Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Nat Commun ; 10(1): 3446, 2019 08 01.
Article En | MEDLINE | ID: mdl-31371721

Feeding is known to be profoundly affected by stress-related emotional states and eating disorders are comorbid with psychiatric symptoms and altered emotional responses. The neural basis underlying feeding regulation by stress-related emotional changes is poorly understood. Here, we identify a novel projection from the paraventricular hypothalamus (PVH) to the ventral lateral septum (LSv) that shows a scalable regulation on feeding and behavioral changes related to emotion. Weak photostimulation of glutamatergic PVH→LSv terminals elicits stress-related self-grooming and strong photostimulation causes fear-related escape jumping associated with respective weak and strong inhibition on feeding. In contrast, inhibition of glutamatergic inputs to LSv increases feeding with signs of reduced anxiety. LSv-projecting neurons are concentrated in rostral PVH. LSv and LSv-projecting PVH neurons are activated by stressors in vivo, whereas feeding bouts were associated with reduced activity of these neurons. Thus, PVH→LSv neurotransmission underlies dynamic feeding by orchestrating emotional states, providing a novel neural circuit substrate underlying comorbidity between eating abnormalities and psychiatric disorders.


Feeding Behavior/physiology , Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Psychological Distress , Animals , Behavior, Animal , Excitatory Amino Acid Agents , Feeding and Eating Disorders , Grooming/physiology , Male , Mice , Models, Animal , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
2.
eNeuro ; 6(4)2019.
Article En | MEDLINE | ID: mdl-31331938

The paraventricular hypothalamus (PVH) regulates stress, feeding behaviors and other homeostatic processes, but whether PVH also drives defensive states remains unknown. Here we showed that photostimulation of PVH neurons in mice elicited escape jumping, a typical defensive behavior. We mapped PVH outputs that densely terminate in the ventral midbrain (vMB) area, and found that activation of the PVH→vMB circuit produced profound defensive behavioral changes, including escape jumping, hiding, hyperlocomotion, and learned aversion. Electrophysiological recordings showed excitatory postsynaptic input onto vMB neurons via PVH fiber activation, and in vivo studies demonstrated that glutamate transmission from PVH→vMB was required for the evoked behavioral responses. Photostimulation of PVH→vMB fibers induced cFos expression mainly in non-dopaminergic neurons. Using a dual optogenetic-chemogenetic strategy, we further revealed that escape jumping and hiding were partially contributed by the activation of midbrain glutamatergic neurons. Taken together, our work unveils a hypothalamic-vMB circuit that encodes defensive properties, which may be implicated in stress-induced defensive responses.


Escape Reaction/physiology , Mesencephalon/physiology , Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Animals , Avoidance Learning/physiology , Behavior, Animal , Eating/physiology , Glutamic Acid/physiology , Male , Mesencephalon/cytology , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/physiology , Optogenetics , Paraventricular Hypothalamic Nucleus/cytology
3.
Sci Adv ; 5(3): eaav1640, 2019 03.
Article En | MEDLINE | ID: mdl-30854429

Animals must consider competing information before deciding to eat: internal signals indicating the desirability of food and external signals indicating the risk involved in eating within a particular environment. The behaviors driven by the former are manifestations of hunger, and the latter, anxiety. The connection between pathologic anxiety and reduced eating in conditions like typical depression and anorexia is well known. Conversely, anti-anxiety drugs such as benzodiazepines increase appetite. Here, we show that GABAergic neurons in the diagonal band of Broca (DBBGABA) are responsive to indications of risk and receive monosynaptic inhibitory input from lateral hypothalamus GABAergic neurons (LHGABA). Activation of this circuit reduces anxiety and causes indiscriminate feeding. We also found that diazepam rapidly reduces DBBGABA activity while inducing indiscriminate feeding. Our study reveals that the LHGABA→DBBGABA neurocircuit overrides anxiogenic environmental cues to allow feeding and that this pathway may underlie the link between eating and anxiety-related disorders.


Basal Forebrain/physiology , Cues , Environment , Feeding Behavior , Hypothalamic Area, Lateral/physiology , Nerve Net , Animals , Anxiety , GABAergic Neurons/physiology , Mice , Synaptic Transmission
4.
Nat Commun ; 9(1): 52, 2018 01 04.
Article En | MEDLINE | ID: mdl-29302029

Abnormal feeding often co-exists with compulsive behaviors, but the underlying neural basis remains unknown. Excessive self-grooming in rodents is associated with compulsivity. Here, we show that optogenetically manipulating the activity of lateral hypothalamus (LH) projections targeting the paraventricular hypothalamus (PVH) differentially promotes either feeding or repetitive self-grooming. Whereas selective activation of GABAergic LH→PVH inputs induces feeding, activation of glutamatergic inputs promotes self-grooming. Strikingly, targeted stimulation of GABAergic LH→PVH leads to rapid and reversible transitions to feeding from induced intense self-grooming, while activating glutamatergic LH→PVH or PVH neurons causes rapid and reversible transitions to self-grooming from voracious feeding induced by fasting. Further, specific inhibition of either LH→PVH GABAergic action or PVH neurons reduces self-grooming induced by stress. Thus, we have uncovered a parallel LH→PVH projection circuit for antagonistic control of feeding and self-grooming through dynamic modulation of PVH neuron activity, revealing a common neural pathway that underlies feeding and compulsive behaviors.


Compulsive Behavior , Feeding Behavior , Mice/physiology , Neural Pathways , Animals , Female , Grooming , Hypothalamic Area, Lateral/physiology , Male , Mice/genetics , Paraventricular Hypothalamic Nucleus/physiology
5.
JCI Insight ; 2(14)2017 Jul 20.
Article En | MEDLINE | ID: mdl-28724789

Diet-induced obesity (DIO) represents the major cause for the current obesity epidemic, but the mechanism underlying DIO is unclear. ß-Adrenergic receptors (ß-ARs) play a major role in sympathetic nervous system-mediated (SNS-mediated) diet-induced energy expenditure (EE). Rbc express abundant ß-ARs; however, a potential role for rbc in DIO remains untested. Here, we demonstrated that high-fat, high-caloric diet (HFD) feeding increased both EE and blood O2 content, and the HFD-induced increases in blood O2 level and in body weight gain were negatively correlated. Deficiency of ß-ARs in rbc reduced glycolysis and ATP levels, diminished HFD-induced increases in both blood O2 content and EE, and resulted in DIO. Importantly, specific activation of cAMP signaling in rbc promoted HFD-induced EE and reduced HFD-induced tissue hypoxia independent of obesity. Both HFD and pharmacological activation cAMP signaling in rbc led to increased glycolysis and ATP levels. These results identify a previously unknown role for rbc ß-ARs in mediating the SNS action on HFD-induced EE by increasing O2 supply, and they demonstrate that HFD-induced EE is limited by blood O2 availability and can be augenmented by increased O2 supply.

6.
eNeuro ; 4(3)2017.
Article En | MEDLINE | ID: mdl-28560316

Leptin receptors (LepRs) expressed in the midbrain contribute to the action of leptin on feeding regulation. The midbrain neurons release a variety of neurotransmitters including dopamine (DA), glutamate and GABA. However, which neurotransmitter mediates midbrain leptin action on feeding remains unclear. Here, we showed that midbrain LepR neurons overlap with a subset of dopaminergic, GABAergic and glutamatergic neurons. Specific removal of vesicular monoamine transporter 2 (VMAT2) in midbrain LepR neurons (KO mice) disrupted DA accumulation in vesicles, but failed to cause a significant change in the evoked release of either glutamate or GABA to downstream neurons. While KO mice showed no differences on chow, they presented a reduced high-fat diet (HFD) intake and resisted to HFD-induced obesity. Specific activation of midbrain LepR neurons promoted VMAT2-dependent feeding on chow and HFD. When tested with an intermittent access to HFD where first 2.5-h HFD eating (binge-like) and 24-h HFD feeding were measured, KO mice exhibited more binge-like, but less 24-h HFD feeding. Interestingly, leptin inhibited 24-h HFD feeding in controls but not in KO mice. Thus, VMAT2-mediated neurotransmission from midbrain LepR neurons contributes to both binge-like eating and HFD feeding regulation.


Feeding Behavior/physiology , Mesencephalon/metabolism , Neurons/metabolism , Receptors, Leptin/metabolism , Synaptic Transmission/physiology , Vesicular Monoamine Transport Proteins/metabolism , Animals , Bulimia/metabolism , Bulimia/pathology , Diet, High-Fat , Disease Models, Animal , Disease Susceptibility/metabolism , Dopamine/metabolism , Female , Glutamic Acid/metabolism , Leptin/administration & dosage , Leptin/metabolism , Male , Mesencephalon/cytology , Mesencephalon/pathology , Mice, Transgenic , Neurons/cytology , Neurons/pathology , Obesity/metabolism , Obesity/pathology , Tissue Culture Techniques , Vesicular Monoamine Transport Proteins/genetics , gamma-Aminobutyric Acid/metabolism
7.
J Neurosci ; 35(29): 10440-50, 2015 Jul 22.
Article En | MEDLINE | ID: mdl-26203139

The hypothalamus is critical for feeding and body weight regulation. Prevailing studies focus on hypothalamic neurons that are defined by selectively expressing transcription factors or neuropeptides including those expressing proopiomelanocortin (POMC) and agouti-related peptides (AgRP). The Cre expression driven by the pancreas-duodenum homeobox 1 promoter is abundant in several hypothalamic nuclei but not in AgRP or POMC neurons. Using this line, we generated mice with disruption of GABA release from a major subset of non-POMC, non-AgRP GABAergic neurons in the hypothalamus. These mice exhibited a reduction in postweaning feeding and growth, and disrupted hyperphagic responses to NPY. Disruption of GABA release severely diminished GABAergic input to the paraventricular hypothalamic nucleus (PVH). Furthermore, disruption of GABA-A receptor function in the PVH also reduced postweaning feeding and blunted NPY-induced hyperphagia. Given the limited knowledge on postweaning feeding, our results are significant in identifying GABA release from a major subset of less appreciated hypothalamic neurons as a key mediator for postweaning feeding and NPY hyperphagia, and the PVH as one major downstream site that contributes significantly to the GABA action. Significance statement: Prevalent studies on feeding in the hypothalamus focus on well characterized, selective groups neurons [e.g., proopiomelanocortin (POMC) and agouti-related peptide (AgRP) neurons], and as a result, the role of the majority of other hypothalamic neurons is largely neglected. Here, we demonstrated an important role for GABAergic projections from non-POMC non-AgRP neurons to the paraventricular hypothalamic nucleus in promoting postweaning (mainly nocturnal) feeding and mediating NPY-induced hyperphagia. Thus, these results signify an importance to study those yet to be defined hypothalamic neurons in the regulation of energy balance and reveal a neural basis for postweaning (nocturnal) feeding and NPY-mediated hyperphagia.


Feeding Behavior/physiology , GABAergic Neurons/physiology , Hyperphagia/physiopathology , Paraventricular Hypothalamic Nucleus/physiology , Agouti-Related Protein/biosynthesis , Animals , Hypothalamus/cytology , Hypothalamus/physiology , In Situ Hybridization , Mice , Mice, Mutant Strains , Neuropeptide Y/metabolism , Organ Culture Techniques , Paraventricular Hypothalamic Nucleus/cytology , Patch-Clamp Techniques , Pro-Opiomelanocortin/biosynthesis
8.
J Neurosci ; 35(8): 3312-8, 2015 Feb 25.
Article En | MEDLINE | ID: mdl-25716832

Lesions of the lateral hypothalamus (LH) cause hypophagia. However, activation of glutamatergic neurons in LH inhibits feeding. These results suggest a potential importance for other LH neurons in stimulating feeding. Our current study in mice showed that disruption of GABA release from adult LH GABAergic neurons reduced feeding. LH GABAergic neurons project extensively to the paraventricular hypothalamic nucleus (PVH), and optogenetic stimulation of GABAergic LH → PVH fibers induced monosynaptic IPSCs in PVH neurons, and potently increased feeding, which depended on GABA release. In addition, disruption of GABA-A receptors in the PVH reduced feeding. Thus, we have identified a new feeding pathway in which GABAergic projections from the LH to the PVH promote feeding.


Eating/physiology , GABAergic Neurons/physiology , Hypothalamic Area, Lateral/physiology , Paraventricular Hypothalamic Nucleus/physiology , Animals , GABAergic Neurons/metabolism , Hypothalamic Area, Lateral/cytology , Inhibitory Postsynaptic Potentials , Mice , Neural Pathways/cytology , Neural Pathways/physiology , Paraventricular Hypothalamic Nucleus/cytology
9.
PLoS One ; 9(4): e93257, 2014.
Article En | MEDLINE | ID: mdl-24695574

ATP6V0C is the bafilomycin A1-binding subunit of vacuolar ATPase, an enzyme complex that critically regulates vesicular acidification. We and others have shown previously that bafilomycin A1 regulates cell viability, autophagic flux and metabolism of proteins that accumulate in neurodegenerative disease. To determine the importance of ATP6V0C for autophagy-lysosome pathway function, SH-SY5Y human neuroblastoma cells differentiated to a neuronal phenotype were nucleofected with non-target or ATP6V0C siRNA and following recovery were treated with either vehicle or bafilomycin A1 (0.3-100 nM) for 48 h. ATP6V0C knockdown was validated by quantitative RT-PCR and by a significant decrease in Lysostracker Red staining. ATP6V0C knockdown significantly increased basal levels of microtubule-associated protein light chain 3-II (LC3-II), α-synuclein high molecular weight species and APP C-terminal fragments, and inhibited autophagic flux. Enhanced LC3 and LAMP-1 co-localization following knockdown suggests that autophagic flux was inhibited in part due to lysosomal degradation and not by a block in vesicular fusion. Knockdown of ATP6V0C also sensitized cells to the accumulation of autophagy substrates and a reduction in neurite length following treatment with 1 nM bafilomycin A1, a concentration that did not produce such alterations in non-target control cells. Reduced neurite length and the percentage of propidium iodide-positive dead cells were also significantly greater following treatment with 3 nM bafilomycin A1. Together these results indicate a role for ATP6V0C in maintaining constitutive and stress-induced ALP function, in particular the metabolism of substrates that accumulate in age-related neurodegenerative disease and may contribute to disease pathogenesis.


Autophagy/physiology , Lysosomes/physiology , Neuroblastoma/metabolism , Neurodegenerative Diseases/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Amyloid beta-Protein Precursor/metabolism , Cell Line, Tumor , Humans , Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism , Neuroblastoma/physiopathology , Neurodegenerative Diseases/physiopathology , alpha-Synuclein/metabolism
10.
J Neurosci ; 33(23): 9592-600, 2013 Jun 05.
Article En | MEDLINE | ID: mdl-23739956

Accumulation of α-synuclein (α-syn) in the brain is a core feature of Parkinson disease (PD) and leads to microglial activation, production of inflammatory cytokines and chemokines, T-cell infiltration, and neurodegeneration. Here, we have used both an in vivo mouse model induced by viral overexpression of α-syn as well as in vitro systems to study the role of the MHCII complex in α-syn-induced neuroinflammation and neurodegeneration. We find that in vivo, expression of full-length human α-syn causes striking induction of MHCII expression by microglia, while knock-out of MHCII prevents α-syn-induced microglial activation, antigen presentation, IgG deposition, and the degeneration of dopaminergic neurons. In vitro, treatment of microglia with aggregated α-syn leads to activation of antigen processing and presentation of antigen sufficient to drive CD4 T-cell proliferation and to trigger cytokine release. These results indicate a central role for microglial MHCII in the activation of both the innate and adaptive immune responses to α-syn in PD and suggest that the MHCII signaling complex may be a target of neuroprotective therapies for the disease.


CD4-Positive T-Lymphocytes/physiology , Dopaminergic Neurons/metabolism , Genes, MHC Class II/physiology , Microglia/metabolism , Nerve Degeneration/metabolism , alpha-Synuclein/biosynthesis , Animals , Animals, Newborn , Cell Proliferation , Cells, Cultured , Dopaminergic Neurons/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology
11.
ACS Chem Neurosci ; 3(12): 1063-72, 2012 Dec 19.
Article En | MEDLINE | ID: mdl-23259041

Rotenone, which selectively inhibits mitochondrial complex I, induces oxidative stress, α-synuclein accumulation, and dopaminergic neuron death, principal pathological features of Parkinson's disease. The autophagy-lysosome pathway degrades damaged proteins and organelles for the intracellular maintenance of nutrient and energy balance. While it is known that rotenone causes autophagic vacuole accumulation, the mechanism by which this effect occurs has not been thoroughly investigated. Treatment of differentiated SH-SY5Y cells with rotenone (10 µM) induced the accumulation of autophagic vacuoles at 6 h and 24 h as indicated by Western blot analysis for microtubule associated protein-light chain 3-II (MAP-LC3-II). Assessment of autophagic flux at these time points indicated that autophagic vacuole accumulation resulted from a decrease in their effective lysosomal degradation, which was substantiated by increased levels of autophagy substrates p62 and α-synuclein. Inhibition of lysosomal degradation may be explained by the observed decrease in cellular ATP levels, which in turn may have caused the observed concomitant increase in acidic vesicle pH. The early (6 h) effects of rotenone on cellular energetics and autophagy-lysosome pathway function preceded the induction of cell death and apoptosis. These findings indicate that the classical mitochondrial toxin rotenone has a pronounced effect on macroautophagy completion that may contribute to its neurotoxic potential.


Autophagy/drug effects , Cell Death/drug effects , Neurons/drug effects , Rotenone/pharmacology , Uncoupling Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Neurons/metabolism , Neurons/pathology , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/pathology
...