Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 611(7937): 769-779, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36385529

RESUMO

APOE4 is the strongest genetic risk factor for Alzheimer's disease1-3. However, the effects of APOE4 on the human brain are not fully understood, limiting opportunities to develop targeted therapeutics for individuals carrying APOE4 and other risk factors for Alzheimer's disease4-8. Here, to gain more comprehensive insights into the impact of APOE4 on the human brain, we performed single-cell transcriptomics profiling of post-mortem human brains from APOE4 carriers compared with non-carriers. This revealed that APOE4 is associated with widespread gene expression changes across all cell types of the human brain. Consistent with the biological function of APOE2-6, APOE4 significantly altered signalling pathways associated with cholesterol homeostasis and transport. Confirming these findings with histological and lipidomic analysis of the post-mortem human brain, induced pluripotent stem-cell-derived cells and targeted-replacement mice, we show that cholesterol is aberrantly deposited in oligodendrocytes-myelinating cells that are responsible for insulating and promoting the electrical activity of neurons. We show that altered cholesterol localization in the APOE4 brain coincides with reduced myelination. Pharmacologically facilitating cholesterol transport increases axonal myelination and improves learning and memory in APOE4 mice. We provide a single-cell atlas describing the transcriptional effects of APOE4 on the aging human brain and establish a functional link between APOE4, cholesterol, myelination and memory, offering therapeutic opportunities for Alzheimer's disease.


Assuntos
Apolipoproteína E4 , Encéfalo , Colesterol , Fibras Nervosas Mielinizadas , Oligodendroglia , Animais , Humanos , Camundongos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Colesterol/metabolismo , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Autopsia , Células-Tronco Pluripotentes Induzidas , Neurônios/metabolismo , Neurônios/patologia , Heterozigoto , Transporte Biológico , Homeostase , Análise de Célula Única , Memória , Envelhecimento/genética , Perfilação da Expressão Gênica , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia
2.
Glia ; 72(2): 452-469, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37969043

RESUMO

Genetic findings have highlighted key roles for microglia in the pathology of neurodegenerative conditions such as Alzheimer's disease (AD). A number of mutations in the microglial protein triggering receptor expressed on myeloid cells 2 (TREM2) have been associated with increased risk for developing AD, most notably the R47H/+ substitution. We employed gene editing and stem cell models to gain insight into the effects of the TREM2 R47H/+ mutation on human-induced pluripotent stem cell-derived microglia. We found transcriptional changes affecting numerous cellular processes, with R47H/+ cells exhibiting a proinflammatory gene expression signature. TREM2 R47H/+ also caused impairments in microglial movement and the uptake of multiple substrates, as well as rendering microglia hyperresponsive to inflammatory stimuli. We developed an in vitro laser-induced injury model in neuron-microglia cocultures, finding an impaired injury response by TREM2 R47H/+ microglia. Furthermore, mouse brains transplanted with TREM2 R47H/+ microglia exhibited reduced synaptic density, with upregulation of multiple complement cascade components in TREM2 R47H/+ microglia suggesting inappropriate synaptic pruning as one potential mechanism. These findings identify a number of potentially detrimental effects of the TREM2 R47H/+ mutation on microglial gene expression and function likely to underlie its association with AD.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Camundongos , Animais , Humanos , Microglia/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação/genética , Doença de Alzheimer/patologia , Sinapses/metabolismo , Encéfalo/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
3.
Mol Syst Biol ; 16(12): e9819, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33289969

RESUMO

Alzheimer's disease (AD) is characterized by the appearance of amyloid-ß plaques, neurofibrillary tangles, and inflammation in brain regions involved in memory. Using mass spectrometry, we have quantified the phosphoproteome of the CK-p25, 5XFAD, and Tau P301S mouse models of neurodegeneration. We identified a shared response involving Siglec-F which was upregulated on a subset of reactive microglia. The human paralog Siglec-8 was also upregulated on microglia in AD. Siglec-F and Siglec-8 were upregulated following microglial activation with interferon gamma (IFNγ) in BV-2 cell line and human stem cell-derived microglia models. Siglec-F overexpression activates an endocytic and pyroptotic inflammatory response in BV-2 cells, dependent on its sialic acid substrates and immunoreceptor tyrosine-based inhibition motif (ITIM) phosphorylation sites. Related human Siglecs induced a similar response in BV-2 cells. Collectively, our results point to an important role for mouse Siglec-F and human Siglec-8 in regulating microglial activation during neurodegeneration.


Assuntos
Inflamação/patologia , Microglia/metabolismo , Degeneração Neural/patologia , Fosfoproteínas/metabolismo , Proteômica , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Sequência de Aminoácidos , Animais , Anticorpos/metabolismo , Morte Celular , Linhagem Celular , Humanos , Inflamação/metabolismo , Interferon gama/metabolismo , Camundongos Transgênicos , Microglia/patologia , Degeneração Neural/metabolismo , Peptídeos/metabolismo , Fagocitose , Fosfotirosina/metabolismo , Proteoma/metabolismo , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/química , Transdução de Sinais , Regulação para Cima
4.
Mol Psychiatry ; 25(1): 148-167, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31391546

RESUMO

Alzheimer's disease is a devastating neurodegenerative disorder with no cure. Countless promising therapeutics have shown efficacy in rodent Alzheimer's disease models yet failed to benefit human patients. While hope remains that earlier intervention with existing therapeutics will improve outcomes, it is becoming increasingly clear that new approaches to understand and combat the pathophysiology of Alzheimer's disease are needed. Human induced pluripotent stem cell (iPSC) technologies have changed the face of preclinical research and iPSC-derived cell types are being utilized to study an array of human conditions, including neurodegenerative disease. All major brain cell types can now be differentiated from iPSCs, while increasingly complex co-culture systems are being developed to facilitate neuroscience research. Many cellular functions perturbed in Alzheimer's disease can be recapitulated using iPSC-derived cells in vitro, and co-culture platforms are beginning to yield insights into the complex interactions that occur between brain cell types during neurodegeneration. Further, iPSC-based systems and genome editing tools will be critical in understanding the roles of the numerous new genes and mutations found to modify Alzheimer's disease risk in the past decade. While still in their relative infancy, these developing iPSC-based technologies hold considerable promise to push forward efforts to combat Alzheimer's disease and other neurodegenerative disorders.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Doença de Alzheimer/genética , Encéfalo/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Humanos , Modelos Biológicos , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo
5.
J Neurosci ; 38(2): 322-334, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29167401

RESUMO

Acetaminophen (paracetamol) is a widely used analgesic and antipyretic drug with only incompletely understood mechanisms of action. Previous work, using models of acute nociceptive pain, indicated that analgesia by acetaminophen involves an indirect activation of CB1 receptors by the acetaminophen metabolite and endocannabinoid reuptake inhibitor AM 404. However, the contribution of the cannabinoid system to antihyperalgesia against inflammatory pain, the main indication of acetaminophen, and the precise site of the relevant CB1 receptors have remained elusive. Here, we analyzed acetaminophen analgesia in mice of either sex with inflammatory pain and found that acetaminophen exerted a dose-dependent antihyperalgesic action, which was mimicked by intrathecally injected AM 404. Both compounds lost their antihyperalgesic activity in CB1-/- mice, confirming the involvement of the cannabinoid system. Consistent with a mechanism downstream of proinflammatory prostaglandin formation, acetaminophen also reversed hyperalgesia induced by intrathecal prostaglandin E2 To distinguish between a peripheral/spinal and a supraspinal action, we administered acetaminophen and AM 404 to hoxB8-CB1-/- mice, which lack CB1 receptors from the peripheral nervous system and the spinal cord. These mice exhibited unchanged antihyperalgesia indicating a supraspinal site of action. Accordingly, local injection of the CB1 receptor antagonist rimonabant into the rostral ventromedial medulla blocked acetaminophen-induced antihyperalgesia, while local rostral ventromedial medulla injection of AM 404 reduced hyperalgesia in wild-type mice but not in CB1-/- mice. Our results indicate that the cannabinoid system contributes not only to acetaminophen analgesia against acute pain but also against inflammatory pain, and suggest that the relevant CB1 receptors reside in the rostral ventromedial medulla.SIGNIFICANCE STATEMENT Acetaminophen is a widely used analgesic drug with multiple but only incompletely understood mechanisms of action, including a facilitation of endogenous cannabinoid signaling via one of its metabolites. Our present data indicate that enhanced cannabinoid signaling is also responsible for the analgesic effects of acetaminophen against inflammatory pain. Local injections of the acetaminophen metabolite AM 404 and of cannabinoid receptor antagonists as well as data from tissue-specific CB1 receptor-deficient mice suggest the rostral ventromedial medulla as an important site of the cannabinoid-mediated analgesia by acetaminophen.


Assuntos
Acetaminofen/farmacologia , Analgésicos não Narcóticos/farmacologia , Bulbo/metabolismo , Dor/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Animais , Antagonistas de Receptores de Canabinoides/farmacologia , Feminino , Inflamação/metabolismo , Inflamação/fisiopatologia , Masculino , Bulbo/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dor/fisiopatologia , Receptor CB1 de Canabinoide/genética
6.
Neuroimage ; 150: 279-291, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28192273

RESUMO

Classical benzodiazepines, which are widely used as sedatives, anxiolytics and anticonvulsants, exert their therapeutic effects through interactions with heteropentameric GABAA receptors composed of two α, two ß and one γ2 subunit. Their high affinity binding site is located at the interface between the γ2 and the adjacent α subunit. The α-subunit gene family consists of six members and receptors can be homomeric or mixed with respect to the α-subunits. Previous work has suggested that benzodiazepine binding site ligands with selectivity for individual GABAA receptor subtypes, as defined by the benzodiazepine-binding α subunit, may have fewer side effects and may even be effective in diseases, such as schizophrenia, autism or chronic pain, that do not respond well to classical benzodiazepines. The distributions of the individual α subunits across the CNS have been extensively characterized. However, as GABAA receptors may contain two different α subunits, the distribution of the subunits does not necessarily reflect the distribution of receptor subtypes with respect to benzodiazepine pharmacology. In the present study, we have used in vivo [18F]flumazenil PET and in vitro [3H]flumazenil autoradiography in combination with GABAA receptor point-mutated mice to characterize the distribution of the two most prevalent GABAA receptor subtypes (α1 and α2) throughout the mouse brain. The results were in agreement with published in vitro data. High levels of α2-containing receptors were found in brain regions of the neuronal network of anxiety. The α1/α2 subunit combinations were predictable from the individual subunit levels. In additional experiments, we explored in vivo [18F]flumazenil PET to determine the degree of receptor occupancy at GABAA receptor subtypes following oral administration of diazepam. The dose to occupy 50% of sensitive receptors, independent of the receptor subtype(s), was 1-2mg/kg, in agreement with published data from ex vivo studies with wild type mice. In conclusion, we have resolved the quantitative distribution of α1- and α2-containing homomeric and mixed GABAA receptors in vivo at the millimeter scale and demonstrate that the regional drug receptor occupancy in vivo at these GABAA receptor subtypes can be determined by [18F]flumazenil PET. Such information should be valuable for drug development programs aiming for subtype-selective benzodiazepine site ligands for new therapeutic indications.


Assuntos
Encéfalo/metabolismo , Neuroimagem/métodos , Tomografia por Emissão de Pósitrons/métodos , Receptores de GABA-A/biossíntese , Animais , Autorradiografia , Diazepam/farmacologia , Flumazenil , Radioisótopos de Flúor , Moduladores GABAérgicos/farmacologia , Camundongos , Camundongos Mutantes , Compostos Radiofarmacêuticos , Receptores de GABA-A/análise
7.
Adv Mater ; 36(8): e2309225, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38018280

RESUMO

Neuroinflammation is a hallmark of neurodegenerative disorders including Alzheimer's disease (AD). Microglia, the brain's immune cells, express many of the AD-risk loci identified in genome wide association studies and present a promising target for anti-inflammatory RNA therapeutics but are difficult to transfect with current methods. Here, several lipid nanoparticle (LNP) formulations are examined, and a lead candidate that supports efficient RNA delivery in cultures of human stem cell-derived microglia-like cells (iMGLs) and animal models of neuroinflammation is identified. The lead microglia LNP (MG-LNP) formulation shows minimal toxicity and improves delivery efficiency to inflammatory iMGLs, suggesting a preference for delivery into activated microglia. Intraperitoneal injection of the MG-LNP formulation generates widespread expression of the delivered reporter construct in all organs, whereas local intracisternal injection directly into the cerebrospinal fluid leads to preferential expression in the brain. It is shown that LNP-mediated delivery of siRNA targeting the PU.1 transcription factor, a known AD-risk locus, successfully reduces PU.1 levels in iMGLs and reduces neuroinflammation in mice injected with LPS and in CK-p25 mice that mimic the chronic neuroinflammation seen in AD patients. The LNP formulation represents an effective RNA delivery vehicle when applied intrathecally and can be broadly utilized to test potential neuroinflammation-directed gene therapies.


Assuntos
Doença de Alzheimer , Nanopartículas , Humanos , Animais , Camundongos , RNA Interferente Pequeno/genética , Doenças Neuroinflamatórias , Estudo de Associação Genômica Ampla , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo
8.
J Immunol ; 186(3): 1769-80, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21191068

RESUMO

Chronic pain resulting from inflammatory and neuropathic disorders causes considerable economic and social burden. For a substantial proportion of patients, conventional drug treatments do not provide adequate pain relief. Consequently, novel approaches to pain management, involving alternative targets and new therapeutic modalities compatible with chronic use, are being sought. Nerve growth factor (NGF) is a major mediator of chronic pain. Clinical testing of NGF antagonists is ongoing, and clinical proof of concept has been established with a neutralizing mAb. Active immunization, with the goal of inducing therapeutically effective neutralizing autoreactive Abs, is recognized as a potential treatment option for chronic diseases. We have sought to determine if such a strategy could be applied to chronic pain by targeting NGF with a virus-like particle (VLP)-based vaccine. A vaccine comprising recombinant murine NGF conjugated to VLPs from the bacteriophage Qß (NGFQß) was produced. Immunization of mice with NGFQß induced anti-NGF-specific IgG Abs capable of neutralizing NGF. Titers could be sustained over 1 y by periodic immunization but declined in the absence of boosting. Vaccination with NGFQß substantially reduced hyperalgesia in collagen-induced arthritis or postinjection of zymosan A, two models of inflammatory pain. Long-term NGFQß immunization did not change sensory or sympathetic innervation patterns or induce cholinergic deficits in the forebrain, nor did it interfere with blood-brain barrier integrity. Thus, autovaccination targeting NGF using a VLP-based approach may represent a novel modality for the treatment of chronic pain.


Assuntos
Hiperalgesia/imunologia , Hiperalgesia/prevenção & controle , Mediadores da Inflamação/uso terapêutico , Fatores de Crescimento Neural/imunologia , Manejo da Dor , Dor/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Doença Aguda , Allolevivirus/imunologia , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/fisiologia , Anticorpos Antivirais/uso terapêutico , Linhagem Celular Tumoral , Doença Crônica , Avaliação Pré-Clínica de Medicamentos , Hiperalgesia/virologia , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Mediadores da Inflamação/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Fatores de Crescimento Neural/efeitos adversos , Fatores de Crescimento Neural/uso terapêutico , Testes de Neutralização , Dor/patologia , Ratos , Fatores de Tempo , Vacinas Conjugadas/efeitos adversos , Vacinas Conjugadas/imunologia , Vacinas Conjugadas/uso terapêutico , Vacinas de Partículas Semelhantes a Vírus/efeitos adversos , Vacinas de Partículas Semelhantes a Vírus/uso terapêutico
9.
J Exp Med ; 220(11)2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37642942

RESUMO

Pervasive neuroinflammation occurs in many neurodegenerative diseases, including Alzheimer's disease (AD). SPI1/PU.1 is a transcription factor located at a genome-wide significant AD-risk locus and its reduced expression is associated with delayed onset of AD. We analyzed single-cell transcriptomic datasets from microglia of human AD patients and found an enrichment of PU.1-binding motifs in the differentially expressed genes. In hippocampal tissues from transgenic mice with neurodegeneration, we found vastly increased genomic PU.1 binding. We then screened for PU.1 inhibitors using a PU.1 reporter cell line and discovered A11, a molecule with anti-inflammatory efficacy and nanomolar potency. A11 regulated genes putatively by recruiting a repressive complex containing MECP2, HDAC1, SIN3A, and DNMT3A to PU.1 motifs, thus representing a novel mechanism and class of molecules. In mouse models of AD, A11 ameliorated neuroinflammation, loss of neuronal integrity, AD pathology, and improved cognitive performance. This study uncovers a novel class of anti-inflammatory molecules with therapeutic potential for neurodegenerative disorders.


Assuntos
Doença de Alzheimer , Doenças Neuroinflamatórias , Animais , Camundongos , Humanos , Oncogenes , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Linhagem Celular , Modelos Animais de Doenças , Camundongos Transgênicos
10.
Sci Transl Med ; 13(583)2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33658354

RESUMO

The E4 allele of the apolipoprotein E gene (APOE) has been established as a genetic risk factor for many diseases including cardiovascular diseases and Alzheimer's disease (AD), yet its mechanism of action remains poorly understood. APOE is a lipid transport protein, and the dysregulation of lipids has recently emerged as a key feature of several neurodegenerative diseases including AD. However, it is unclear how APOE4 perturbs the intracellular lipid state. Here, we report that APOE4, but not APOE3, disrupted the cellular lipidomes of human induced pluripotent stem cell (iPSC)-derived astrocytes generated from fibroblasts of APOE4 or APOE3 carriers, and of yeast expressing human APOE isoforms. We combined lipidomics and unbiased genome-wide screens in yeast with functional and genetic characterization to demonstrate that human APOE4 induced altered lipid homeostasis. These changes resulted in increased unsaturation of fatty acids and accumulation of intracellular lipid droplets both in yeast and in APOE4-expressing human iPSC-derived astrocytes. We then identified genetic and chemical modulators of this lipid disruption. We showed that supplementation of the culture medium with choline (a soluble phospholipid precursor) restored the cellular lipidome to its basal state in APOE4-expressing human iPSC-derived astrocytes and in yeast expressing human APOE4 Our study illuminates key molecular disruptions in lipid metabolism that may contribute to the disease risk linked to the APOE4 genotype. Our study suggests that manipulating lipid metabolism could be a therapeutic approach to help alleviate the consequences of carrying the APOE4 allele.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Apolipoproteínas E , Homeostase , Humanos , Neuroglia
11.
Brain Res ; 1741: 146889, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32439345

RESUMO

Diminished synaptic inhibition in the superficial spinal dorsal horn contributes to exaggerated pain responses that accompany peripheral inflammation and neuropathy. α2GABAA receptors (α2GABAAR) constitute the most abundant GABAAR subtype at this site and are the targets of recently identified antihyperalgesic compounds. Surprisingly, hoxb8-α2-/- mice that lack α2GABAAR from the spinal cord and peripheral sensory neurons exhibit unaltered sensitivity to acute painful stimuli and develop normal inflammatory and neuropathic hyperalgesia. Here, we provide a comprehensive analysis of GABAergic neurotransmission, of behavioral phenotypes and of possible compensatory mechanisms in hoxb8-α2-/- mice. Our results confirm that hoxb8-α2-/- mice show significantly diminished GABAergic inhibitory postsynaptic currents (IPSCs) in the superficial dorsal horn but no hyperalgesic phenotype. We also confirm that the potentiation of dorsal horn GABAergic IPSCs by the α2-preferring GABAAR modulator HZ-166 is reduced in hoxb8-α2-/- mice and that hoxb8-α2-/- mice are resistant to the analgesic effects of HZ-166. Tonic GABAergic currents, glycinergic IPSCs, and sensory afferent-evoked EPSCs did not show significant changes in hoxb8-α2-/- mice rendering a compensatory up-regulation of other GABAAR subtypes or of glycine receptors unlikely. Although expression of serotonin and of the serotonin producing enzyme tryptophan hydroxylase (TPH2) was significantly increased in the dorsal horn of hoxb8-α2-/- mice, ablation of serotonergic terminals from the lumbar spinal cord failed to unmask a nociceptive phenotype. Our results are consistent with an important contribution of α2GABAAR to spinal nociceptive control but their ablation early in development appears to activate yet-to-be identified compensatory mechanisms that protect hoxb8-α2-/- mice from hyperalgesia.


Assuntos
Neurônios GABAérgicos/metabolismo , Hiperalgesia/metabolismo , Fenótipo , Receptores de GABA-A/deficiência , Medula Espinal/metabolismo , Transmissão Sináptica/fisiologia , Animais , Feminino , Células HEK293 , Humanos , Hiperalgesia/genética , Potenciais Pós-Sinápticos Inibidores/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Receptores de GABA-A/genética
12.
Neuropharmacology ; 143: 71-78, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30240781

RESUMO

Benzodiazepines (BDZ), which potentiate the action of GABA at four subtypes of GABAA receptors (α1, α2, α3, and α5GABAARs), are highly effective against anxiety disorders, but also cause severe side effects greatly limiting their clinical application. Both, preclinical studies in genetically engineered mice, and preclinical and clinical trials with subtype-selective compounds indicate that undesired effects can in principle be avoided by targeting specific GABAAR subtypes. While there is general consensus that activity at α1GABAARs should be avoided, controversy exists as to whether α2 or α3GABAARs need to be targeted for anxiolysis. While previous experiments in GABAAR point-mutated mice demonstrated a critical role of α2GABAARs, studies solely relying on pharmacological approaches suggested a dominant contribution of α3GABAARs. As most α1GABAAR-sparing BDZ site agonists discriminate little between α2 and α3GABAARs, these claims rest almost exclusively on a single compound, TP003, that has been reported to be a selective α3GABAAR modulator. Here, we have revisited the in vitro pharmacological profile of TP003 and, in addition, tested TP003 in GABAAR triple point-mutated mice, in which only either α1, α2, or α3GABAARs were left BDZ sensitive. These experiments revealed that TP003 behaves as a partial, rather non-selective BDZ site agonist in vitro that acts in vivo through α1, α2, and α3GABAARs (α5GABAAR-mediated effects were not tested). With respect to anxiolysis, our results support a critical contribution of α2GABAARs, but not of α3GABAARs. TP003 should therefore not be considered an α3GABAAR selective agent. Previously published studies using TP003 should be interpreted with caution.


Assuntos
Ansiolíticos/farmacologia , Ansiedade/tratamento farmacológico , Ansiedade/metabolismo , Agonistas de Receptores de GABA-A/farmacologia , Imidazóis/farmacologia , Piridinas/farmacologia , Animais , Ansiolíticos/química , Sítios de Ligação , Agonistas de Receptores de GABA-A/química , Células HEK293 , Humanos , Hipnóticos e Sedativos/química , Hipnóticos e Sedativos/farmacologia , Imidazóis/química , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos da Linhagem 129 , Camundongos Transgênicos , Estrutura Molecular , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Relaxantes Musculares Centrais/química , Relaxantes Musculares Centrais/farmacologia , Piridinas/química , Ratos , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Proteínas Recombinantes/metabolismo
13.
Nat Commun ; 9(1): 3230, 2018 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-30104684

RESUMO

Chronic itch is a highly debilitating condition affecting about 10% of the general population. The relay of itch signals is under tight control by inhibitory circuits of the spinal dorsal horn, which may offer a hitherto unexploited therapeutic opportunity. Here, we found that specific pharmacological targeting of inhibitory α2 and α3GABAA receptors reduces acute histaminergic and non-histaminergic itch in mice. Systemic treatment with an α2/α3GABAA receptor selective modulator alleviates also chronic itch in a mouse model of atopic dermatitis and in dogs sensitized to house dust mites, without inducing sedation, motor dysfunction, or loss of antipruritic activity after prolonged treatment. Transsynaptic circuit tracing, immunofluorescence, and electrophysiological experiments identify spinal α2 and α3GABAA receptors as likely molecular targets underlying the antipruritic effect. Our results indicate that drugs targeting α2 and α3GABAA receptors are well-suited to alleviate itch, including non-histaminergic chronic itch for which currently no approved treatment exists.


Assuntos
Prurido/tratamento farmacológico , Receptores de GABA-A/metabolismo , Medula Espinal/patologia , Animais , Doença Crônica , Modelos Animais de Doenças , Cães , Peptídeo Liberador de Gastrina/metabolismo , Células HEK293 , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/uso terapêutico , Humanos , Hidrocarbonetos Fluorados/farmacologia , Hidrocarbonetos Fluorados/uso terapêutico , Hipersensibilidade/complicações , Hipersensibilidade/tratamento farmacológico , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Camundongos Endogâmicos C57BL , Inibição Neural/efeitos dos fármacos , Mutação Puntual/genética , Prurido/complicações
14.
Neuropharmacology ; 108: 172-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27108932

RESUMO

Neuropathic pain is associated with impaired inhibitory control of spinal dorsal horn neurons, which are involved in processing pain signals. The metabotropic GABAB receptor is an important component of the inhibitory system and is highly expressed in primary nociceptors and intrinsic dorsal horn neurons to control their excitability. Activation of GABAB receptors with the orthosteric agonist baclofen effectively reliefs neuropathic pain but is associated with severe side effects that prevent its widespread application. The recently developed positive allosteric GABAB receptor modulators lack most of these side effects and are therefore promising drugs for the treatment of pain. Here we tested the high affinity positive allosteric modulator rac-BHFF for its ability to relief neuropathic pain induced by chronic constriction of the sciatic nerve in mice. rac-BHFF significantly increased the paw withdrawal threshold to mechanical stimulation in healthy mice, indicating an endogenous GABABergic tone regulating the sensitivity to mechanical stimuli. Surprisingly, rac-BHFF displayed no analgesic activity in neuropathic mice although GABAB receptor expression was not affected in the dorsal horn as shown by quantitative receptor autoradiography. However, activation of spinal GABAB receptors by intrathecal injection of baclofen reduced hyperalgesia and its analgesic effect was considerably potentiated by co-application of rac-BHFF. These results indicate that under conditions of neuropathic pain the GABAergic tone is too low to provide a basis for allosteric modulation of GABAB receptors. However, allosteric modulators would be well suited as an add-on to reduce the dose of baclofen required to achieve analgesia.


Assuntos
Analgesia/métodos , Baclofeno/administração & dosagem , Benzofuranos/administração & dosagem , Moduladores GABAérgicos/administração & dosagem , Neuralgia/tratamento farmacológico , Receptores de GABA-B/fisiologia , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Animais , Relação Dose-Resposta a Droga , Agonistas dos Receptores de GABA-B/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/metabolismo , Neuropatia Ciática/tratamento farmacológico , Neuropatia Ciática/metabolismo
15.
Neuropharmacology ; 109: 366-375, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27392635

RESUMO

Data from genetically modified mice suggest that benzodiazepine (BDZ)-site agonists with improved selectivity for α2-subtype GABAA receptors (α2GABAAR) are potentially useful for the treatment of neuropathic pain. Subtype-selective compounds available for preclinical tests in rodents support this concept but have not been approved for human use, hindering proof-of-concept studies in patients. We recently proposed that N-desmethyl clobazam (NDMC), the main metabolite of the licensed BDZ clobazam (CBZ), is responsible for most of the antihyperalgesia observed in mice after CBZ administration. In order to assess a potentially favorable pharmacological profile of NDMC, we analyzed differences in the GABAAR subtype specificity of CBZ, NDMC and diazepam (DZP) in recombinant receptors. DZP and CBZ potentiated sedating α1GABAARs and antihyperalgesic α2GABAARs with similar efficacies, whereas NDMC preferred α2GABAARs over α1GABAARs across a wide concentration range. In vivo, DZP and NDMC reduced neuropathic pain at doses between 3 and 30 mg/kg. At these doses, DZP had strong locomotor sedating effects while NDMC caused no or only weak sedation. Sedative effects of NDMC became apparent when the action of NDMC was restricted to α1GABAARs. However, when GABAAR point-mutated mice were studied that allow the analysis of antihyperalgesia and sedation in isolation, we found that, compared to DZP, NDMC had a significantly improved therapeutic window, consistent with its more favorable α2/α1 in vitro activity ratio. Given that NDMC should share the safety profile of its parent compound CBZ, it should be well-suited for proof-of-concept studies in human volunteers or patients.


Assuntos
Analgésicos não Narcóticos/metabolismo , Benzodiazepinas/metabolismo , Hiperalgesia/metabolismo , Receptores de GABA-A/metabolismo , Analgésicos não Narcóticos/uso terapêutico , Animais , Benzodiazepinas/uso terapêutico , Clobazam , Relação Dose-Resposta a Droga , Feminino , Células HEK293 , Humanos , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Ligação Proteica/fisiologia
16.
J Clin Invest ; 126(7): 2547-60, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27270175

RESUMO

Diminished inhibitory neurotransmission in the superficial dorsal horn of the spinal cord is thought to contribute to chronic pain. In inflammatory pain, reductions in synaptic inhibition occur partially through prostaglandin E2- (PGE2-) and PKA-dependent phosphorylation of a specific subtype of glycine receptors (GlyRs) that contain α3 subunits. Here, we demonstrated that 2,6-di-tert-butylphenol (2,6-DTBP), a nonanesthetic propofol derivative, reverses inflammation-mediated disinhibition through a specific interaction with heteromeric αßGlyRs containing phosphorylated α3 subunits. We expressed mutant GlyRs in HEK293T cells, and electrophysiological analyses of these receptors showed that 2,6-DTBP interacted with a conserved phenylalanine residue in the membrane-associated stretch between transmembrane regions 3 and 4 of the GlyR α3 subunit. In native murine spinal cord tissue, 2,6-DTBP modulated synaptic, presumably αß heteromeric, GlyRs only after priming with PGE2. This observation is consistent with results obtained from molecular modeling of the α-ß subunit interface and suggests that in α3ßGlyRs, the binding site is accessible to 2,6-DTBP only after PKA-dependent phosphorylation. In murine models of inflammatory pain, 2,6-DTBP reduced inflammatory hyperalgesia in an α3GlyR-dependent manner. Together, our data thus establish that selective potentiation of GlyR function is a promising strategy against chronic inflammatory pain and that, to our knowledge, 2,6-DTBP has a unique pharmacological profile that favors an interaction with GlyRs that have been primed by peripheral inflammation.


Assuntos
Hiperalgesia/metabolismo , Inflamação/metabolismo , Manejo da Dor/métodos , Receptores de Glicina/metabolismo , Medula Espinal/metabolismo , Sítio Alostérico , Animais , Feminino , Células HEK293 , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Moleculares , Neurônios , Dor , Fenóis/química , Fenilalanina/química , Fosforilação , Conformação Proteica , Proteínas Recombinantes/química
17.
Adv Pharmacol ; 73: 71-96, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25637438

RESUMO

GABAA receptors (GABA(A)Rs) and glycine receptors are key elements of the spinal control of nociception and pain. Compromised functioning of these two transmitter systems contributes to chronic pain states. Restoring their proper function through positive allosteric modulators should constitute a rational approach to the treatment of chronic pain syndromes involving diminished inhibitory spinal pain control. Although classical benzodiazepines (i.e., full agonists at the benzodiazepine binding site of GABA(A)Rs) potentiate synaptic inhibition in spinal pain controlling circuits, they lack clinically relevant analgesic activity in humans. Recent data obtained from experiments in GABA(A)R point-mutated mice suggests dose-limiting sedative effects of classical nonspecific benzodiazepines as the underlying cause. Experiments in genetically engineered mice resistant to the sedative effects of classical benzodiazepines and studies with novel less sedating benzodiazepines have indeed shown that profound antihyperalgesia can be obtained at least in preclinical pain models. Present evidence suggests that compounds with high intrinsic activity at α2-GABA(A)R and minimal agonistic activity at α1-GABA(A)R should possess relevant antihyperalgesic activity without causing unwanted sedation. On-going preclinical studies in genetically engineered mice and clinical trials with more selective benzodiazepine site agonists should soon provide additional insights into this emerging topic.


Assuntos
Analgésicos/farmacologia , Dor/tratamento farmacológico , Receptores de GABA-A/efeitos dos fármacos , Animais , Benzodiazepinas/farmacologia , Modelos Animais de Doenças , Agonistas de Receptores de GABA-A/farmacologia , Humanos , Camundongos , Dor/fisiopatologia , Mutação Puntual , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo
18.
Nat Commun ; 6: 6803, 2015 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-25865415

RESUMO

Agonists at the benzodiazepine-binding site of GABAA receptors (BDZs) enhance synaptic inhibition through four subtypes (α1, α2, α3 and α5) of GABAA receptors (GABAAR). When applied to the spinal cord, they alleviate pathological pain; however, insufficient efficacy after systemic administration and undesired effects preclude their use in routine pain therapy. Previous work suggested that subtype-selective drugs might allow separating desired antihyperalgesia from unwanted effects, but the lack of selective agents has hitherto prevented systematic analyses. Here we use four lines of triple GABAAR point-mutated mice, which express only one benzodiazepine-sensitive GABAAR subtype at a time, to show that targeting only α2GABAARs achieves strong antihyperalgesia and reduced side effects (that is, no sedation, motor impairment and tolerance development). Additional pharmacokinetic and pharmacodynamic analyses in these mice explain why clinically relevant antihyperalgesia cannot be achieved with nonselective BDZs. These findings should foster the development of innovative subtype-selective BDZs for novel indications such as chronic pain.


Assuntos
Benzodiazepinas/farmacologia , Moduladores GABAérgicos/farmacologia , Hiperalgesia/genética , Hipnóticos e Sedativos/farmacologia , Dor/genética , Mutação Puntual , Receptores de GABA-A/genética , Analgesia/métodos , Animais , Arginina/genética , Arginina/metabolismo , Diazepam/farmacologia , Expressão Gênica , Histidina/genética , Histidina/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Masculino , Camundongos , Midazolam/farmacologia , Dor/tratamento farmacológico , Dor/metabolismo , Dor/fisiopatologia , Manejo da Dor/métodos , Receptores de GABA-A/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia
19.
Neuropharmacology ; 98: 78-89, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25839900

RESUMO

The endocannabinoid system (ECS) comprises the cannabinoid receptors CB1 and CB2 and their endogenous arachidonic acid-derived agonists 2-arachidonoyl glycerol and anandamide, which play important neuromodulatory roles. Recently, a novel class of negative allosteric CB1 receptor peptide ligands, hemopressin-like peptides derived from alpha hemoglobin, has been described, with yet unknown origin and function in the CNS. Using monoclonal antibodies we now identified the localization of RVD-hemopressin (pepcan-12) and N-terminally extended peptide endocannabinoids (pepcans) in the CNS and determined their neuronal origin. Immunohistochemical analyses in rodents revealed distinctive and specific staining in major groups of noradrenergic neurons, including the locus coeruleus (LC), A1, A5 and A7 neurons, which appear to be major sites of production/release in the CNS. No staining was detected in dopaminergic neurons. Peptidergic axons were seen throughout the brain (notably hippocampus and cerebral cortex) and spinal cord, indicative of anterograde axonal transport of pepcans. Intriguingly, the chromaffin cells in the adrenal medulla were also strongly stained for pepcans. We found specific co-expression of pepcans with galanin, both in the LC and adrenal gland. Using LC-MS/MS, pepcan-12 was only detected in non-perfused brain (∼ 40 pmol/g), suggesting that in the CNS it is secreted and present in extracellular compartments. In adrenal glands, significantly more pepcan-12 (400-700 pmol/g) was measured in both non-perfused and perfused tissues. Thus, chromaffin cells may be a major production site of pepcan-12 found in blood. These data uncover important areas of peptide endocannabinoid occurrence with exclusive noradrenergic immunohistochemical staining, opening new doors to investigate their potential physiological function in the ECS. This article is part of the Special Issue entitled 'Fluorescent Tools in Neuropharmacology'.


Assuntos
Medula Suprarrenal/metabolismo , Sistema Nervoso Central/metabolismo , Endocanabinoides/química , Endocanabinoides/metabolismo , Peptídeos/metabolismo , Medula Suprarrenal/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Benzilaminas/farmacologia , Sistema Nervoso Central/crescimento & desenvolvimento , Cromatografia Líquida , Colchicina/farmacologia , Feminino , Hemoglobinas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibidores da Captação de Neurotransmissores/farmacologia , Fragmentos de Peptídeos , Ratos , Ratos Wistar , Espectrometria de Massas em Tandem , Moduladores de Tubulina/farmacologia
20.
Neuron ; 85(6): 1289-304, 2015 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-25789756

RESUMO

The gate control theory of pain proposes that inhibitory neurons of the spinal dorsal horn exert critical control over the relay of nociceptive signals to higher brain areas. Here we investigated how the glycinergic subpopulation of these neurons contributes to modality-specific pain and itch processing. We generated a GlyT2::Cre transgenic mouse line suitable for virus-mediated retrograde tracing studies and for spatially precise ablation, silencing, and activation of glycinergic neurons. We found that these neurons receive sensory input mainly from myelinated primary sensory neurons and that their local toxin-mediated ablation or silencing induces localized mechanical, heat, and cold hyperalgesia; spontaneous flinching behavior; and excessive licking and biting directed toward the corresponding skin territory. Conversely, local pharmacogenetic activation of the same neurons alleviated neuropathic hyperalgesia and chloroquine- and histamine-induced itch. These results establish glycinergic neurons of the spinal dorsal horn as key elements of an inhibitory pain and itch control circuit.


Assuntos
Rede Nervosa/fisiopatologia , Neurônios/citologia , Dor/fisiopatologia , Prurido/fisiopatologia , Corno Dorsal da Medula Espinal/citologia , Animais , Modelos Animais de Doenças , Glicina/metabolismo , Hiperalgesia/patologia , Camundongos , Camundongos Transgênicos , Rede Nervosa/metabolismo , Rede Nervosa/patologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/patologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Células do Corno Posterior/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA