Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 201
Filter
Add more filters











Publication year range
1.
Nat Commun ; 15(1): 5353, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918403

ABSTRACT

Nociceptin/orphanin-FQ (N/OFQ) is a recently appreciated critical opioid peptide with key regulatory functions in several central behavioral processes including motivation, stress, feeding, and sleep. The functional relevance of N/OFQ action in the mammalian brain remains unclear due to a lack of high-resolution approaches to detect this neuropeptide with appropriate spatial and temporal resolution. Here we develop and characterize NOPLight, a genetically encoded sensor that sensitively reports changes in endogenous N/OFQ release. We characterized the affinity, pharmacological profile, spectral properties, kinetics, ligand selectivity, and potential interaction with intracellular signal transducers of NOPLight in vitro. Its functionality was established in acute brain slices by exogeneous N/OFQ application and chemogenetic induction of endogenous N/OFQ release from PNOC neurons. In vivo studies with fibre photometry enabled direct recording of NOPLight binding to exogenous N/OFQ receptor ligands, as well as detection of endogenous N/OFQ release within the paranigral ventral tegmental area (pnVTA) during natural behaviors and chemogenetic activation of PNOC neurons. In summary, we show here that NOPLight can be used to detect N/OFQ opioid peptide signal dynamics in tissue and freely behaving animals.


Subject(s)
Neurons , Nociceptin , Opioid Peptides , Receptors, Opioid , Animals , Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Receptors, Opioid/genetics , Neurons/metabolism , Humans , Mice , Male , Ventral Tegmental Area/metabolism , Nociceptin Receptor , HEK293 Cells , Brain/metabolism , Mice, Inbred C57BL , Ligands , Biosensing Techniques/methods
2.
Nat Commun ; 15(1): 5439, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937485

ABSTRACT

Efficient control of feeding behavior requires the coordinated adjustment of complex motivational and affective neurocircuits. Neuropeptides from energy-sensing hypothalamic neurons are potent feeding modulators, but how these endogenous signals shape relevant circuits remains unclear. Here, we examine how the orexigenic neuropeptide Y (NPY) adapts GABAergic inputs to the bed nucleus of the stria terminalis (BNST). We find that fasting increases synaptic connectivity between agouti-related peptide (AgRP)-expressing 'hunger' and BNST neurons, a circuit that promotes feeding. In contrast, GABAergic input from the central amygdala (CeA), an extended amygdala circuit that decreases feeding, is reduced. Activating NPY-expressing AgRP neurons evokes these synaptic adaptations, which are absent in NPY-deficient mice. Moreover, fasting diminishes the ability of CeA projections in the BNST to suppress food intake, and NPY-deficient mice fail to decrease anxiety in order to promote feeding. Thus, AgRP neurons drive input-specific synaptic plasticity, enabling a selective shift in hunger and anxiety signaling during starvation through NPY.


Subject(s)
Agouti-Related Protein , Feeding Behavior , Neuronal Plasticity , Neuropeptide Y , Septal Nuclei , Starvation , Animals , Neuropeptide Y/metabolism , Neuropeptide Y/genetics , Neuronal Plasticity/physiology , Agouti-Related Protein/metabolism , Agouti-Related Protein/genetics , Feeding Behavior/physiology , Septal Nuclei/metabolism , Septal Nuclei/physiology , Mice , Starvation/metabolism , Male , Amygdala/metabolism , Amygdala/physiology , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/physiology , GABAergic Neurons/metabolism , Eating/physiology , Fasting/physiology , Anxiety/metabolism , Anxiety/physiopathology , Hunger/physiology
3.
Science ; 384(6694): 438-446, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38662831

ABSTRACT

Liver mitochondria play a central role in metabolic adaptations to changing nutritional states, yet their dynamic regulation upon anticipated changes in nutrient availability has remained unaddressed. Here, we found that sensory food perception rapidly induced mitochondrial fragmentation in the liver through protein kinase B/AKT (AKT)-dependent phosphorylation of serine 131 of the mitochondrial fission factor (MFFS131). This response was mediated by activation of hypothalamic pro-opiomelanocortin (POMC)-expressing neurons. A nonphosphorylatable MFFS131G knock-in mutation abrogated AKT-induced mitochondrial fragmentation in vitro. In vivo, MFFS131G knock-in mice displayed altered liver mitochondrial dynamics and impaired insulin-stimulated suppression of hepatic glucose production. Thus, rapid activation of a hypothalamus-liver axis can adapt mitochondrial function to anticipated changes of nutritional state in control of hepatic glucose metabolism.


Subject(s)
Food , Gluconeogenesis , Glucose , Liver , Membrane Proteins , Mitochondria, Liver , Mitochondrial Dynamics , Mitochondrial Proteins , Perception , Animals , Male , Mice , Gene Knock-In Techniques , Glucose/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Liver/metabolism , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Neurons/metabolism , Phosphorylation , Pro-Opiomelanocortin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Transgenic
4.
Nat Commun ; 15(1): 3443, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658557

ABSTRACT

The hypothalamus contains a remarkable diversity of neurons that orchestrate behavioural and metabolic outputs in a highly plastic manner. Neuronal diversity is key to enabling hypothalamic functions and, according to the neuroscience dogma, it is predetermined during embryonic life. Here, by combining lineage tracing of hypothalamic pro-opiomelanocortin (Pomc) neurons with single-cell profiling approaches in adult male mice, we uncovered subpopulations of 'Ghost' neurons endowed with atypical molecular and functional identity. Compared to 'classical' Pomc neurons, Ghost neurons exhibit negligible Pomc expression and are 'invisible' to available neuroanatomical approaches and promoter-based reporter mice for studying Pomc biology. Ghost neuron numbers augment in diet-induced obese mice, independent of neurogenesis or cell death, but weight loss can reverse this shift. Our work challenges the notion of fixed, developmentally programmed neuronal identities in the mature hypothalamus and highlight the ability of specialised neurons to reversibly adapt their functional identity to adult-onset obesogenic stimuli.


Subject(s)
Hypothalamus , Neurons , Obesity , Pro-Opiomelanocortin , Single-Cell Analysis , Animals , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/genetics , Neurons/metabolism , Obesity/metabolism , Obesity/pathology , Male , Mice , Hypothalamus/metabolism , Hypothalamus/cytology , Disease Models, Animal , Diet, High-Fat , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis , Mice, Obese
5.
Diabetes ; 73(7): 1058-1071, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38608276

ABSTRACT

The Rab-GTPase-activating protein (RabGAP) TBC1D4 (AS160) represents a key component in the regulation of glucose transport into skeletal muscle and white adipose tissue (WAT) and is therefore crucial during the development of insulin resistance and type 2 diabetes. Increased daily activity has been shown to be associated with improved postprandial hyperglycemia in allele carriers of a loss-of-function variant in the human TBC1D4 gene. Using conventional Tbc1d4-deficient mice (D4KO) fed a high-fat diet, we show that moderate endurance exercise training leads to substantially improved glucose and insulin tolerance and enhanced expression levels of markers for mitochondrial activity and browning in WAT from D4KO animals. Importantly, in vivo and ex vivo analyses of glucose uptake revealed increased glucose clearance in interscapular brown adipose tissue and WAT from trained D4KO mice. Thus, chronic exercise is able to overcome the genetically induced insulin resistance caused by Tbc1d4 depletion. Gene variants in TBC1D4 may be relevant in future precision medicine as determinants of exercise response.


Subject(s)
Adipose Tissue, White , GTPase-Activating Proteins , Insulin Resistance , Mice, Knockout , Physical Conditioning, Animal , Insulin Resistance/genetics , Insulin Resistance/physiology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Animals , Mice , Physical Conditioning, Animal/physiology , Adipose Tissue, White/metabolism , Diet, High-Fat , Male , Adipose Tissue, Brown/metabolism , Muscle, Skeletal/metabolism , Glucose/metabolism , Mice, Inbred C57BL
6.
Cell Rep ; 43(4): 113960, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38507407

ABSTRACT

GFRAL-expressing neurons actuate aversion and nausea, are targets for obesity treatment, and may mediate metformin effects by long-term GDF15-GFRAL agonism. Whether GFRAL+ neurons acutely regulate glucose and energy homeostasis is, however, underexplored. Here, we report that cell-specific activation of GFRAL+ neurons using a variety of techniques causes a torpor-like state, including hypothermia, the release of stress hormones, a shift from glucose to lipid oxidation, and impaired insulin sensitivity, glucose tolerance, and skeletal muscle glucose uptake but augmented glucose uptake in visceral fat. Metabolomic analysis of blood and transcriptomics of muscle and fat indicate alterations in ketogenesis, insulin signaling, adipose tissue differentiation and mitogenesis, and energy fluxes. Our findings indicate that acute GFRAL+ neuron activation induces endocrine and gluco- and thermoregulatory responses associated with nausea and torpor. While chronic activation of GFRAL signaling promotes weight loss in obesity, these results show that acute activation of GFRAL+ neurons causes hypothermia and hyperglycemia.


Subject(s)
Glucose , Hypothermia , Nausea , Neurons , Torpor , Animals , Neurons/metabolism , Nausea/metabolism , Hypothermia/metabolism , Torpor/physiology , Glucose/metabolism , Mice , Male , Muscle, Skeletal/metabolism , Mice, Inbred C57BL , Insulin/metabolism , Insulin Resistance , Signal Transduction
7.
Nat Metab ; 6(3): 473-493, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38378998

ABSTRACT

Agouti-related peptide (AgRP)-expressing and proopiomelanocortin (POMC)-expressing neurons reciprocally regulate food intake. Here, we combine non-interacting recombinases to simultaneously express functionally opposing chemogenetic receptors in AgRP and POMC neurons for comparing metabolic responses in male and female mice with simultaneous activation of AgRP and inhibition of POMC neurons with isolated activation of AgRP neurons or isolated inhibition of POMC neurons. We show that food intake is regulated by the additive effect of AgRP neuron activation and POMC neuron inhibition, while systemic insulin sensitivity and gluconeogenesis are differentially modulated by isolated-versus-simultaneous regulation of AgRP and POMC neurons. We identify a neurocircuit engaging Npy1R-expressing neurons in the paraventricular nucleus of the hypothalamus, where activated AgRP neurons and inhibited POMC neurons cooperate to promote food consumption and activate Th+ neurons in the nucleus tractus solitarii. Collectively, these results unveil how food intake is precisely regulated by the simultaneous bidirectional interplay between AgRP and POMC neurocircuits.


Subject(s)
Neurons , Pro-Opiomelanocortin , Mice , Male , Female , Animals , Pro-Opiomelanocortin/metabolism , Agouti-Related Protein/metabolism , Neurons/metabolism , Hypothalamus/metabolism
8.
Cells ; 13(1)2024 01 04.
Article in English | MEDLINE | ID: mdl-38201306

ABSTRACT

Enteroendocrine cells (EECs) constitute only a small proportion of Villin-1 (Vil1)-expressing intestinal epithelial cells (IECs) of the gastrointestinal tract; yet, in sum, they build the largest endocrine organ of the body, with each of them storing and releasing a distinct set of peptides for the control of feeding behavior, glucose metabolism, and gastrointestinal motility. Like all IEC types, EECs are continuously renewed from intestinal stem cells in the crypt base and terminally differentiate into mature subtypes while moving up the crypt-villus axis. Interestingly, EECs adjust their hormonal secretion according to their migration state as EECs receive altering differentiation signals along the crypt-villus axis and thus undergo functional readaptation. Cell-specific targeting of mature EEC subtypes by specific promoters is challenging because the expression of EEC-derived peptides and their precursors is not limited to EECs but are also found in other organs, such as the brain (e.g., Cck and Sst) as well as in the pancreas (e.g., Sst and Gcg). Here, we describe an intersectional genetic approach that enables cell type-specific targeting of functionally distinct EEC subtypes by combining a newly generated Dre-recombinase expressing mouse line (Vil1-2A-DD-Dre) with multiple existing Cre-recombinase mice and mouse strains with rox and loxP sites flanked stop cassettes for transgene expression. We found that transgene expression in triple-transgenic mice is highly specific in I but not D and L cells in the terminal villi of the small intestine. The targeting of EECs only in terminal villi is due to the integration of a defective 2A separating peptide that, combined with low EEC intrinsic Vil1 expression, restricts our Vil1-2A-DD-Dre mouse line and the intersectional genetic approach described here only applicable for the investigation of mature EEC subpopulations.


Subject(s)
Duodenum , Intestine, Small , Mice , Animals , Enteroendocrine Cells , Mice, Transgenic , Peptides
9.
Nat Commun ; 14(1): 7824, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38016943

ABSTRACT

Dysregulation of hypothalamic ceramides has been associated with disrupted neuronal pathways in control of energy and glucose homeostasis. However, the specific ceramide species promoting neuronal lipotoxicity in obesity have remained obscure. Here, we find increased expression of the C16:0 ceramide-producing ceramide synthase (CerS)6 in cultured hypothalamic neurons exposed to palmitate in vitro and in the hypothalamus of obese mice. Conditional deletion of CerS6 in hypothalamic neurons attenuates high-fat diet (HFD)-dependent weight gain and improves glucose metabolism. Specifically, CerS6 deficiency in neurons expressing pro-opiomelanocortin (POMC) or steroidogenic factor 1 (SF-1) alters feeding behavior and alleviates the adverse metabolic effects of HFD feeding on insulin sensitivity and glucose tolerance. POMC-expressing cell-selective deletion of CerS6 prevents the diet-induced alterations of mitochondrial morphology and improves cellular leptin sensitivity. Our experiments reveal functions of CerS6-derived ceramides in hypothalamic lipotoxicity, altered mitochondrial dynamics, and ER/mitochondrial stress in the deregulation of food intake and glucose metabolism in obesity.


Subject(s)
Obesity , Pro-Opiomelanocortin , Animals , Mice , Ceramides/metabolism , Diet, High-Fat/adverse effects , Glucose/metabolism , Homeostasis , Hypothalamus/metabolism , Mice, Obese , Neurons/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/metabolism
10.
Science ; 381(6665): eabl7398, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37769095

ABSTRACT

Systemic metabolism has to be constantly adjusted to the variance of food intake and even be prepared for anticipated changes in nutrient availability. Therefore, the brain integrates multiple homeostatic signals with numerous cues that predict future deviations in energy supply. Recently, our understanding of the neural pathways underlying these regulatory principles-as well as their convergence in the hypothalamus as the key coordinator of food intake, energy expenditure, and glucose metabolism-have been revealed. These advances have changed our view of brain-dependent control of metabolic physiology. In this Review, we discuss new concepts about how alterations in these pathways contribute to the development of prevalent metabolic diseases such as obesity and type 2 diabetes mellitus and how this emerging knowledge may provide new targets for their treatment.


Subject(s)
Brain-Gut Axis , Diabetes Mellitus, Type 2 , Eating , Energy Metabolism , Hypothalamus , Neural Pathways , Obesity , Humans , Diabetes Mellitus, Type 2/physiopathology , Homeostasis , Hypothalamus/physiology , Obesity/physiopathology , Neural Pathways/physiopathology
11.
Nat Metab ; 5(8): 1352-1363, 2023 08.
Article in English | MEDLINE | ID: mdl-37592007

ABSTRACT

Survival under selective pressure is driven by the ability of our brain to use sensory information to our advantage to control physiological needs. To that end, neural circuits receive and integrate external environmental cues and internal metabolic signals to form learned sensory associations, consequently motivating and adapting our behaviour. The dopaminergic midbrain plays a crucial role in learning adaptive behaviour and is particularly sensitive to peripheral metabolic signals, including intestinal peptides, such as glucagon-like peptide 1 (GLP-1). In a single-blinded, randomized, controlled, crossover basic human functional magnetic resonance imaging study relying on a computational model of the adaptive learning process underlying behavioural responses, we show that adaptive learning is reduced when metabolic sensing is impaired in obesity, as indexed by reduced insulin sensitivity (participants: N = 30 with normal insulin sensitivity; N = 24 with impaired insulin sensitivity). Treatment with the GLP-1 receptor agonist liraglutide normalizes impaired learning of sensory associations in men and women with obesity. Collectively, our findings reveal that GLP-1 receptor activation modulates associative learning in people with obesity via its central effects within the mesoaccumbens pathway. These findings provide evidence for how metabolic signals can act as neuromodulators to adapt our behaviour to our body's internal state and how GLP-1 receptor agonists work in clinics.


Subject(s)
Insulin Resistance , Liraglutide , Male , Humans , Female , Liraglutide/pharmacology , Liraglutide/therapeutic use , Glucagon-Like Peptide-1 Receptor , Glucagon-Like Peptide 1 , Obesity/drug therapy
12.
bioRxiv ; 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37292957

ABSTRACT

Nociceptin/orphanin-FQ (N/OFQ) is a recently appreciated critical opioid peptide with key regulatory functions in several central behavioral processes including motivation, stress, feeding, and sleep. The functional relevance of N/OFQ action in the mammalian brain remains unclear due to a lack of high-resolution approaches to detect this neuropeptide with appropriate spatial and temporal resolution. Here we develop and characterize NOPLight, a genetically encoded sensor that sensitively reports changes in endogenous N/OFQ release. We characterized the affinity, pharmacological profile, spectral properties, kinetics, ligand selectivity, and potential interaction with intracellular signal transducers of NOPLight in vitro. Its functionality was established in acute brain slices by exogeneous N/OFQ application and chemogenetic induction of endogenous N/OFQ release from PNOC neurons. In vivo studies with fiber photometry enabled a direct recording of binding by N/OFQ receptor ligands, as well as the detection of natural or chemogenetically-evoked endogenous N/OFQ release within the paranigral ventral tegmental area (pnVTA). In summary, we show that NOPLight can be used to detect N/OFQ opioid peptide signal dynamics in tissue and freely-behaving animals.

13.
Mol Metab ; 71: 101707, 2023 05.
Article in English | MEDLINE | ID: mdl-36933618

ABSTRACT

BACKGROUND/PURPOSE: Litter size is a biological variable that strongly influences adult physiology in rodents. Despite evidence from previous decades and recent studies highlighting its major impact on metabolism, information about litter size is currently underreported in the scientific literature. Here, we urge that this important biological variable should be explicitly stated in research articles. RESULTS/CONCLUSION: Below, we briefly describe the scientific evidence supporting the impact of litter size on adult physiology and outline a series of recommendations and guidelines to be implemented by investigators, funding agencies, editors in scientific journals, and animal suppliers to fill this important gap.


Subject(s)
Rodentia , Pregnancy , Animals , Female , Litter Size/physiology
14.
Cell Metab ; 35(4): 571-584.e6, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36958330

ABSTRACT

Western diets rich in fat and sugar promote excess calorie intake and weight gain; however, the underlying mechanisms are unclear. Despite a well-documented association between obesity and altered brain dopamine function, it remains elusive whether these alterations are (1) pre-existing, increasing the individual susceptibility to weight gain, (2) secondary to obesity, or (3) directly attributable to repeated exposure to western diet. To close this gap, we performed a randomized, controlled study (NCT05574660) with normal-weight participants exposed to a high-fat/high-sugar snack or a low-fat/low-sugar snack for 8 weeks in addition to their regular diet. The high-fat/high-sugar intervention decreased the preference for low-fat food while increasing brain response to food and associative learning independent of food cues or reward. These alterations were independent of changes in body weight and metabolic parameters, indicating a direct effect of high-fat, high-sugar foods on neurobehavioral adaptations that may increase the risk for overeating and weight gain.


Subject(s)
Reward , Snacks , Humans , Obesity/metabolism , Weight Gain , Sugars
15.
Cell Rep Med ; 4(1): 100897, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36652907

ABSTRACT

Feeding behavior must be continuously adjusted to match energy needs. Recent discoveries in murine models identified uridine as a regulator of energy balance. Here, we explore its contribution to the complex control of food intake in humans by administering a single dose of uridine monophosphate (UMP; 0.5 or 1 g) to healthy participants in two placebo-controlled studies designed to assess food behavior (registration: DRKS00014874). We establish that endogenous circulating uridine correlates with hunger and ensuing food consumption. It also dynamically decreases upon caloric ingestion, prompting its potential role in a negative feedback loop regulating energy intake. We further demonstrate that oral UMP administration temporarily increases circulating uridine and-when within the physiological range-enhances hunger and caloric intake proportionally to participants' basal energy needs. Overall, uridine appears as a potential target to tackle dysfunctions of feeding behavior in humans.


Subject(s)
Energy Intake , Hunger , Humans , Animals , Mice , Uridine , Energy Intake/physiology , Hunger/physiology , Uridine Monophosphate , Eating
16.
Glia ; 71(4): 1120-1141, 2023 04.
Article in English | MEDLINE | ID: mdl-36583573

ABSTRACT

The sphingolipids galactosylceramide (GalCer), sulfatide (ST) and sphingomyelin (SM) are essential for myelin stability and function. GalCer and ST are synthesized mostly from C22-C24 ceramides, generated by Ceramide Synthase 2 (CerS2). To clarify the requirement for C22-C24 sphingolipid synthesis in myelin biosynthesis and stability, we generated mice lacking CerS2 specifically in myelinating cells (CerS2ΔO/ΔO ). At 6 weeks of age, normal-appearing myelin had formed in CerS2ΔO/ΔO mice, however there was a reduction in myelin thickness and the percentage of myelinated axons. Pronounced loss of C22-C24 sphingolipids in myelin of CerS2ΔO/ΔO mice was compensated by greatly increased levels of C18 sphingolipids. A distinct microglial population expressing high levels of activation and phagocytic markers such as CD64, CD11c, MHC class II, and CD68 was apparent at 6 weeks of age in CerS2ΔO/ΔO mice, and had increased by 10 weeks. Increased staining for denatured myelin basic protein was also apparent in 6-week-old CerS2ΔO/ΔO mice. By 16 weeks, CerS2ΔO/ΔO mice showed pronounced myelin atrophy, motor deficits, and axon beading, a hallmark of axon stress. 90% of CerS2ΔO/ΔO mice died between 16 and 26 weeks of age. This study highlights the importance of sphingolipid acyl chain length for the structural integrity of myelin, demonstrating how a modest reduction in lipid chain length causes exposure of a denatured myelin protein epitope and expansion of phagocytic microglia, followed by axon pathology, myelin degeneration, and motor deficits. Understanding the molecular trigger for microglial activation should aid the development of therapeutics for demyelinating and neurodegenerative diseases.


Subject(s)
Microglia , Myelin Sheath , Mice , Animals , Microglia/metabolism , Myelin Sheath/metabolism , Ceramides/metabolism , Sphingolipids/metabolism
17.
Nat Cell Biol ; 25(1): 20-29, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36543979

ABSTRACT

Impaired proinsulin-to-insulin processing in pancreatic ß-cells is a key defective step in both type 1 diabetes and type 2 diabetes (T2D) (refs. 1,2), but the mechanisms involved remain to be defined. Altered metabolism of sphingolipids (SLs) has been linked to development of obesity, type 1 diabetes and T2D (refs. 3-8); nonetheless, the role of specific SL species in ß-cell function and demise is unclear. Here we define the lipid signature of T2D-associated ß-cell failure, including an imbalance of specific very-long-chain SLs and long-chain SLs. ß-cell-specific ablation of CerS2, the enzyme necessary for generation of very-long-chain SLs, selectively reduces insulin content, impairs insulin secretion and disturbs systemic glucose tolerance in multiple complementary models. In contrast, ablation of long-chain-SL-synthesizing enzymes has no effect on insulin content. By quantitatively defining the SL-protein interactome, we reveal that CerS2 ablation affects SL binding to several endoplasmic reticulum-Golgi transport proteins, including Tmed2, which we define as an endogenous regulator of the essential proinsulin processing enzyme Pcsk1. Our study uncovers roles for specific SL subtypes and SL-binding proteins in ß-cell function and T2D-associated ß-cell failure.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Humans , Proinsulin/genetics , Proinsulin/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Sphingolipids/metabolism , Diabetes Mellitus, Type 1/metabolism , Insulin/metabolism , Homeostasis , Carrier Proteins/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism
18.
Mol Metab ; 66: 101626, 2022 12.
Article in English | MEDLINE | ID: mdl-36356831

ABSTRACT

OBJECTIVE: Nonalcoholic fatty liver disease (NAFLD) ranges from steatosis to nonalcoholic steatohepatitis (NASH), which often progresses to hepatocellular carcinoma (HCC) through a largely undefined mechanism. NASH and HCC depend on inflammatory signaling, whose master regulator is the NFκB transcription factor family, activated by canonical and non-canonical pathways. METHODS: Here, we investigated non-canonical NFκB-inducing kinase (NIK/MAP3K14) in metabolic NASH, NASH to HCC transition, and DEN-induced HCC. To this end, we performed dietary and chemical interventions in mice that were analyzed via single nucleus sequencing, gene expression and histochemical methods. Ultimately, we verified our mouse results in human patient samples. RESULTS: We revealed that hepatocyte-specific NIK deficiency (NIKLKO) ameliorated metabolic NASH complications and reduced hepatocarcinogenesis, independent of its role in the NFκB pathway. Instead, hepatic NIK attenuated hepatoprotective JAK2/STAT5 signaling that is a prerequisite for NASH and NASH to HCC progression in mice and humans. CONCLUSIONS: Our data suggest NIK-mediated inhibitory JAK2 phosphorylation at serine 633 that might be amenable for future therapeutic interventions in patients.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Humans , Mice , Carcinoma, Hepatocellular/metabolism , Hepatocytes/metabolism , Janus Kinase 2/metabolism , Liver Neoplasms/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , STAT5 Transcription Factor/metabolism , NF-kappaB-Inducing Kinase
19.
JCI Insight ; 7(21)2022 11 08.
Article in English | MEDLINE | ID: mdl-36345942

ABSTRACT

Dopamine acts on neurons in the arcuate nucleus (ARC) of the hypothalamus, which controls homeostatic feeding responses. Here we demonstrate a differential enrichment of dopamine receptor 1 (Drd1) expression in food intake-promoting agouti related peptide (AgRP)/neuropeptide Y (NPY) neurons and a large proportion of Drd2-expressing anorexigenic proopiomelanocortin (POMC) neurons. Owing to the nature of these receptors, this translates into a predominant activation of AgRP/NPY neurons upon dopamine stimulation and a larger proportion of dopamine-inhibited POMC neurons. Employing intersectional targeting of Drd2-expressing POMC neurons, we reveal that dopamine-mediated POMC neuron inhibition is Drd2 dependent and that POMCDrd2+ neurons exhibit differential expression of neuropeptide signaling mediators compared with the global POMC neuron population, which manifests in enhanced somatostatin responsiveness of POMCDrd2+ neurons. Selective chemogenetic activation of POMCDrd2+ neurons uncovered their ability to acutely suppress feeding and to preserve body temperature in fasted mice. Collectively, the present study provides the molecular and functional characterization of POMCDrd2+ neurons and aids our understanding of dopamine-dependent control of homeostatic energy-regulatory neurocircuits.


Subject(s)
Dopamine , Pro-Opiomelanocortin , Animals , Mice , Agouti-Related Protein/metabolism , Body Temperature , Dopamine/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/metabolism
20.
Nat Metab ; 4(10): 1402-1419, 2022 10.
Article in English | MEDLINE | ID: mdl-36266547

ABSTRACT

The hypothalamus plays a key role in coordinating fundamental body functions. Despite recent progress in single-cell technologies, a unified catalog and molecular characterization of the heterogeneous cell types and, specifically, neuronal subtypes in this brain region are still lacking. Here, we present an integrated reference atlas, 'HypoMap,' of the murine hypothalamus, consisting of 384,925 cells, with the ability to incorporate new additional experiments. We validate HypoMap by comparing data collected from Smart-Seq+Fluidigm C1 and bulk RNA sequencing of selected neuronal cell types with different degrees of cellular heterogeneity. Finally, via HypoMap, we identify classes of neurons expressing glucagon-like peptide-1 receptor (Glp1r) and prepronociceptin (Pnoc), and validate them using single-molecule in situ hybridization. Collectively, HypoMap provides a unified framework for the systematic functional annotation of murine hypothalamic cell types, and it can serve as an important platform to unravel the functional organization of hypothalamic neurocircuits and to identify druggable targets for treating metabolic disorders.


Subject(s)
Glucagon-Like Peptide-1 Receptor , Hypothalamus , Mice , Animals , Glucagon-Like Peptide-1 Receptor/genetics , Hypothalamus/metabolism , Neurons/metabolism , Sequence Analysis, RNA , Gene Expression
SELECTION OF CITATIONS
SEARCH DETAIL