Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biomol Struct Dyn ; : 1-21, 2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-38285669

RESUMO

Chronic pain is a common and debilitating condition with a huge social and economic burden worldwide. Currently, available drugs in clinics are not adequately effective and possess a variety of severe side effects leading to treatment withdrawal and poor quality of life. Recent findings highlight the potential role of autotaxin (ATX) as a promising novel target for chronic pain management, extending beyond its previously established involvement in arthritis and other neurological disorders, such as Alzheimer's disease. In the present study, we used a virtual screening strategy by targeting ATX against commercially available natural compounds (enamine- phenotypic screening library) to identify the potential inhibitors for the treatment of chronic pain. After initial identification using molecular docking based virtual screening, molecular mechanics (MM/GBSA), ADMET profiling and molecular dynamics simulation were performed to verify top hits. The computational screening resulted in the identification of fifteen top scoring structurally diverse hits that have free energy of binding (ΔG) values in the range of -25.792 (for compound Enamine_1850) to -74.722 Kcal/mol (for compound Enamine_1687). Moreover, the top-scoring hits have favourable ADME properties as calculated using in-silico algorithms. Additionally, the molecular dynamics simulation revealed the stable nature of protein-ligand interaction and provided information about amino acid residues involved in binding. This study led to the identification of potential autotaxin inhibitors with favourable pharmacokinetic properties. Identified hits may further be investigated for their safety and efficacy potential using in-vitro and in-vivo models of chronic pain.Communicated by Ramaswamy H. Sarma.

2.
Mol Neurobiol ; 61(8): 6089-6100, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38277118

RESUMO

Cold injury or frostbite is a common medical condition that causes serious clinical complications including sensory abnormalities and chronic pain ultimately affecting overall well-being. Opioids are the first-choice drug for the treatment of frostbite-induced chronic pain; however, their notable side effects, including sedation, motor incoordination, respiratory depression, and drug addiction, present substantial obstacle to their clinical utility. To address this challenge, we have exploited peripheral mu-opioid receptors as potential target for the treatment of frostbite-induced chronic pain. In this study, we investigated the effect of dermorphin [D-Arg2, Lys4] (1-4) amide (DALDA), a peripheral mu-opioid receptor agonist, on frostbite injury and hypersensitivity induced by deep freeze magnet exposure in rats. Animals with frostbite injury displayed significant hypersensitivity to mechanical, thermal, and cold stimuli which was significant ameliorated on treatment with different doses of DALDA (1, 3, and 10 mg/kg) and ibuprofen (100 mg/kg). Further, molecular biology investigations unveiled heightened oxido-nitrosative stress, coupled with a notable upregulation in the expression of TRP channels (TRPA1, TRPV1, and TRPM8), glial cell activation, and neuroinflammation (TNF-α, IL-1ß) in the sciatic nerve, dorsal root ganglion (DRG), and spinal cord of frostbite-injured rats. Treatment with DALDA leads to substantial reduction in TRP channels, microglial activation, and suppression of the inflammatory cascade in the ipsilateral L4-L5 DRG and spinal cord of rats. Overall, findings from the present study suggest that activation of peripheral mu-opioid receptors mitigates chronic pain in rats by modulating the expression of TRP channels and suppressing glial cell activation and neuroinflammation.


Assuntos
Congelamento das Extremidades , Microglia , Doenças Neuroinflamatórias , Peptídeos Opioides , Ratos Sprague-Dawley , Animais , Masculino , Ratos , Congelamento das Extremidades/tratamento farmacológico , Congelamento das Extremidades/complicações , Congelamento das Extremidades/patologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/patologia , Hiperalgesia/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Peptídeos Opioides/metabolismo , Peptídeos Opioides/farmacologia , Peptídeos Opioides/uso terapêutico , Dor/tratamento farmacológico , Dor/metabolismo , Dor/patologia , Receptores Opioides mu/metabolismo , Receptores Opioides mu/agonistas , Canais de Potencial de Receptor Transitório/metabolismo
3.
Cell Signal ; 115: 111028, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38176530

RESUMO

Frostbite, a debilitating condition, significantly affects the well-being of military veterans and high-altitude residents, causing severe clinical complications such as chronic pain that markedly impacts overall quality of life. There has been a notable increase in the development of pre-clinical models for studying frostbite injury, but their suitability for pain evaluation remains limited. The major hurdle in the development of novel therapeutics for the treatment of frostbite-induced chronic pain is the unavailability of well-established preclinical models. In this study, we employed deep-frozen magnets to induce frostbite injury and conducted validation for chronic pain through assessments of face, predictive, and mechanistic validity. Behavioral assays demonstrated that frostbite injury exhibited significant mechanical, thermal & cold hypersensitivity in rats. Further, molecular analysis indicated that frostbite injury triggered the activation of TRP channels (TRPA1, TRPV1 and TRPM8), microgliosis, and neuroinflammation in the dorsal root ganglion (DRG) and spinal cord of rats. Notably, NR2B protein expressions were significantly upregulated in the DRG of injured rats, while no changes were observed in spinal NR2B expressions. Furthermore, the administration of ibuprofen (25, 50, and 100 mg/kg, i.p.) resulted in a significant improvement in behavioral, biochemical, and molecular alterations in frostbite-injured rats. Overall, results suggested that established frostbite model effectively recapitulates face, pharmacological, and mechanistic validity, highlighting its potential for screening future treatment modalities and exploring the intricate mechanisms associated with frostbite-induced chronic pain.


Assuntos
Dor Crônica , Congelamento das Extremidades , Ratos , Animais , Dor Crônica/metabolismo , Hiperalgesia/metabolismo , Qualidade de Vida , Ratos Sprague-Dawley
4.
Neurotherapeutics ; 21(1): e00302, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38241153

RESUMO

Paclitaxel, a frequently utilized chemotherapeutic agent, often gives rise to severe and distressing sensory neuropathy in patients undergoing chemotherapy. Unfortunately, current therapeutics for chemotherapy-induced neuropathic pain (CINP) demonstrate limited effectiveness and are burdened with the potential for central side effects such as sedation, respiratory depression, cognitive impairment, and addiction, posing substantial clinical challenges. In light of these limitations, present study is designed to investigate the therapeutic potential of Dermorphin [D-Arg2, Lys4] (1-4) amide (DALDA), a preferential peripherally acting mu-opioid receptor agonist, in rat model of CINP. The primary objective was to assess the analgesic properties of DALDA and elucidate the underlying mechanisms governing its therapeutic activity. Our findings revealed that DALDA treatment significantly ameliorated paclitaxel-induced evoked and spontaneous ongoing pain in rats without causing drug addiction and other central side effects. Molecular analyses further unveiled that paclitaxel administration resulted in increased expression of TRP channels, NR2B, voltage-gated sodium channels (VGSCs) and neuroinflammatory markers in both the dorsal root ganglion (DRG) and the spinal cord (L4-L5 region) of rats. DALDA treatment significantly downregulated ion channels (TRPs, VGSCs) and NR2B expressions, concomitant with the inhibition of microglial activation, resulting in the suppression of oxido-nitrosative stress and neuroinflammatory cascade. Findings from the current study suggests that peripheral mu-opioid receptors may offer a potential target for the treatment of patients suffering from CINP, offering new avenues for improved pain relief while minimizing central side effects.


Assuntos
Antineoplásicos , Neuralgia , Peptídeos Opioides , Humanos , Ratos , Animais , Amidas/uso terapêutico , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Paclitaxel/toxicidade , Gânglios Espinais/metabolismo
5.
Int Immunopharmacol ; 124(Pt B): 110944, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37801968

RESUMO

Opioids are employed in the management of chemotherapy-induced neuropathic pain (CINP) when other pain management approaches have failed and proven ineffective. However, their use in CINP is generally considered as a second-line or adjunctive therapy owing to their central side effects and development of tolerance with their long-term usage. Targeting peripheral sites may offer several advantages over the conventional CNS-based approaches as peripheral targets modulate pain signals at their source, thereby relieving pain with higher specificity, efficacy and minimizing adverse effects associated with off-site CNS actions. Therefore, present study was designed with an aim to investigate the effect of loperamide, a peripherally acting mu-opioid receptor agonist, on paclitaxel-induced neuropathic pain in rats and elucidate its underlying mechanism. Loperamide treatment significantly attenuated mechanical, and cold hypersensitivity and produced significant place preference behaviour in neuropathic rats indicating its potential to treat both evoked and spontaneous pain. More importantly, loperamide treatment in naïve rats did not produce place preference to drug-paired chamber pointing towards its non-addictive analgesic potential. Further, molecular investigations revealed increased expression of ion channels such as TRPA1, TRPM8; voltage-gated sodium channels (VGSCs) and neuroinflammatory markers in the dorsal root ganglion (DRG) and lumbar (L4-L5) spinal cord of neuropathic rats, which was significantly downregulated upon loperamide treatment. These findings collectively suggest that activation of peripheral mu-opioid receptors contributes to the amelioration of both evoked and spontaneous pain in neuropathic rats by downregulating TRP channels and VGSCs along with suppression of oxido-nitrosative stress and neuroinflammatory cascade.


Assuntos
Antineoplásicos , Neuralgia , Ratos , Animais , Loperamida/uso terapêutico , Loperamida/farmacologia , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Analgésicos Opioides/uso terapêutico , Analgésicos Opioides/farmacologia , Antineoplásicos/efeitos adversos , Receptores Opioides
6.
Metab Brain Dis ; 37(8): 2853-2870, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36178640

RESUMO

Alpha-Synuclein (α-Syn) accumulation is central to the pathogenesis of Parkinson's disease (PD), hence the quest for finding potential therapeutics that may promote the α-Syn clearance is the need of the hour. To this, activation of the evolutionarily conserved protein and key regulator of the autophagy, 5'AMP-activated protein kinase (AMPK) is well-known to induce autophagy and subsequently the clearance of α-Syn aggregates. Alpha-mangostin (AM) a polyphenolic xanthone obtained from Garcinia Mangostana L. was previously reported to activate AMPK-dependent autophagy in various pre-clinical cancer models. However, no studies evidenced the effect of AM on AMPK-dependent autophagy activation in the PD. Therefore, the present study aimed to investigate the neuroprotective activity of AM in the chronic rotenone mouse model of PD against rotenone-induced α-Syn accumulation and to dissect molecular mechanisms underlying the observed neuroprotection. The findings showed that AM exerts neuroprotection against rotenone-induced α-Syn accumulation in the striatum and cortex by activating AMPK, upregulating autophagy (LC3II/I, Beclin-1), and lysosomal (TFEB) markers. Of note, an in-vitro study utilizing rat pheochromocytoma cells verified that AM conferred the neuroprotection only through AMPK activation, as the presence of inhibitors of AMPK (dorsomorphin) and autophagy (3-methyl adenine) failed to mitigate rotenone-induced α-Syn accumulation. Moreover, AM also counteracted rotenone-induced behavioral deficits, oxidative stress, and degeneration of nigro-striatal dopaminergic neurons. In conclusion, AM provided neuroprotection by ameliorating the rotenone-induced α-Syn accumulation through AMPK-dependent autophagy activation and it can be considered as a therapeutic agent which might be having a higher translational value in the treatment of PD.


Assuntos
Doença de Parkinson , Rotenona , Animais , Ratos , Camundongos , Rotenona/toxicidade , alfa-Sinucleína/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Proteínas Quinases Ativadas por AMP , Neuroproteção , Autofagia
7.
Metab Brain Dis ; 37(7): 2197-2211, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35239143

RESUMO

Schizophrenia (SZ) is a severe progressive neurodegenerative as well as disruptive behavior disorder affecting innumerable people throughout the world. The discovery of potential biomarkers in the clinical scenario would lead to the development of effective methods of diagnosis and would provide an understanding of the prognosis of the disease. Moreover, breakthrough inventions for the treatment and prevention of this mysterious disease could evolve as a result of a thorough understanding of the clinical biomarkers. In this review, we have discussed about specific biomarkers of SZ an emphasis has been laid to delineate (1) diagnostic biomarkers like neuroimmune biomarkers, metabolic biomarkers, oligodendrocyte biomarkers and biomarkers of negative and cognitive symptoms, (2) therapeutic biomarkers like various neurotransmitter systems and (3) prognostic biomarkers. All the biomarkers were evaluated in drug-naïve (at least for 4 weeks) patients in order to achieve a clear comparison between schizophrenic patients and healthy controls. Also, an attempt has been made to elucidate the potential genes which serve as predictors and tools for the determination of biomarkers and would ultimately help in the prevention and treatment of this deadly illness.


Assuntos
Esquizofrenia , Humanos , Esquizofrenia/tratamento farmacológico , Biomarcadores/metabolismo , Prognóstico , Neurotransmissores
8.
Inflammopharmacology ; 30(2): 549-563, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35243557

RESUMO

Chronic pain is among the most burdensome and devastating disorders affecting millions of people worldwide. Recent studies suggest the role of kinesin nanomotors in development and maintenance of chronic pain. KIF17 is a member of kinesin superfamily that binds to NR2B cargo system via mLin10 scaffolding protein and makes the NMDARs functional at cell surface. NMDA receptor activation is known to induce the central sensitization and excitotoxicity which can be recognized by the glial cells followed by the release of cytokine storm at spinal and supraspinal level leading to chronic pain. In this study, we have investigated the role of aurora kinase in the regulation of KIF17 and NR2B trafficking in the animal model of chronic inflammatory pain. Tozasertib (10, 20, and 40 mg/kg i.p.), a pan aurora kinase inhibitor, significantly attenuates acute inflammatory pain and suppresses enhanced pain hypersensitivity to heat, cold, and mechanical stimuli in CFA-injected rats. Molecular investigations suggest enhanced expression of KIF17/mLin10/NR2B in L4-L5 dorsal root ganglion (DRG) and spinal cord of CFA-injected rats which was significantly attenuated on treatment with tozasertib. Moreover, tozasertib treatment significantly attenuated CFA-induced oxido-nitrosative stress and macrophage activation in DRG and microglia activation in spinal cord of rats. Findings from the current study suggest that tozasertib mediates anti-nociceptive activity by inhibiting aurora kinase-mediated KIF17/mLin10/NR2B signaling.


Assuntos
Dor Crônica , Cinesinas , Piperazinas , Receptores de N-Metil-D-Aspartato , Animais , Dor Crônica/tratamento farmacológico , Dor Crônica/metabolismo , Humanos , Hiperalgesia/tratamento farmacológico , Piperazinas/farmacologia , Ratos , Receptor Cross-Talk , Receptores de N-Metil-D-Aspartato/metabolismo
9.
Life Sci ; 288: 120187, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-34856209

RESUMO

Chemotherapy-induced neuropathic pain (CINP) is among the most common clinical complications associated with the use of anti-cancer drugs. CINP occurs in nearly 68.1% of the cancer patients receiving chemotherapeutic drugs. Most of the clinically available analgesics are ineffective in the case of CINP patients as the pathological mechanisms involved with different chemotherapeutic drugs are distinct from each other. CINP triggers the somatosensory nervous system, increases the neuronal firing and activation of nociceptive mediators including transient receptor protein vanilloid 1 (TRPV1). TRPV1 is widely present in the peripheral nociceptive nerve cells and it has been reported that the higher expression of TRPV1 in DRGs serves a critical role in the potentiation of CINP. The therapeutic glory of TRPV1 is well recognized in clinics which gives a promising insight into the treatment of pain. But the adverse effects associated with some of the antagonists directed the scientists towards RNA interference (RNAi), a tool to silence gene expression. Thus, ongoing research is focused on developing small interfering RNA (siRNA)-based therapeutics targeting TRPV1. In this review, we have discussed the involvement of TRPV1 in the nociceptive signaling associated with CINP and targeting this nociceptor, using siRNA will potentially arm us with effective therapeutic interventions for the clinical management of CINP.


Assuntos
Antineoplásicos/efeitos adversos , Neoplasias/tratamento farmacológico , Neuralgia/terapia , RNA Interferente Pequeno/administração & dosagem , Canais de Cátion TRPV/antagonistas & inibidores , Animais , Humanos , Neoplasias/patologia , Neuralgia/induzido quimicamente , Neuralgia/patologia , RNA Interferente Pequeno/genética , Transdução de Sinais , Canais de Cátion TRPV/genética
10.
ACS Chem Neurosci ; 12(22): 4195-4208, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34723483

RESUMO

Bone cancer pain (BCP) is a distinct pain state showing characteristics of both neuropathic and inflammatory pain. On average, almost 46% of cancer patients exhibit BCP with numbers flaring up to as high as 76% for terminally ill patients. Patients suffering from BCP experience a compromised quality of life, and the unavailability of effective therapeutics makes this a more devastating condition. In every individual cancer patient, the pain is driven by different mechanisms at different sites. The mechanisms behind the manifestation of BCP are very complex and poorly understood, which creates a substantial barrier to drug development. Nevertheless, some of the key mechanisms involved have been identified and are being explored further to develop targeted molecules. Developing a multitarget approach might be beneficial in this case as the underlying mechanism is not fixed and usually a number of these pathways are simultaneously dysregulated. In this review, we have discussed the role of recently identified novel modulators and mechanisms involved in the development of BCP. They include ion channels and receptors involved in sensing alteration of temperature and acidic microenvironment, immune system activation, sodium channels, endothelins, protease-activated receptors, neurotrophins, motor proteins mediated trafficking of glutamate receptor, and some bone-specific mechanisms. Apart from this, we have also discussed some of the novel approaches under preclinical and clinical development for the treatment of bone cancer pain.


Assuntos
Neoplasias Ósseas , Dor do Câncer , Animais , Neoplasias Ósseas/complicações , Neoplasias Ósseas/tratamento farmacológico , Dor do Câncer/tratamento farmacológico , Modelos Animais de Doenças , Humanos , Dor/tratamento farmacológico , Dor/etiologia , Qualidade de Vida , Microambiente Tumoral
11.
Metab Brain Dis ; 36(7): 2141-2153, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34386880

RESUMO

Alcohol consumption for a longer period of time is linked with neuronal damage and an increase in inflammatory signaling resulting in cell death and dementia. Natural compounds are the focus of research due to their high efficacy and good safety profile. Here we have investigated the effect of chronic epigallocatechin-3-gallate (EGCG) administration against the alcohol-induced cognitive deficit rats. Male Wistar rats were exposed to the 12% ethanol (10 g/kg; oral gavage) for ten weeks and treated with EGCG (25, 50, and 100 mg/kg) for the same duration. Ethanol exposure led to the impaired spatial memory and learning in rats assessed using the Morris water maze and elevated plus-maze test. Further, we assessed the role of EGCG in mitigating the oxidative stress, neuroinflammatory and cell death signaling associated markers. Co-administration with EGCG significantly prevented all the behavioral, biochemical and molecular alterations in the different brain regions of ethanol-treated rats in a dose-dependent manner. EGCG suppressed the acetylcholinesterase activity, increased oxidative-nitrosative stress, cytokines (TNF-alpha and IL-1beta), NF-kappa ß and caspase-3 levels in both the cortex and hippocampus of ethanol-treated rats. Our preliminary study demonstrated that EGCG improves the oxido-nitrosative stress, inflammation, and cell death signaling associated with ethanol-induced cognitive dysfunction. This suggests the potential role of EGCG in mitigating the cognitive deficits associated with chronic alcohol consumption.


Assuntos
Acetilcolinesterase , Disfunção Cognitiva , Acetilcolinesterase/metabolismo , Animais , Catequina/análogos & derivados , Morte Celular , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Etanol/toxicidade , Hipocampo/metabolismo , Masculino , Aprendizagem em Labirinto , Estresse Oxidativo , Ratos , Ratos Wistar
12.
ACS Chem Neurosci ; 12(16): 2956-2963, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34324307

RESUMO

Chronic pain is among the most prevalent burdensome disorders worldwide. The N-methyl-d-aspartate (NMDA) receptor system plays a critical role in central sensitization, a primary feature of chronic pain. Despite the proven efficacy of exogenous ligands to this receptor system in preclinical studies, evidence for the clinical efficacy of NMDA antagonists for the treatment of chronic pain is weak. Researchers are studying alternate approaches, rather than direct inhibition of the NMDA receptors in pain processing neurons. This indirect approach utilizes the modulation of molecular switches that regulates the synthesis, maturation, and transport of receptors from cellular organelles to the synaptic membrane. Kinesins are nanomotors that anterogradely transport the cargo using microtubule tracks across the neurons. Various members of the kinesin family, including KIF17, KIF11, KIF5b, and KIF21a, regulate the intracellular transport of NMDA receptors. Pharmacological targeting of these ATP-driven nanomotors could be a useful tool for manipulating the NMDAR functioning. It could provide the potential for the development of a novel strategy for the management of chronic pain.


Assuntos
Dor Crônica , Cinesinas , Dor Crônica/tratamento farmacológico , Humanos , Cinesinas/metabolismo , Neurônios/metabolismo , Transporte Proteico , Receptores de N-Metil-D-Aspartato/metabolismo
13.
Eur J Pharmacol ; 889: 173619, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33011242

RESUMO

Neuropathic pain is a critical burdensome problem due to the complex interplay of several pathological mechanisms and lack of availability of effective therapeutic interventions. The available therapeutic options are associated with a variety of limitations, including severe side effects, and unmet medical needs, warranting further research to identify and validate potential targets. Adenosine receptors system is a widely studied target, which evidently was successful in alleviation of neuropathic pain in several experimental paradigms, and researchers are putting efforts in building its clinical roadmap. The adenosine receptors act by different mechanisms and targeting adenosine receptors for neuropathic pain includes several important pathways such as p38-mitogen-activated protein kinases (MAPK), extracellular signal-regulated kinases (ERK), brain-derived neurotrophic factor (BDNF) signalling, γ-aminobutyric acid (GABA) as well as the ion channel modulations. Various studies have also shown the relevance of targeting adenosine receptors in chemotherapy-induced neuropathic pain and diabetic neuropathy. Several drugs acting on adenosine receptors have undergone clinical trials for management of neuropathic pain, whereas many other drugs are yet to be studied to find a potential anti-nociceptive agent. In this review, we have discussed the roadmap of adenosine receptors as a potential target for the treatment of neuropathic pain.


Assuntos
Agonistas Adrenérgicos/metabolismo , Analgésicos/metabolismo , Neuralgia/metabolismo , Receptores Purinérgicos P1/metabolismo , Transdução de Sinais/fisiologia , Agonistas Adrenérgicos/administração & dosagem , Analgésicos/administração & dosagem , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Humanos , Neuralgia/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
ACS Chem Neurosci ; 11(9): 1210-1217, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32243128

RESUMO

Alcohol addiction is one of the highly prevalent neurological disorders and a major threat to public health in the 21st century. Alcohol addiction affects people from all age groups and often leads to other serious comorbidities. The pathophysiology of alcohol addiction involves imbalance between the excitatory and inhibitory neurotransmitters in the brain. These changes occur in various regions of the brain including reward circuit such as the ventral tegmental area (VTA), nucleus accumbens (NAc), and prefrontal cortex. In this review, we have discussed several neurochemical circuitries which get manipulated and maladapted during alcohol addiction. To date there is no effective therapeutic intervention in clinics devoid of side effects that can successfully treat the patients suffering from alcohol addiction. Understanding the neurobiological intricacies of alcohol addiction is critical for the development of novel anti-addiction therapeutics. Apart from this, we have also discussed the recent therapeutic milestones for the management of alcohol addiction including vasopressin receptors, corticotrophin-releasing factor, GABA receptors, glucocorticoid receptors, brain stimulation and mindfulness-oriented recovery enhancement.


Assuntos
Alcoolismo , Comportamento Aditivo , Alcoolismo/tratamento farmacológico , Humanos , Núcleo Accumbens , Recompensa , Área Tegmentar Ventral
15.
ACS Chem Neurosci ; 11(6): 830-839, 2020 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-32083459

RESUMO

The opioid crisis is a major threat of the 21st century, with a remarkable juxtaposition of use and abuse. Opioids are the most potent and efficacious class of analgesics, but despite their proven therapeutic efficacy, they have recently been degraded to third-line therapy for the management of chronic pain in clinics. The reason behind this is the development of potential side effects and tolerance after repeated dosing. Opioid tolerance is the major limiting factor leading to the withdrawal of treatment, severe side effects due to dose escalation, and sometimes even death of the patients. Every day more than 90 people die due to opioids overdose in America, and a similar trend has been seen across the globe. Over the past two decades, researchers have been trying to dissect the neurobiological mechanism of opioid tolerance. Research on opioid tolerance shifted toward central nervous system-based adaptations because tolerance is much more than just a cellular phenomenon. Thus, neurobiological adaptations associated with opioid tolerance are important to understand in order to find newer pain therapeutics. These adaptations are associated with alterations in ascending and descending pain pathways, reward circuitry modulations, receptor desensitization and down-regulation, receptor internalization, heterodimerization, and altered epigenetic regulation. The present Review is focused on novel circuitries associated with opioid tolerance in different areas of the brain, such as periaqueductal gray, rostral ventromedial medulla, dorsal raphe nucleus, ventral tegmental area, and nucleus accumbens. Understanding the neurobiological modulations associated with chronic opioid exposure and tolerance will pave the way for the development of novel pharmacological tools for safer and better management of chronic pain in patients.


Assuntos
Analgésicos Opioides , Epigênese Genética , Analgésicos/uso terapêutico , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Tolerância a Medicamentos , Epigênese Genética/efeitos dos fármacos , Humanos , Dor/tratamento farmacológico
16.
Neurosci Lett ; 716: 134652, 2020 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-31778768

RESUMO

Parkinson's disease (PD) is a progressive, late-onset, and degenerative disorder that affects the central nervous system with an unknown etiology. Due to its incredible complexity in disease nature, many of the existing treatment approaches show a vain recovery in Parkinson's patients. Therefore, an in search of disease-modifying therapeutics for an effective recovery is essential. Alpha mangostin is an important polyphenolic xanthone reported for its neuroprotective effect against rotenone-induced α-synuclein aggregation and loss of tyrosine hydroxylase positive (TH+)-neurons in SH-SY5Y cells. Hence, the current study aims to test its protective effect in managing the in-vivo rat model of PD. To justify this aim, adult male Sprague Dawley rats (250 ± 20 g) were subjected to chronic treatment of rotenone (2 mg/kg/day, s.c.) for 21 days. In parallel alpha mangostin treatment (10 mg/kg, i.p) was administered along with rotenone for 21 days. Chronic rotenone treatment for 21 days increased lipid peroxidation, nitrite concentration, and decreased glutathione levels. Further, depletion of TH+-dopaminergic neuron expression in substantia nigra pars compacta (SNc), and the development of motor and behavioral deficits in rotenone treated animals like cognitive impairment, muscle incoordination, and neuromuscular weakness were observed. Moreover, western blot studies ascertained the reduced normal alpha-synuclein levels and increased phosphorylated α-synuclein levels in comparison to the vehicle-treated group. Treatment with alpha mangostin significantly restored the locomotor activity, memory deficits, and improved the levels of antioxidant enzymes. It also significantly reduced the levels of phosphorylated α-synuclein which in turn gave protection against TH+-dopaminergic neuronal loss in SNc, suggesting it's anti-oxidant and anti-aggregatory potential against α-synuclein. In conclusion through our current results, we could suggest that alpha mangostin has a potential neuroprotective effect against rotenone-induced PD and might be used as a neuroprotective agent. Further mechanistic studies on preclinical and clinical levels are required to be conducted with alpha mangostin to avail and foresee it as a potential agent in the treatment and management of PD.


Assuntos
Encéfalo/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Transtornos Parkinsonianos/patologia , Xantonas/farmacologia , Animais , Encéfalo/patologia , Neurônios Dopaminérgicos/patologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Transtornos Parkinsonianos/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Rotenona/toxicidade , Desacopladores/toxicidade
17.
ACS Chem Neurosci ; 10(9): 3914-3926, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31385687

RESUMO

Parkinson's disease (PD) is the second-most common neurodegenerative disorder, neuropathologically characterized by the aggregation of misfolded α-synuclein (α-syn) protein, which appears to be central to the onset and progression of PD pathology. Evidence from pioneering studies has highly advocated the existence of impaired autophagy pathways in the brains of PD patients. Autophagy is an evolutionarily conserved, homeostatic mechanism for minimizing abnormal protein aggregates and facilitating organelle turnover. Any aberration in constitutive autophagy activity results in the aggregation of misfolded α-syn, which, in turn, may further inhibit their own degradation-leading to a vicious cycle of neuronal death. Despite the plethora of available literature, there are still lacunas existing in our understanding of the exact cellular interplay between autophagy impairment and α-syn accumulation-mediated neurotoxicity. In this context, clearance of aggregated α-syn via up-regulation of the autophagy-lysosomal pathway could provide a pharmacologically viable approach to the treatment of PD. The present Review highlights the basics of autophagy and detrimental cross-talk between α-syn and chaperone-mediated autophagy, and α-syn and macroautophagy. It also depicts the interaction between α-syn and novel targets, LRRK2 and mTOR, followed by the role of autophagy in PD from a therapeutic perspective. More importantly, it further updates the reader's understanding of various newer therapeutic avenues that may accomplish disease modification via promoting clearance of toxic α-syn through activation of autophagy.


Assuntos
Autofagia/fisiologia , Lisossomos/metabolismo , Doença de Parkinson/metabolismo , Animais , Humanos , Lisossomos/patologia , Doença de Parkinson/patologia , alfa-Sinucleína/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...