Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain ; 146(10): 4033-4039, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37249190

RESUMO

Melzak and Wall's gate control theory proposed that innocuous input into the dorsal horn of the spinal cord represses pain-inducing nociceptive input. Here we show that input from proprioceptive parvalbumin-expressing sensory neurons tonically represses nociceptor activation within dorsal root ganglia. Deletion of parvalbumin-positive sensory neurons leads to enhanced nociceptor activity measured with GCaMP3, increased input into wide dynamic range neurons of the spinal cord and increased acute and spontaneous pain behaviour, as well as potentiated innocuous sensation. Parvalbumin-positive sensory neurons express the enzymes and transporters necessary to produce vesicular GABA that is known to be released from depolarized somata. These observations support the view that gate control mechanisms occur peripherally within dorsal root ganglia.


Assuntos
Parvalbuminas , Células Receptoras Sensoriais , Humanos , Transmissão Sináptica , Dor , Gânglios Espinais
3.
Neuron ; 109(9): 1497-1512.e6, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33823138

RESUMO

Deletion of SCN9A encoding the voltage-gated sodium channel NaV1.7 in humans leads to profound pain insensitivity and anosmia. Conditional deletion of NaV1.7 in sensory neurons of mice also abolishes pain, suggesting that the locus of analgesia is the nociceptor. Here we demonstrate, using in vivo calcium imaging and extracellular recording, that NaV1.7 knockout mice have essentially normal nociceptor activity. However, synaptic transmission from nociceptor central terminals in the spinal cord is greatly reduced by an opioid-dependent mechanism. Analgesia is also reversed substantially by central but not peripheral application of opioid antagonists. In contrast, the lack of neurotransmitter release from olfactory sensory neurons is opioid independent. Male and female humans with NaV1.7-null mutations show naloxone-reversible analgesia. Thus, inhibition of neurotransmitter release is the principal mechanism of anosmia and analgesia in mouse and human Nav1.7-null mutants.


Assuntos
Analgesia , Canal de Sódio Disparado por Voltagem NAV1.7/deficiência , Neurônios Receptores Olfatórios/metabolismo , Dor/genética , Transmissão Sináptica/fisiologia , Adulto , Animais , Feminino , Humanos , Masculino , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Transtornos do Olfato/congênito , Transtornos do Olfato/genética
4.
Brain ; 144(6): 1711-1726, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-33693512

RESUMO

Patients with neuropathic pain often experience innocuous cooling as excruciating pain. The cell and molecular basis of this cold allodynia is little understood. We used in vivo calcium imaging of sensory ganglia to investigate how the activity of peripheral cold-sensing neurons was altered in three mouse models of neuropathic pain: oxaliplatin-induced neuropathy, partial sciatic nerve ligation, and ciguatera poisoning. In control mice, cold-sensing neurons were few in number and small in size. In neuropathic animals with cold allodynia, a set of normally silent large diameter neurons became sensitive to cooling. Many of these silent cold-sensing neurons responded to noxious mechanical stimuli and expressed the nociceptor markers Nav1.8 and CGRPα. Ablating neurons expressing Nav1.8 resulted in diminished cold allodynia. The silent cold-sensing neurons could also be activated by cooling in control mice through blockade of Kv1 voltage-gated potassium channels. Thus, silent cold-sensing neurons are unmasked in diverse neuropathic pain states and cold allodynia results from peripheral sensitization caused by altered nociceptor excitability.


Assuntos
Temperatura Baixa/efeitos adversos , Hiperalgesia/metabolismo , Neuralgia/metabolismo , Neurônios/metabolismo , Nociceptores/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/metabolismo , Superfamília Shaker de Canais de Potássio/metabolismo , Sensação Térmica/fisiologia
5.
Sci Adv ; 6(8): eaax4568, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32128393

RESUMO

Expression of the voltage-gated sodium channel NaV1.7 in sensory neurons is required for pain sensation. We examined the role of NaV1.7 in the dorsal horn of the spinal cord using an epitope-tagged NaV1.7 knock-in mouse. Immuno-electron microscopy showed the presence of NaV1.7 in dendrites of superficial dorsal horn neurons, despite the absence of mRNA. Rhizotomy of L5 afferent nerves lowered the levels of NaV1.7 in the dorsal horn. Peripheral nervous system-specific NaV1.7 null mutant mice showed central deficits, with lamina II dorsal horn tonic firing neurons more than halved and single spiking neurons more than doubled. NaV1.7 blocker PF05089771 diminished excitability in dorsal horn neurons but had no effect on NaV1.7 null mutant mice. These data demonstrate an unsuspected functional role of primary afferent neuron-generated NaV1.7 in dorsal horn neurons and an expression pattern that would not be predicted by transcriptomic analysis.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/genética , Células do Corno Posterior/fisiologia , Células Receptoras Sensoriais/fisiologia , Animais , Fenômenos Eletrofisiológicos , Potenciais Pós-Sinápticos Excitadores , Expressão Gênica , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/ultraestrutura , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/ultraestrutura , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia
6.
Wellcome Open Res ; 3: 101, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30271888

RESUMO

Background: Functional deletion of the Scn9a (sodium voltage-gated channel alpha subunit 9) gene encoding sodium channel Nav1.7 makes humans and mice pain-free. Opioid signalling contributes to this analgesic state. We have used pharmacological and genetic approaches to identify the opioid receptors involved in this form of analgesia. We also examined the regulation of proenkephalin expression by the transcription factor Nfat5 that binds upstream of the Penk gene. Methods: We used specific µ-, δ- and κ-opioid receptor antagonists alone or in combination to examine which opioid receptors were necessary for Nav1.7 loss-associated analgesia in mouse behavioural assays of thermal pain. We also used µ- and δ-opioid receptor null mutant mice alone and in combination in behavioural assays to examine the role of these receptors in Nav1.7 knockouts pain free phenotype. Finally, we examined the levels of Penk mRNA in Nfat5-null mutant mice, as this transcription factor binds to consensus sequences upstream of the Penk gene. Results: The pharmacological block or deletion of both µ- and δ-opioid receptors was required to abolish Nav1.7-null opioid-related analgesia. κ-opioid receptor antagonists were without effect. Enkephalins encoded by the Penk gene are upregulated in Nav1.7 nulls. Deleting Nfat5, a transcription factor with binding motifs upstream of Penk, induces the same level of enkephalin mRNA expression as found in Nav1 .7 nulls, but without consequent analgesia. These data confirm that a combination of events linked to Scn9a gene loss is required for analgesia. Higher levels of endogenous enkephalins, potentiated opioid receptors, diminished electrical excitability and loss of neurotransmitter release together contribute to the analgesic phenotype found in Nav1.7-null mouse and human mutants. Conclusions: These observations help explain the failure of Nav1.7 channel blockers alone to produce analgesia and suggest new routes for analgesic drug development.

7.
Wellcome Open Res ; 3: 78, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30079380

RESUMO

Background: Sensory neurons play an essential role in almost all pain conditions, and have recently been classified into distinct subsets on the basis of their transcriptomes. Here we have analysed alterations in dorsal root ganglia (DRG) gene expression using microarrays in mouse models related to human chronic pain. Methods: Six different pain models were studied in male C57BL/6J mice: (1) bone cancer pain using cancer cell injection in the intramedullary space of the femur; (2) neuropathic pain using partial sciatic nerve ligation; (3) osteoarthritis pain using mechanical joint loading; (4) chemotherapy-induced pain with oxaliplatin; (5) chronic muscle pain using hyperalgesic priming; and (6) inflammatory pain using intraplantar complete Freund's adjuvant. Microarray analyses were performed using RNA isolated from dorsal root ganglia and compared to sham/vehicle treated controls. Results: Differentially expressed genes (DEGs) were identified. Known and previously unreported genes were found to be dysregulated in each pain model. The transcriptomic profiles for each model were compared and expression profiles of DEGs within subsets of DRG neuronal populations were analysed to determine whether specific neuronal subsets could be linked to each of the pain models.  Conclusions: Each pain model exhibits a unique set of altered transcripts implying distinct cellular responses to different painful stimuli. No simple direct link between genetically distinct sets of neurons and particular pain models could be discerned.

8.
Sci Signal ; 11(535)2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29921656

RESUMO

Mechanically activated, slowly adapting currents in sensory neurons have been linked to noxious mechanosensation. The conotoxin NMB-1 (noxious mechanosensation blocker-1) blocks such currents and inhibits mechanical pain. Using a biotinylated form of NMB-1 in mass spectrometry analysis, we identified 67 binding proteins in sensory neurons and a sensory neuron-derived cell line, of which the top candidate was annexin A6, a membrane-associated calcium-binding protein. Annexin A6-deficient mice showed increased sensitivity to mechanical stimuli. Sensory neurons from these mice showed increased activity of the cation channel Piezo2, which mediates a rapidly adapting mechano-gated current linked to proprioception and touch, and a decrease in mechanically activated, slowly adapting currents. Conversely, overexpression of annexin A6 in sensory neurons inhibited rapidly adapting currents that were partially mediated by Piezo2. Furthermore, overexpression of annexin A6 in sensory neurons attenuated mechanical pain in a mouse model of osteoarthritis, a disease in which mechanically evoked pain is particularly problematic. These data suggest that annexin A6 can be exploited to inhibit chronic mechanical pain.


Assuntos
Anexina A6/fisiologia , Conotoxinas/metabolismo , Mecanotransdução Celular , Dor/prevenção & controle , Fragmentos de Peptídeos/metabolismo , Células Receptoras Sensoriais/fisiologia , Animais , Artrite Experimental/etiologia , Artrite Experimental/fisiopatologia , Biotinilação , Células Cultivadas , Canais Iônicos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoartrite/etiologia , Osteoartrite/fisiopatologia , Dor/metabolismo , Dor/patologia
9.
Brain ; 141(4): 1028-1039, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29394316

RESUMO

Many studies support the pro-nociceptive role of brain-derived neurotrophin factor (BDNF) in pain processes in the peripheral and central nervous system. We have previously shown that nociceptor-derived BDNF is involved in inflammatory pain. Microglial-derived BDNF has also been shown to be involved in neuropathic pain. However, the distinct contribution of primary afferent-derived BNDF to chronic pain processing remains undetermined. In this study, we used Avil-CreERT2 mice to delete Bdnf from all adult peripheral sensory neurons. Conditional BDNF knockouts were healthy with no sensory neuron loss. Behavioural assays and in vivo electrophysiology indicated that spinal excitability was normal. Following formalin inflammation or neuropathy with a modified Chung model, we observed normal development of acute pain behaviour, but a deficit in second phase formalin-induced nocifensive responses and a reversal of neuropathy-induced mechanical hypersensitivity during the later chronic pain phase in conditional BDNF knockout mice. In contrast, we observed normal development of acute and chronic neuropathic pain in the Seltzer model, indicating differences in the contribution of BDNF to distinct models of neuropathy. We further used a model of hyperalgesic priming to examine the contribution of primary afferent-derived BDNF in the transition from acute to chronic pain, and found that primed BDNF knockout mice do not develop prolonged mechanical hypersensitivity to an inflammatory insult. Our data suggest that BDNF derived from sensory neurons plays a critical role in mediating the transition from acute to chronic pain.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Dor Crônica/patologia , Gânglios Espinais/patologia , Células Receptoras Sensoriais/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Carragenina/toxicidade , Dor Crônica/induzido quimicamente , Modelos Animais de Doenças , Feminino , Formaldeído/toxicidade , Hiperalgesia/etiologia , Masculino , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Medição da Dor
10.
EMBO J ; 37(3): 427-445, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29335280

RESUMO

The voltage-gated sodium channel NaV1.7 plays a critical role in pain pathways. We generated an epitope-tagged NaV1.7 mouse that showed normal pain behaviours to identify channel-interacting proteins. Analysis of NaV1.7 complexes affinity-purified under native conditions by mass spectrometry revealed 267 proteins associated with Nav1.7 in vivo The sodium channel ß3 (Scn3b), rather than the ß1 subunit, complexes with Nav1.7, and we demonstrate an interaction between collapsing-response mediator protein (Crmp2) and Nav1.7, through which the analgesic drug lacosamide regulates Nav1.7 current density. Novel NaV1.7 protein interactors including membrane-trafficking protein synaptotagmin-2 (Syt2), L-type amino acid transporter 1 (Lat1) and transmembrane P24-trafficking protein 10 (Tmed10) together with Scn3b and Crmp2 were validated by co-immunoprecipitation (Co-IP) from sensory neuron extract. Nav1.7, known to regulate opioid receptor efficacy, interacts with the G protein-regulated inducer of neurite outgrowth (Gprin1), an opioid receptor-binding protein, demonstrating a physical and functional link between Nav1.7 and opioid signalling. Further information on physiological interactions provided with this normal epitope-tagged mouse should provide useful insights into the many functions now associated with the NaV1.7 channel.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Dor/fisiopatologia , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides/metabolismo , Células Receptoras Sensoriais/metabolismo , Acetamidas/farmacologia , Analgésicos/farmacologia , Animais , Linhagem Celular , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lacosamida , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Ligação Proteica , Mapeamento de Interação de Proteínas , Transporte Proteico/fisiologia , Sinaptotagmina II/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Subunidade beta-3 do Canal de Sódio Disparado por Voltagem/metabolismo
11.
Nat Commun ; 6: 8967, 2015 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-26634308

RESUMO

Loss-of-function mutations in the SCN9A gene encoding voltage-gated sodium channel Nav1.7 cause congenital insensitivity to pain in humans and mice. Surprisingly, many potent selective antagonists of Nav1.7 are weak analgesics. We investigated whether Nav1.7, as well as contributing to electrical signalling, may have additional functions. Here we report that Nav1.7 deletion has profound effects on gene expression, leading to an upregulation of enkephalin precursor Penk mRNA and met-enkephalin protein in sensory neurons. In contrast, Nav1.8-null mutant sensory neurons show no upregulated Penk mRNA expression. Application of the opioid antagonist naloxone potentiates noxious peripheral input into the spinal cord and dramatically reduces analgesia in both female and male Nav1.7-null mutant mice, as well as in a human Nav1.7-null mutant. These data suggest that Nav1.7 channel blockers alone may not replicate the analgesic phenotype of null mutant humans and mice, but may be potentiated with exogenous opioids.


Assuntos
Encefalinas/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Insensibilidade Congênita à Dor/metabolismo , Adulto , Animais , Encefalinas/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Insensibilidade Congênita à Dor/genética , Insensibilidade Congênita à Dor/fisiopatologia , Sensação , Células Receptoras Sensoriais/metabolismo
12.
EMBO Mol Med ; 6(2): 239-58, 2014 02.
Artigo em Inglês | MEDLINE | ID: mdl-24378569

RESUMO

Muscular dystrophies are severe genetic diseases for which no efficacious therapies exist. Experimental clinical treatments include intra-arterial administration of vessel-associated stem cells, called mesoangioblasts (MABs). However, one of the limitations of this approach is the relatively low number of cells that engraft the diseased tissue, due, at least in part, to the sub-optimal efficiency of extravasation, whose mechanisms for MAB are unknown. Leukocytes emigrate into the inflamed tissues by crossing endothelial cell-to-cell junctions and junctional proteins direct and control leukocyte diapedesis. Here, we identify the endothelial junctional protein JAM-A as a key regulator of MAB extravasation. We show that JAM-A gene inactivation and JAM-A blocking antibodies strongly enhance MAB engraftment in dystrophic muscle. In the absence of JAM-A, the exchange factors EPAC-1 and 2 are down-regulated, which prevents the activation of the small GTPase Rap-1. As a consequence, junction tightening is reduced, allowing MAB diapedesis. Notably, pharmacological inhibition of Rap-1 increases MAB engraftment in dystrophic muscle, which results into a significant improvement of muscle function offering a novel strategy for stem cell-based therapies.


Assuntos
Moléculas de Adesão Celular/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Distrofias Musculares/terapia , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Transplante de Células-Tronco , Células-Tronco/citologia , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Cardiotoxinas , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/deficiência , Movimento Celular , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Camundongos , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Distrofias Musculares/patologia , Distrofias Musculares/cirurgia , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/deficiência , Sarcoglicanas/deficiência , Sarcoglicanas/metabolismo
13.
J Vis Exp ; (83): e50532, 2014 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-24472871

RESUMO

Patient-derived iPSCs could be an invaluable source of cells for future autologous cell therapy protocols. iPSC-derived myogenic stem/progenitor cells similar to pericyte-derived mesoangioblasts (iPSC-derived mesoangioblast-like stem/progenitor cells: IDEMs) can be established from iPSCs generated from patients affected by different forms of muscular dystrophy. Patient-specific IDEMs can be genetically corrected with different strategies (e.g. lentiviral vectors, human artificial chromosomes) and enhanced in their myogenic differentiation potential upon overexpression of the myogenesis regulator MyoD. This myogenic potential is then assessed in vitro with specific differentiation assays and analyzed by immunofluorescence. The regenerative potential of IDEMs is further evaluated in vivo, upon intramuscular and intra-arterial transplantation in two representative mouse models displaying acute and chronic muscle regeneration. The contribution of IDEMs to the host skeletal muscle is then confirmed by different functional tests in transplanted mice. In particular, the amelioration of the motor capacity of the animals is studied with treadmill tests. Cell engraftment and differentiation are then assessed by a number of histological and immunofluorescence assays on transplanted muscles. Overall, this paper describes the assays and tools currently utilized to evaluate the differentiation capacity of IDEMs, focusing on the transplantation methods and subsequent outcome measures to analyze the efficacy of cell transplantation.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/fisiologia , Regeneração/fisiologia , Medicina Regenerativa/métodos , Animais , Modelos Animais de Doenças , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Modelos Animais , Fibras Musculares Esqueléticas/citologia , Músculo Esquelético/citologia , Distrofia Muscular Animal/cirurgia
14.
Obes Surg ; 24(2): 241-52, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23996294

RESUMO

Laparoscopic Roux-en-Y gastric bypass (LRYGBP) reduces appetite and induces significant and sustainable weight loss. Circulating gut hormones changes engendered by LRYGBP are implicated in mediating these beneficial effects. Laparoscopic sleeve gastrectomy (LSG) is advocated as an alternative to LRYGBP, with comparable short-term weight loss and metabolic outcomes. LRYGBP and LSG are anatomically distinct procedures causing differential entero-endocrine cell nutrient exposure and thus potentially different gut hormone changes. Studies reporting the comparative effects of LRYGBP and LSG on appetite and circulating gut hormones are controversial, with no data to date on the effects of LSG on circulating peptide YY3-36 (PYY3-36) levels, the specific PYY anorectic isoform. In this study, we prospectively investigated appetite and gut hormone changes in response to LRYGBP and LSG in adiposity-matched non-diabetic patients. Anthropometric indices, leptin, fasted and nutrient-stimulated acyl-ghrelin, active glucagon-like peptide-1 (GLP-1), PYY3-36 levels and appetite were determined pre-operatively and at 6 and 12 weeks post-operatively in obese, non-diabetic females, with ten undergoing LRYGBP and eight adiposity-matched females undergoing LSG. LRYGBP and LSG comparably reduced adiposity. LSG decreased fasting and post-prandial plasma acyl-ghrelin compared to pre-surgery and to LRYGBP. Nutrient-stimulated PYY3-36 and active GLP-1 concentrations increased post-operatively in both groups. However, LRYGBP induced greater, more sustained PYY3-36 and active GLP-1 increments compared to LSG. LRYGBP suppressed fasting hunger compared to LSG. A similar increase in post-prandial fullness was observed post-surgery following both procedures. LRYGBP and LSG produced comparable enhanced satiety and weight loss. However, LSG and LRYGBP differentially altered gut hormone profiles.


Assuntos
Apetite , Gastrectomia , Derivação Gástrica , Grelina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Laparoscopia , Obesidade Mórbida/cirurgia , Fragmentos de Peptídeos/metabolismo , Peptídeo YY/metabolismo , Adolescente , Adulto , Índice de Massa Corporal , Feminino , Humanos , Pessoa de Meia-Idade , Obesidade Mórbida/metabolismo , Estudos Prospectivos , Resultado do Tratamento , Redução de Peso
15.
J Clin Invest ; 123(8): 3539-51, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23867619

RESUMO

Polymorphisms in the fat mass and obesity-associated gene (FTO) are associated with human obesity and obesity-prone behaviors, including increased food intake and a preference for energy-dense foods. FTO demethylates N6-methyladenosine, a potential regulatory RNA modification, but the mechanisms by which FTO predisposes humans to obesity remain unclear. In adiposity-matched, normal-weight humans, we showed that subjects homozygous for the FTO "obesity-risk" rs9939609 A allele have dysregulated circulating levels of the orexigenic hormone acyl-ghrelin and attenuated postprandial appetite reduction. Using functional MRI (fMRI) in normal-weight AA and TT humans, we found that the FTO genotype modulates the neural responses to food images in homeostatic and brain reward regions. Furthermore, AA and TT subjects exhibited divergent neural responsiveness to circulating acyl-ghrelin within brain regions that regulate appetite, reward processing, and incentive motivation. In cell models, FTO overexpression reduced ghrelin mRNA N6-methyladenosine methylation, concomitantly increasing ghrelin mRNA and peptide levels. Furthermore, peripheral blood cells from AA human subjects exhibited increased FTO mRNA, reduced ghrelin mRNA N6-methyladenosine methylation, and increased ghrelin mRNA abundance compared with TT subjects. Our findings show that FTO regulates ghrelin, a key mediator of ingestive behavior, and offer insight into how FTO obesity-risk alleles predispose to increased energy intake and obesity in humans.


Assuntos
Apetite , Grelina/sangue , Proteínas/genética , Aciltransferases/genética , Aciltransferases/metabolismo , Adolescente , Adulto , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Encéfalo/fisiologia , Ingestão de Alimentos/psicologia , Alimentos , Neuroimagem Funcional , Expressão Gênica , Regulação da Expressão Gênica , Estudos de Associação Genética , Células HEK293 , Humanos , Imageamento por Ressonância Magnética , Masculino , Metilação , Camundongos , Camundongos Knockout , Polimorfismo de Nucleotídeo Único , Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recompensa , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...