine-induced HTR frequency in 20-, 30- and 60-day old mice, (2) d-fenfluramine-evoked c-fos expression in PFC, and (3) whether blockade of inhibitory serotonergic 5-HT1A- or adrenergic ɑ2-receptors would prevent suppressive effect of MA on d-fenfluramine-induced HTR. RESULTS: EMD 281014 (0.001-0.05 mg/kg) or MA (0.1-5 mg/kg) blocked d-fenfluramine-induced HTR dose-dependently during ageing. The 5-HT1A antagonist WAY 100635 countered the inhibitory effect of MA on d-fenfluramine-induced HTR in 30-day old mice, whereas the adrenergic ɑ2 antagonist RS 79948 reversed MA's inhibitory effect in both 20- and 30- day old mice. d-Fenfluramine significantly increased c-fos expressions in PFC regions. MA (1 mg/kg) pretreatment significantly increased d-fenfluramine-evoked c-fos expression in different regions of PFC. EMD 281014 (0.05 mg/kg) failed to prevent d-fenfluramine-induced c-fos expression, but significantly increased it in one PFC region (PrL at - 2.68 mm). CONCLUSION: EMD 281014 suppressed d-fenfluramine-induced HTR but failed to prevent d-fenfluramine-evoked c-fos expression which suggest involvement of additional serotonergic receptors in the mediation of evoked c-fos. The suppressive effect of MA on d-fenfluramine-evoked HTR is due to well-recognized functional interactions between stimulatory 5-HT2A- and the inhibitory 5-HT1A- and ɑ2-receptors. MA-evoked increases in c-fos expression in PFC regions are due to the activation of diverse monoaminergic receptors through increased synaptic concentrations of 5-HT, NE and/or DA, which may also account for the additive effect of MA on d-fenfluramine-evoked changes in c-fos expression. Our findings suggest potential drug receptor functional interaction during development when used in combination.


Assuntos
Fenfluramina , Metanfetamina , Córtex Pré-Frontal , Proteínas Proto-Oncogênicas c-fos , Animais , Humanos , Camundongos , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia , Envelhecimento/metabolismo , Fenfluramina/metabolismo , Fenfluramina/farmacologia , Metanfetamina/metabolismo , Metanfetamina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Receptor 5-HT2A de Serotonina/efeitos dos fármacos , Receptor 5-HT2A de Serotonina/metabolismo , Serotonina/metabolismo , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo
7.
Neuropsychopharmacology ; 48(3): 478-488, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36109595

RESUMO

Drug-associated conditioned cues promote subjects to recall drug reward memory, resulting in drug-seeking and reinstatement. A consolidated memory becomes unstable after recall, such that the amnestic agent can disrupt the memory during the reconsolidation stage, which implicates a potential therapeutic strategy for weakening maladaptive memories. The basolateral amygdala (BLA) involves the association of conditioned cues with reward and aversive valences and projects the information to the nucleus accumbens (NAc) that mediates reward-seeking. However, whether the BLA-NAc projection plays a role in drug-associated memory reactivation and reconsolidation is unknown. We used methamphetamine (MeAM) conditioned place preference (CPP) to investigate the role of BLA-NAc neural projection in the memory reconsolidation. Two weeks before CPP training, we infused adeno-associated virus (AAV) carrying the designer receptor exclusively activated by designer drugs (DREADD) or control constructs. We infused clozapine-N-oxide (CNO) after the recall test to manipulate the neural activity of BLA-NAc projections in mice. We found that after recall, DREADD-mediated inhibition of BLA neurons projecting to the NAc core blunted consolidated MeAM-associated memory. Inhibition of BLA glutamatergic nerve terminals in the NAc core 1 h after recall disrupted consolidated MeAM-associated memory. However, inhibiting this pathway after the time window of reconsolidation failed to affect memory. Furthermore, under the condition without memory retrieval, DREADD-mediated activation of BLA-NAc core projection was required for amnesic agents to disrupt consolidated MeAM-associated memory. Our findings provide evidence that the BLA-NAc pathway activity is involved in the post-retrieval processing of MeAM-associated memory in CPP.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Metanfetamina , Camundongos , Animais , Metanfetamina/farmacologia , Metanfetamina/metabolismo , Tonsila do Cerebelo/metabolismo , Núcleo Accumbens/metabolismo , Memória/fisiologia
8.
Int J Mol Sci ; 23(19)2022 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-36232524

RESUMO

Methamphetamine (METH) is a highly addictive drug abused by millions of users worldwide, thus becoming a global health concern with limited management options. The inefficiency of existing treatment methods has driven research into understanding the mechanisms underlying METH-induced disorders and finding effective treatments. This study aims to understand the complex interactions of the gastrointestinal-immune-nervous systems following an acute METH dose administration as one of the potential underlying molecular mechanisms concentrating on the impact of METH abuse on gut permeability. Findings showed a decreased expression of tight junction proteins ZO-1 and EpCAm in intestinal tissue and the presence of FABP-1 in sera of METH treated mice suggests intestinal wall disruption. The increased presence of CD45+ immune cells in the intestinal wall further confirms gut wall inflammation/disruption. In the brain, the expression of inflammatory markers Ccl2, Cxcl1, IL-1ß, TMEM119, and the presence of albumin were higher in METH mice compared to shams, suggesting METH-induced blood-brain barrier disruption. In the spleen, cellular and gene changes are also noted. In addition, mice treated with an acute dose of METH showed anxious behavior in dark and light, open field, and elevated maze tests compared to sham controls. The findings on METH-induced inflammation and anxiety may provide opportunities to develop effective treatments for METH addiction in the future.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas , Estimulantes do Sistema Nervoso Central , Metanfetamina , Albuminas/metabolismo , Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Animais , Ansiedade , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Molécula de Adesão da Célula Epitelial/metabolismo , Inflamação/metabolismo , Metanfetamina/metabolismo , Metanfetamina/toxicidade , Camundongos
9.
Brain Behav Immun ; 106: 247-261, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36089218

RESUMO

The α-synuclein (α-syn) is involved in methamphetamine (METH)-induced neurotoxicity. Neurons can transfer excessive α-syn to neighboring neurons and glial cells. The effects of α-syn aggregation in astrocytes after METH exposure on the blood-brain barrier (BBB) remains unclear. Our previous study demonstrated that nuclear receptor-related protein 1 (Nurr1), a member of the nuclear receptor family widely expressed in the brain, was involved in the process of METH-induced α-syn accumulated in astrocytes to activate neuroinflammation. The role Nurr1 plays in astrocyte-mediated neuroinflammation, which results in BBB injury induced by METH, remains uncertain. This study found that METH up-regulated α-syn expression in neurons extended to astrocytes, thereby eliciting astrocyte activation, increasing and decreasing IL-1ß, IL-6, TNF-α, and GDNF levels by down-regulating Nurr1 expression, and ultimately damaging the BBB. Specifically, the permeability of BBB to Evans blue and sodium fluorescein (NaF) increased; IgG deposits in the brain parenchyma increased; the Claudin5, Occludin, and PDGFRß levels decreased. Several ultrastructural pathological changes occurred in the BBB, such as abnormal cerebral microvascular diameter, astrocyte end-foot swelling, decreased pericyte coverage, and loss of tight junctions. However, knockout or inhibition of α-syn or astrocyte-specific overexpression of Nurr1 partially alleviated these symptoms and BBB injury. Moreover, the in vitro experiments confirmed that METH increased α-syn level in the primary cultured neurons, which could be further transferred to primary cultured astrocytes, resulting in decreased Nurr1 levels. The decreased Nurr1 levels mediated the increase of IL-1ß, IL-6, and TNF-α, and the decrease of GDNF, thereby changing the permeability to NaF, transendothelial electrical resistance, and Claudin5 and Occludin levels of primary cultured brain microvascular endothelial cells. Based on our findings, we proposed a new mechanism to elucidate METH-induced BBB injury and presented α-syn and Nurr1 as promising drug intervention targets to reduce BBB injury and resulting neurotoxicity in METH abusers.


Assuntos
Estimulantes do Sistema Nervoso Central , Metanfetamina , Síndromes Neurotóxicas , Astrócitos/metabolismo , Barreira Hematoencefálica/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Células Endoteliais/metabolismo , Azul Evans/metabolismo , Azul Evans/farmacologia , Fluoresceína/metabolismo , Fluoresceína/farmacologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Humanos , Imunoglobulina G , Interleucina-6/metabolismo , Metanfetamina/metabolismo , Doenças Neuroinflamatórias , Neurônios/metabolismo , Síndromes Neurotóxicas/metabolismo , Ocludina/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , alfa-Sinucleína/metabolismo
10.
Psychopharmacology (Berl) ; 239(9): 2997-3008, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35881147

RESUMO

RATIONALE: MicroRNA (miRNA) control of post-transcription gene expression in the nucleus accumbens (NAc) has been implicated in methamphetamine (METH) dependence. Conditioned place preference (CPP) is a classical animal procedure that reflects the rewarding effects of addictive drugs. miR-222-3p has been reported to play a key role in various neurological diseases and is strongly associated with alcohol dependence. Nevertheless, the role of miR-222-3p in METH dependence remains unclear. OBJECTIVE: To explore the molecular mechanisms underlying the role of miR-222-3p in the NAc in METH-induced CPP. METHODS: miR-222-3p expression in the NAc of METH-induced CPP mice was detected by quantitative real-time (qPCR). Following adeno-associated virus (AAV)-mediated overexpression or knockdown of miR-222-3p in the NAc, mice were subjected to CPP to investigate the effects of miR-222-3p on METH-induced CPP. Target genes of mir-222-3p were predicted using bioinformatics analysis. Candidate target genes for METH-induced CPP were validated by qPCR. RESULTS: miR-222-3p expression in the NAc was decreased in CPP mice. Overexpression of miR-222-3p in the NAc blunted METH-induced CPP. Ppp3r1, Cdkn1c, Fmr1, and PPARGC1A were identified as target gene transcripts potentially mediating the effects of miR-222-3p on METH-induced CPP. CONCLUSION: Our results highlight miR-222-3p as a key epigenetic regulator in METH-induced CPP and suggest a potential role for miR-222-3p in the regulation of METH-induced reward-related changes in the brain.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas , Estimulantes do Sistema Nervoso Central , Metanfetamina , MicroRNAs , Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Animais , Estimulantes do Sistema Nervoso Central/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Proteína do X Frágil da Deficiência Intelectual , Metanfetamina/metabolismo , Metanfetamina/farmacologia , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Núcleo Accumbens
11.
J Neurovirol ; 28(1): 99-112, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35175539

RESUMO

Macrophages are key elements of the innate immune system. Their HIV-1 infection is a complex process that involves multiple interacting factors and various steps and is further altered by exposure of infected cells to methamphetamine (Meth), a common drug of abuse in people living with HIV. This is reflected by dynamic changes in the intracellular and secreted proteomes of these cells. Quantification of these changes poses a challenge for experimental design and associated analytics. In this study, we measured the effect of Meth on expression of intracellular and secreted galectins-1, -3, and -9 in HIV-1 infected human monocyte-derived macrophages (hMDM) using SWATH-MS, which was further followed by MRM targeted mass spectrometry validation. Cells were exposed to Meth either prior to or after infection. Our results are the first to perform comprehensive quantifications of galectins in primary hMDM cells during HIV-1 infection and Meth exposure a building foundation for future studies on the molecular mechanisms underlying cellular pathology of hMDM resulting from viral infection and a drug of abuse-Meth.


Assuntos
Infecções por HIV , Soropositividade para HIV , HIV-1 , Metanfetamina , Humanos , Macrófagos , Metanfetamina/metabolismo , Metanfetamina/farmacologia
12.
Curr Top Med Chem ; 21(11): 964-975, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34061003

RESUMO

The peroxisome proliferator activated receptors (PPARs) are a superfamily of well-recognized ligand-binding nuclear receptors comprising three isoforms: PPARα, PPARγ, and PPARß/δ. In response to endogenous lipid messengers, PPARs trigger the transcription of genes related to a wider spectrum of physiological phenomena, including fatty acid oxidation, inflammation, adipogenesis, among many others. Thus, the importance of PPARs as putative protective therapy in health issues has increased the interest of studying these nuclear receptors, including the management of neurodegenerative disorders, multiple sclerosis, and likely addiction. In recent years, several pieces of evidence from animal models have demonstrated the promising role of PPARs as a critical element for interventions in addictive behaviors by reducing the reinforcing properties of addictive substances such as alcohol. However, there is a lack of data in the scope and has so far been unexplored the function of PPARs in additional drugs such as cannabis, opioids, methamphetamine, or cocaine. A similar scenario has been found for the management of binge-type eating disorders. Thus, here we review recent advances in understanding the relevance of the PPAR controlling addiction.


Assuntos
Comportamento Aditivo/tratamento farmacológico , Terapia de Alvo Molecular/métodos , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Preparações Farmacêuticas/metabolismo , Álcoois/metabolismo , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Cannabis/metabolismo , Cocaína/metabolismo , Cocaína/farmacologia , Humanos , Ligantes , Metanfetamina/metabolismo , Metanfetamina/farmacologia , Doenças Neurodegenerativas/tratamento farmacológico , Nicotina/metabolismo , Oxirredução , Isoformas de Proteínas , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo
13.
Toxicol Appl Pharmacol ; 416: 115442, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33609514

RESUMO

Cathinone derivatives are the most representative group within new drugs market, which have been described as neurotoxic. Since cathinones, as pentedrone and methylone, are available as racemates, it is our aim to study the neuronal cytotoxicity induced by each enantiomer. Therefore, a dopaminergic SH-SY5Y cell line was used to evaluate the hypothesis of enantioselectivity of pentedrone and methylone enantiomers on cytotoxicity, oxidative stress, and membrane efflux transport (confirmed by in silico studies). Our study demonstrated enantioselectivity of these cathinones, being the S-(+)-pentedrone and R-(+)-methylone the most oxidative enantiomers and also the most cytotoxic, suggesting the oxidative stress as main cytotoxic mechanism, as previously described in in vitro studies. Additionally, the efflux transporter multidrug resistance associated protein 1 (MRP1) seems to play, together with GSH, a selective protective role against the cytotoxicity induced by R-(-)-pentedrone enantiomer. It was also observed an enantioselectivity in the binding to P-glycoprotein (P-gp), another efflux protein, being the R-(-)-pentedrone and S-(-)-methylone the most transported enantiomeric compounds. These results were confirmed, in silico, by docking studies, revealing that R-(-)-pentedrone is the enantiomer with highest affinity to MRP1 and S-(-)-methylone and R-(-)-pentedrone are the enantiomers with highest affinity to P-gp. In conclusion, our data demonstrated that pentedrone and methylone present enantioselectivity in their cytotoxicity, which seems to involve different oxidative reactivity as well as different affinity to the P-gp and MRP1 that together with GSH play a protective role.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Alcaloides/toxicidade , Neurônios Dopaminérgicos/efeitos dos fármacos , Metanfetamina/análogos & derivados , Metilaminas/toxicidade , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Pentanonas/toxicidade , Alcaloides/química , Alcaloides/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Relação Dose-Resposta a Droga , Glutationa/metabolismo , Humanos , Metanfetamina/química , Metanfetamina/metabolismo , Metanfetamina/toxicidade , Metilaminas/química , Metilaminas/metabolismo , Simulação de Acoplamento Molecular , Pentanonas/química , Pentanonas/metabolismo , Ligação Proteica , Estereoisomerismo
14.
Anal Chem ; 93(3): 1604-1611, 2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33356171

RESUMO

Carboxylic metabolites are an important class of metabolites, which widely exist in mammals with various types. Chemical isotope labeling liquid chromatography-mass spectrometry (CIL-LC-MS) has been widely used for the detection of carboxylated metabolites. However, high coverage analysis of carboxylated metabolites in biological samples is still challenging due to improper reactivity and selectivity of labeling reagents to carboxylated metabolites. In this study, we used N-methylphenylethylamine (MPEA) to label various types of carboxylated metabolites including short-chain fatty acids (SCFAs), medium-chain fatty acids (MCFAs), long-chain fatty acids (LCFAs), polycarboxylic acids (polyCAs), amino acids (AAs), and aromatic acids. Additionally, metabolites containing other functional groups, such as phenol, sulfhydryl, and phosphate groups, could not be labeled under the conditions of MPEA labeling. After MPEA labeling, the detection sensitivity of carboxylic acids was increased by 1-2 orders of magnitude, and their chromatographic retention on a reversed-phase (RP) column was enhanced (RT > 3 min). Under optimized labeling conditions, we used MPEA and d3-N-methylphenylethylamine (d3-MPEA) for high coverage screening of carboxylated metabolites in HepG2 cells by ultrahigh-performance liquid chromatography coupled to high-resolution mass spectrometry (UHPLC-HRMS). As a result, a total of 403 potential carboxylated metabolites were obtained of which 68 were confirmed based on our established in-house chemically labeled metabolite database (CLMD). SCFAs, MCFAs, LCFAs, polyCAs, AAs, and aromatic acids were all detected in HepG2 cell extracts. Due to the successful identification of AAs, the current method increased the coverage of carboxylated metabolites compared with our previous work. Moreover, 133 and 109 carboxylated metabolites with changed contents were obtained in HepG2 cells incubated with curcumin and R-3-hydroxybutyric acid, respectively. In general, our established method realized high coverage analysis of carboxylated metabolites in HepG2 cells.


Assuntos
Aminoácidos/análise , Ácidos Carboxílicos/análise , Ácidos Graxos/análise , Metanfetamina/análogos & derivados , Aminoácidos/metabolismo , Ácidos Carboxílicos/metabolismo , Cromatografia Líquida de Alta Pressão , Ácidos Graxos/metabolismo , Células Hep G2 , Humanos , Espectrometria de Massas , Metanfetamina/química , Metanfetamina/metabolismo , Estrutura Molecular
15.
Int J Mol Sci ; 21(17)2020 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-32839415

RESUMO

Methamphetamine (MA) is a highly addictive central nervous system stimulant. Drug addiction is not a static condition but rather a chronically relapsing disorder. Hair is a valuable and stable specimen for chronic toxicological monitoring as it retains toxicants and metabolites. The primary focus of this study was to discover the metabolic effects encompassing diverse pathological symptoms of MA addiction. Therefore, metabolic alterations were investigated in human hair following heavy MA abuse using both targeted and untargeted mass spectrometry and through integrated network analysis. The statistical analyses (t-test, variable importance on projection score, and receiver-operator characteristic curve) demonstrated that 32 metabolites (in targeted metabolomics) as well as 417 and 224 ion features (in positive and negative ionization modes of untargeted metabolomics, respectively) were critically dysregulated. The network analysis showed that the biosynthesis or metabolism of lipids, such as glycosphingolipids, sphingolipids, glycerophospholipids, and ether lipids, as well as the metabolism of amino acids (glycine, serine and threonine; cysteine and methionine) is affected by heavy MA abuse. These findings reveal crucial metabolic effects caused by MA addiction, with emphasis on the value of human hair as a diagnostic specimen for determining drug addiction, and will aid in identifying robust diagnostic markers and therapeutic targets.


Assuntos
Anfetamina/análise , Estimulantes do Sistema Nervoso Central/análise , Cabelo/química , Metanfetamina/análise , Transtornos Relacionados ao Uso de Substâncias/diagnóstico , Adulto , Aminoácidos/química , Aminoácidos/classificação , Aminoácidos/isolamento & purificação , Aminoácidos/metabolismo , Anfetamina/administração & dosagem , Anfetamina/metabolismo , Estudos de Casos e Controles , Estimulantes do Sistema Nervoso Central/administração & dosagem , Estimulantes do Sistema Nervoso Central/metabolismo , Glicerofosfolipídeos/química , Glicerofosfolipídeos/classificação , Glicerofosfolipídeos/isolamento & purificação , Glicerofosfolipídeos/metabolismo , Glicoesfingolipídeos/química , Glicoesfingolipídeos/classificação , Glicoesfingolipídeos/isolamento & purificação , Glicoesfingolipídeos/metabolismo , Humanos , Metabolismo dos Lipídeos/fisiologia , Masculino , Metabolômica/métodos , Metanfetamina/administração & dosagem , Metanfetamina/metabolismo , Pessoa de Meia-Idade , Análise de Componente Principal , Esfingolipídeos/química , Esfingolipídeos/classificação , Esfingolipídeos/isolamento & purificação , Esfingolipídeos/metabolismo , Detecção do Abuso de Substâncias/métodos , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Espectrometria de Massas em Tandem
16.
Med Sci Monit ; 25: 8515-8526, 2019 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-31712546

RESUMO

BACKGROUND Methamphetamine (METH), a confirmed neurotoxic drug, has also reportedly caused several intestinal inflammatory injury cases. The NLRP3 (Nod-like receptor 3 protein) inflammasome can induce several inflammatory injuries by activating IL-1ß and IL-18 when overexpressed. We designed experiments to determine whether METH can cause intestinal inflammatory injury via NLRP3 inflammasome overexpression. MATERIAL AND METHODS IEC-6 cells were classified as control, METH (0.5 mM), and METH (0.5 mM)+MCC950 (100 µM) groups. C57BL/6 mice were separated into control, NS, METH (5 mg/kg), and METH (5 mg/kg)+MCC950 (10 mg/kg) groups (n=10). We detected apoptosis, transepithelial electrical resistance (TEER), and proinflammatory factors (IL-6, INF-γ, TNF-alpha, and NF-kappaB) in the METH cell model. We also assessed proinflammatory factors (IL-6, INF-γ, TNF-alpha, and NF-kappaB) and observed intestinal tissues stained with hematoxylin and eosin (HE) in the METH animal model to explore intestinal inflammatory injury due to METH. After adding MCC950 (an NLRP3 inflammasome inhibitor), we additionally detected NLRP3 inflammasome components (NLRP3, Caspase-1, and ASC), IL-1ß, and IL-18 to estimate the relationship of the NLRP3 inflammasome with intestinal inflammatory injury due to METH. RESULTS METH can lead apoptosis, increase proinflammatory factors (e.g., IL-6, INF-γ, TNF-alpha, and NF-kappaB), and decrease TEER in the METH cell model. In the METH animal model, METH can cause obvious injury and increase proinflammatory factors (e.g., IL-6, INF-γ, TNF-alpha, and NF-kappaB). All the intestinal inflammatory changes due to METH depended on overexpression of the NLRP3 inflammasome and could be ameliorated by MCC950, except for ASC and NF-kappaB. CONCLUSIONS METH, in addition to being a confirmed neurotoxic drug, can also cause severe intestinal inflammatory injury via NLRP3 inflammasome overexpression. NF-kappaB may be an activator of the NLRP3 inflammasome in METH intestinal inflammatory injury.


Assuntos
Mucosa Intestinal/efeitos dos fármacos , Metanfetamina/efeitos adversos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Caspase 1/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Inflamassomos/genética , Inflamassomos/metabolismo , Inflamação/metabolismo , Masculino , Metanfetamina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA
17.
Anal Chem ; 91(14): 9190-9197, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31265257

RESUMO

Macrophages comprise a major component of the human innate immune system that is involved in maintaining homeostasis and responding to infections or other insults. Besides cytokines and chemokines, macrophages presumably influence the surrounding environment by secreting various types of metabolites. Characterization of secreted metabolites under normal and pathological conditions is critical for understanding the complex innate immune system. To investigate the secreted metabolome, we developed a novel workflow consisting of one Reverse Phase (RP) C18 column linked in tandem with a Cogent cholesterol-modified RP C18. This system was used to compare the secreted metabolomes of human monocyte-derived macrophages (hMDM) under normal conditions to those exposed to methamphetamine (Meth). This new experimental approach allowed us to measure 92 metabolites, identify 11 of them as differentially expressed, separate and identify three hydroxymethamphetamine (OHMA) isomers, and identify a new, yet unknown metabolite with a m/z of 192. This study is the first of its kind to address the secreted metabolomic response of hMDM to an insult by Meth. Besides the discovery of novel metabolites secreted by macrophages, we provide a novel methodology to investigate metabolomic profiling.


Assuntos
Macrófagos/efeitos dos fármacos , Metaboloma/efeitos dos fármacos , Metabolômica/métodos , Metanfetamina/metabolismo , Cromatografia Líquida , Humanos , Extração Líquido-Líquido , Espectrometria de Massas , Extração em Fase Sólida
18.
Eur Psychiatry ; 59: 15-24, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30981746

RESUMO

BACKGROUND: Stimulant drugs can cause persistent changes in the brain. Imaging studies show that these changes are most apparent in dopamine transporter (DAT) or receptor availability within the striatum. METHODS: This work focuses on influences of stimulant use on dopaminergic function assessed using nuclear-medicine imaging (PET/SPECT). Included are 39 studies on 655 cocaine, amphetamine, methamphetamine or nicotine users, as well as 690 healthy controls. Metaanalyses were conducted separately for D2/D3 receptors and dopamine transporters of the entire striatum, its subregions caudate and putamen respectively. RESULTS: Meta-analyses results regarding nicotine did not show significant effects between smokers and nonsmokers. In cocaine users there was a significant decrease in dopamine receptor availability in all regions. The striatal DAT availability was significantly increased in cocaine users. Methamphetamine users showed a significantly decreased dopamine receptor and transporter density in all regions. Significant results also indicate a lower transporter availability in all regions. Amphetamine users showed reduced DAT availability in the striatum, as well as in the sub regions. CONCLUSION: This meta-analysis provides evidence that there are ongoing changes in the dopaminergic system associated with the use of stimulants. Especially the results of cocaine, methamphetamine and amphetamine use mainly showed a downregulation. In addition, this meta-analysis is the first to include nicotine. This subset of studies showed evidence for a decreased receptor and DAT availability but no significant results were found in the metaanalyses.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Dependência de Heroína/metabolismo , Metanfetamina/metabolismo , Receptores de Dopamina D2/metabolismo , Encéfalo/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Dopamina , Proteínas da Membrana Plasmática de Transporte de Dopamina/efeitos dos fármacos , Humanos , Neostriado/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único
19.
Psychopharmacology (Berl) ; 236(3): 1001-1014, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30911791

RESUMO

BACKGROUND AND RATIONALE: Polydrug use is a widespread phenomenon, especially among adolescents and young adults. Synthetic cathinones are frequently consumed in combination with other drugs of abuse. However, there is very little information regarding the consequences of this specific consumption pattern. OBJECTIVES: The aim of this review is to introduce this topic and highlight the gaps in the existing literature. In three different sections, we focus on specific interactions of synthetic cathinones with alcohol, cannabinoids, and the stimulants nicotine and cocaine. We then dedicate a section to the existence of sex and gender differences in the effects of synthetic cathinones and the long-term psychophysiological consequences of adolescent and prenatal exposure to these drugs. MAJOR FINDINGS: Epidemiological studies, case reports, and results obtained in animal models point to the existence of pharmacological and pharmacokinetic interactions between synthetic cathinones and other drugs of abuse. This pattern of polyconsumption can cause the potentiation of negative effects, and the dissociation between objective and subjective effects can increase the combined use of the drugs and the risk of toxicity leading to serious health problems. Certain animal studies indicate a higher vulnerability and effect of cathinones in females. In humans, most of the users are men and case reports show long-term psychotic symptoms after repeated use. CONCLUSIONS: The co-use of synthetic cathinones and the other drugs of abuse analyzed indicates potentiation of diverse effects including dependence and addiction, neurotoxicity, and impaired cognition and emotional responses. The motivations for and effects of synthetic cathinone use appear to be influenced by sex/gender. The long-term consequences of their use by adolescents and pregnant women deserve further investigation.


Assuntos
Estimulantes do Sistema Nervoso Central/metabolismo , Drogas Ilícitas/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Alcaloides/efeitos adversos , Alcaloides/metabolismo , Animais , Canabinoides/efeitos adversos , Canabinoides/metabolismo , Estimulantes do Sistema Nervoso Central/efeitos adversos , Cocaína/efeitos adversos , Cocaína/sangue , Interações Medicamentosas/fisiologia , Humanos , Drogas Ilícitas/farmacologia , Metanfetamina/efeitos adversos , Metanfetamina/análogos & derivados , Metanfetamina/metabolismo , Camundongos , Síndromes Neurotóxicas/diagnóstico , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/psicologia , Polimedicação , Ratos , Transtornos Relacionados ao Uso de Substâncias/diagnóstico , Transtornos Relacionados ao Uso de Substâncias/psicologia
20.
Behav Brain Res ; 366: 67-76, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-30902659

RESUMO

Chronic methamphetamine (MA) use can lead to increased symptoms of depression and anxiety during abstinence. Less is known about the specific brain regions that are altered following repeated MA that may be associated with these behavioral perturbations. Furthermore, MA has been reported to recruit and activate microglia in the brain, which may exacerbate stress-associated behavioral changes. In the present study, male and female mice were injected with MA (5 mg/kg) or saline once daily for 10 days, and during early withdrawal were assessed for alterations in immediate early gene (c-Fos) responses to a forced swim stressor. Chronic MA exposure increased floating and decreased swim time in the forced swim test in male and female mice tested 48 h after the final dose, indicating elevated depressive-like behavior. Furthermore, assessment of nest building, a measure of distress or despair-like behavior, revealed a sex-specific effect with only MA-treated females showing impairments. The c-Fos response to forced swim was attenuated by prior MA exposure in the central amygdala, CA3 hippocampal region, prefrontal cortex, and bed nucleus of the stria terminalis (BST). In the BST this attenuation occurred only in males. Neither the total number of microglia or activated microglia were altered by chronic MA exposure in regions examined. The primary findings indicate that chronic MA exposure attenuates activation of select stress-associated brain regions, a dysregulation that might contribute to alterations in mood-related behaviors.


Assuntos
Metanfetamina/metabolismo , Neurônios/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Animais , Ansiedade/metabolismo , Encéfalo/metabolismo , Região CA3 Hipocampal/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Corticosterona/farmacologia , Depressão/metabolismo , Teste de Esforço/métodos , Feminino , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Metanfetamina/efeitos adversos , Metanfetamina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Estresse Fisiológico/fisiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Natação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA