Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Int J Mol Sci ; 24(4)2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36834616

ABSTRACT

Molecular profiling of the hypothalamus in response to metabolic shifts is a critical cue to better understand the principle of the central control of whole-body energy metabolism. The transcriptional responses of the rodent hypothalamus to short-term calorie restriction have been documented. However, studies on the identification of hypothalamic secretory factors that potentially contribute to the control of appetite are lacking. In this study, we analyzed the differential expression of hypothalamic genes and compared the selected secretory factors from the fasted mice with those of fed control mice using bulk RNA-sequencing. We verified seven secretory genes that were significantly altered in the hypothalamus of fasted mice. In addition, we determined the response of secretory genes in cultured hypothalamic cells to treatment with ghrelin and leptin. The current study provides further insights into the neuronal response to food restriction at the molecular level and may be useful for understanding the hypothalamic control of appetite.


Subject(s)
Hypothalamus , Starvation , Mice , Animals , Hypothalamus/metabolism , Leptin/metabolism , Starvation/metabolism , Appetite/physiology , Fasting/physiology , Ghrelin/metabolism , Gene Expression Profiling
2.
Aging Cell ; 20(4): e13342, 2021 04.
Article in English | MEDLINE | ID: mdl-33724708

ABSTRACT

One of the most fundamental challenges for all living organisms is to sense and respond to alternating nutritional conditions in order to adapt their metabolism and physiology to promote survival and achieve balanced growth. Here, we applied metabolomics and lipidomics to examine temporal regulation of metabolism during starvation in wild-type Caenorhabditis elegans and in animals lacking the transcription factor HLH-30. Our findings show for the first time that starvation alters the abundance of hundreds of metabolites and lipid species in a temporal- and HLH-30-dependent manner. We demonstrate that premature death of hlh-30 animals under starvation can be prevented by supplementation of exogenous fatty acids, and that HLH-30 is required for complete oxidation of long-chain fatty acids. We further show that RNAi-mediated knockdown of the gene encoding carnitine palmitoyl transferase I (cpt-1) only impairs survival of wild-type animals and not of hlh-30 animals. Strikingly, we also find that compromised generation of peroxisomes by prx-5 knockdown renders hlh-30 animals hypersensitive to starvation, which cannot be rescued by supplementation of exogenous fatty acids. Collectively, our observations show that mitochondrial functions are compromised in hlh-30 animals and that hlh-30 animals rewire their metabolism to largely depend on functional peroxisomes during starvation, underlining the importance of metabolic plasticity to maintain survival.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Mitochondria/metabolism , Signal Transduction/genetics , Starvation/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Dietary Supplements , Fatty Acids/administration & dosage , Fatty Acids/metabolism , Gene Knockdown Techniques , Longevity/genetics , Mutation , Oxidation-Reduction , Peroxisomes/metabolism , RNA Interference , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Starvation/genetics
3.
PLoS One ; 15(3): e0230156, 2020.
Article in English | MEDLINE | ID: mdl-32134989

ABSTRACT

Kampo, a system of traditional Japanese therapy utilizing mixtures of herbal medicine, is widely accepted in the Japanese medical system. Kampo originated from traditional Chinese medicine, and was gradually adopted into a Japanese style. Although its effects on a variety of diseases are appreciated, the underlying mechanisms remain mostly unclear. Using a quantitative tf-LC3 system, we conducted a high-throughput screen of 128 kinds of Kampo to evaluate the effects on autophagy. The results revealed a suppressive effect of Shigyakusan/TJ-35 on autophagic activity. TJ-35 specifically suppressed dephosphorylation of ULK1 and TFEB, among several TORC1 substrates, in response to nutrient deprivation. TFEB was dephosphorylated by calcineurin in a Ca2+ dependent manner. Cytosolic Ca2+ concentration was increased in response to nutrient starvation, and TJ-35 suppressed this increase. Thus, TJ-35 prevents the starvation-induced Ca2+ increase, thereby suppressing induction of autophagy.


Subject(s)
Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/drug effects , Drugs, Chinese Herbal/pharmacology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Calcineurin/metabolism , Calcium/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphorylation , Starvation/metabolism
4.
Physiol Rep ; 8(1): e14338, 2020 01.
Article in English | MEDLINE | ID: mdl-31925945

ABSTRACT

We previously showed that 2 weeks of a severe food restricted (sFR) diet (40% of the caloric intake of the control (CT) diet) up-regulated the circulating renin angiotensin (Ang) system (RAS) in female Fischer rats, most likely as a result of the fall in plasma volume. In this study, we investigated the role of the central RAS in the mean arterial pressure (MAP) and heart rate (HR) dysregulation associated with sFR. Although sFR reduced basal mean MAP and HR, the magnitude of the pressor response to intracerebroventricular (icv) microinjection of Ang-[1-8] was not affected; however, HR was 57 ± 13 bpm lower 26 min after Ang-[1-8] microinjection in the sFR rats and a similar response was observed after losartan was microinjected. The major catabolic pathway of Ang-[1-8] in the hypothalamus was via Ang-[1-7]; however, no differences were detected in the rate of Ang-[1-8] synthesis or degradation between CT and sFR animals. While sFR had no effect on the AT1 R binding in the subfornical organ (SFO), the organum vasculosum laminae terminalis (OVLT) and median preoptic nucleus (MnPO) of the paraventricular anteroventral third ventricle, ligand binding increased 1.4-fold in the paraventricular nucleus (PVN) of the hypothalamus. These findings suggest that sFR stimulates the central RAS by increasing AT1 R expression in the PVN as a compensatory response to the reduction in basal MAP and HR. These findings have implications for people experiencing a period of sFR since an activated central RAS could increase their risk of disorders involving over activation of the RAS including renal and cardiovascular diseases.


Subject(s)
Angiotensin I/metabolism , Arterial Pressure/physiology , Caloric Restriction , Heart Rate/physiology , Hypothalamus/metabolism , Peptide Fragments/metabolism , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System/physiology , Starvation/metabolism , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme 2/metabolism , Animals , Arterial Pressure/drug effects , Autoradiography , Female , Heart Rate/drug effects , Injections, Intraventricular , Losartan/pharmacology , Organum Vasculosum/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Peptide Fragments/pharmacology , Peptidyl-Dipeptidase A/metabolism , Preoptic Area/metabolism , Rats , Rats, Inbred F344 , Renin-Angiotensin System/drug effects , Subfornical Organ/metabolism
5.
Biosci Trends ; 11(5): 557-564, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-29033400

ABSTRACT

Cardiomyocyte nutrient deprivation is a common clinical event that mediates various cardiac ischemic processes and is associated with autophagy activation and cell survival or death. Luteolin-7-O-glucoside (LUTG) was one of the flavonoid glycosides isolated from Dracocephalum tanguticum. Previous research had showed that LUTG pretreatment had significant protective effects against doxorubicin-induced cardiotoxicity. However, whether LUTG could protect cardiomyocytes from starvation-induced injury was not clear. In this study, cardioprotection and mechanisms of LUTG against starvation-induced injury were investigated in vitro. 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2-tetrazolium bromide (MTT) assay showed starvation-induced autophagy is a homeostatic and protective response for H9c2 cell survival. LUTG could protect against injury induced by starvation in H9c2 cells. Acridine orange (AO) staining showed that pretreatment with LUTG enhanced lysosomal autophagy. Western blotting indicated that LUTG enhanced autophagy by down-regulating the expression of phospho-extracellular signal regulated kinase1/2 (p-ERK), phospho-protein kinase B (p-Akt) and phospho-mammalian target of rapamycin (p-mTOR). These results suggest that LUTG might act as a promising therapeutic agent for preventing starvation-induced cardiotoxicity by upregulation of autophagy through the Akt/mTOR and ERK signal pathway.


Subject(s)
Autophagy/drug effects , Cardiotonic Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Flavones/pharmacology , Glucosides/pharmacology , Myocytes, Cardiac/drug effects , Starvation/pathology , Animals , Cardiotonic Agents/isolation & purification , Cell Culture Techniques , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Survival/drug effects , Drugs, Chinese Herbal/isolation & purification , Flavones/isolation & purification , Glucosides/isolation & purification , Lamiaceae/chemistry , MAP Kinase Signaling System/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Starvation/metabolism
6.
PLoS One ; 12(2): e0172917, 2017.
Article in English | MEDLINE | ID: mdl-28235079

ABSTRACT

SLC38A9 is characterized as a lysosomal component of the amino acid sensing Ragulator-RAG GTPase complex, controlling the mechanistic target of rapamycin complex 1 (mTORC1). Here, immunohistochemistry was used to map SLC38A9 in mouse brain and staining was detected throughout the brain, in cortex, hypothalamus, thalamus, hippocampus, brainstem and cerebellum. More specifically, immunostaining was found in areas known to be involved in amino acid sensing and signaling pathways e.g. piriform cortex and hypothalamus. SLC38A9 immunoreactivity co-localized with both GABAergic and glutamatergic neurons, but not with astrocytes. SLC38A9 play a key role in the mTORC1 pathway, and therefore we performed in vivo starvation and high-fat diet studies, to measure gene expression alterations in specific brain tissues and in larger brain regions. Following starvation, Slc38a9 was upregulated in brainstem and cortex, and in anterior parts of the brain (Bregma 3.2 to -2.1mm). After high-fat diet, Slc38a9 was specifically upregulated in hypothalamus, while overall downregulation was noticed throughout the brain (Bregma 3.2 to -8.6mm).


Subject(s)
Amino Acid Transport Systems/metabolism , Hypothalamus/metabolism , Amino Acid Transport Systems/genetics , Animals , Brain/metabolism , Diet, High-Fat , Gene Expression , Male , Mice, Inbred C57BL , Starvation/metabolism , Up-Regulation
7.
J Clin Invest ; 126(12): 4727-4734, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27906690

ABSTRACT

Leptin is an adipose tissue hormone that functions as an afferent signal in a negative feedback loop that maintains homeostatic control of adipose tissue mass. This endocrine system thus serves a critical evolutionary function by protecting individuals from the risks associated with being too thin (starvation) or too obese (predation and temperature dysregulation). Mutations in leptin or its receptor cause massive obesity in mice and humans, and leptin can effectively treat obesity in leptin-deficient patients. Leptin acts on neurons in the hypothalamus and elsewhere to elicit its effects, and mutations that affect the function of this neural circuit cause Mendelian forms of obesity. Leptin levels fall during starvation and elicit adaptive responses in many other physiologic systems, the net effect of which is to reduce energy expenditure. These effects include cessation of menstruation, insulin resistance, alterations of immune function, and neuroendocrine dysfunction, among others. Some or all of these effects are also seen in patients with constitutively low leptin levels, such as occur in lipodystrophy. Leptin is an approved treatment for generalized lipodystrophy, a condition associated with severe metabolic disease, and has also shown potential for the treatment of other types of diabetes. In addition, leptin restores reproductive capacity and increases bone mineral density in patients with hypothalamic amenorrhea, an infertility syndrome in females. Most obese patients have high endogenous levels of leptin, in some instances as a result of mutations in the neural circuit on which leptin acts, though in most cases, the pathogenesis of leptin resistance is not known. Obese patients with leptin resistance show a variable response to exogenous leptin but may respond to a combination of leptin plus amylin. Overall, the identification of leptin has provided a framework for studying the pathogenesis of obesity in the general population, clarified the nature of the biologic response to starvation, and helped to advance our understanding of the neural mechanisms that control feeding.


Subject(s)
Adipose Tissue , Diabetes Mellitus , Leptin , Lipodystrophy , Obesity , Starvation , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Diabetes Mellitus/drug therapy , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Leptin/genetics , Leptin/metabolism , Leptin/therapeutic use , Lipodystrophy/drug therapy , Lipodystrophy/genetics , Lipodystrophy/metabolism , Lipodystrophy/pathology , Mice , Neurons/metabolism , Neurons/pathology , Obesity/drug therapy , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Starvation/drug therapy , Starvation/genetics , Starvation/metabolism , Starvation/pathology
8.
Endocrinology ; 157(8): 3253-65, 2016 08.
Article in English | MEDLINE | ID: mdl-27323240

ABSTRACT

Hypothalamic-pituitary-thyroid (HPT) axis activity is important for energy homeostasis, and is modified by stress. Maternal separation (MS) alters the stress response and predisposes to metabolic disturbances in the adult. We therefore studied the effect of MS on adult HPT axis activity. Wistar male and female pups were separated from their mothers 3 h/d during postnatal day (PND)2-PND21 (MS), or left nonhandled (NH). Open field and elevated plus maze tests revealed increased locomotion in MS males and anxiety-like behavior in MS females. At PND90, MS females had increased body weight gain, Trh expression in the hypothalamic paraventricular nucleus, and white adipose tissue mass. MS males had increased expression of TRH-degrading enzyme in tanycytes, reduced TSH and T3, and enhanced corticosterone serum concentrations. MS stimulated brown adipose tissue deiodinase 2 activity in either sex. Forty-eight hours of fasting (PND60) augmented serum corticosterone levels similarly in MS or NH females but more in MS than in NH male rats. MS reduced the fasting-induced drop in hypothalamic paraventricular nucleus-Trh expression of males but not of females and abolished the fasting-induced increase in Trh expression in both sexes. Fasting reduced serum concentrations of TSH, T4, and T3, less in MS than in NH males, whereas in females, TSH decreased in MS but not in NH rats, but T4 and T3 decreased similarly in NH and MS rats. In conclusion, MS produced long-term changes in the activity of the HPT axis that were sex specific; response to fasting was partially blunted in males, which could affect their adaptive response to negative energy balance.


Subject(s)
Aminopeptidases/genetics , Hypothalamus/metabolism , Maternal Deprivation , Pyrrolidonecarboxylic Acid/analogs & derivatives , Starvation/physiopathology , Thyroid Gland/physiology , Thyrotropin-Releasing Hormone/genetics , Aminopeptidases/metabolism , Animals , Animals, Newborn , Female , Male , Pyrrolidonecarboxylic Acid/metabolism , Rats , Rats, Wistar , Sex Characteristics , Starvation/genetics , Starvation/metabolism , Thyrotropin-Releasing Hormone/metabolism
9.
J Biol Chem ; 289(32): 21909-25, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24891504

ABSTRACT

Loss of muscle proteins and the consequent weakness has important clinical consequences in diseases such as cancer, diabetes, chronic heart failure, and in aging. In fact, excessive proteolysis causes cachexia, accelerates disease progression, and worsens life expectancy. Muscle atrophy involves a common pattern of transcriptional changes in a small subset of genes named atrophy-related genes or atrogenes. Whether microRNAs play a role in the atrophy program and muscle loss is debated. To understand the involvement of miRNAs in atrophy we performed miRNA expression profiling of mouse muscles under wasting conditions such as fasting, denervation, diabetes, and cancer cachexia. We found that the miRNA signature is peculiar of each catabolic condition. We then focused on denervation and we revealed that changes in transcripts and microRNAs expression did not occur simultaneously but were shifted. Indeed, whereas transcriptional control of the atrophy-related genes peaks at 3 days, changes of miRNA expression maximized at 7 days after denervation. Among the different miRNAs, microRNA-206 and -21 were the most induced in denervated muscles. We characterized their pattern of expression and defined their role in muscle homeostasis. Indeed, in vivo gain and loss of function experiments revealed that miRNA-206 and miRNA-21 were sufficient and required for atrophy program. In silico and in vivo approaches identified transcription factor YY1 and the translational initiator factor eIF4E3 as downstream targets of these miRNAs. Thus miRNAs are important for fine-tuning the atrophy program and their modulation can be a novel potential therapeutic approach to counteract muscle loss and weakness in catabolic conditions.


Subject(s)
MicroRNAs/genetics , Muscular Atrophy/etiology , Muscular Atrophy/genetics , 3' Untranslated Regions , Animals , Base Sequence , Cachexia/genetics , Cachexia/metabolism , Disease Models, Animal , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Profiling , Male , Mice , Mice, Inbred BALB C , MicroRNAs/metabolism , Molecular Sequence Data , Muscle Denervation , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Starvation/genetics , Starvation/metabolism , Time Factors , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
10.
J Integr Bioinform ; 11(1): 235, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24675236

ABSTRACT

Our understanding of complex biological processes can be enhanced by combining different kinds of high-throughput experimental data, but the use of incompatible identifiers makes data integration a challenge. We aimed to improve methods for integrating and visualizing different types of omics data. To validate these methods, we applied them to two previous studies on starvation in mice, one using proteomics and the other using transcriptomics technology. We extended the PathVisio software with new plugins to link proteins, transcripts and pathways. A low overall correlation between proteome and transcriptome data was detected (Spearman rank correlation: 0.21). At the level of individual genes, correlation was highly variable. Many mRNA/protein pairs, such as fructose biphosphate aldolase B and ATP Synthase, show good correlation. For other pairs, such as ferritin and elongation factor 2, an interesting effect is observed, where mRNA and protein levels change in opposite directions, suggesting they are not primarily regulated at the transcriptional level. We used pathway diagrams to visualize the integrated datasets and found it encouraging that transcriptomics and proteomics data supported each other at the pathway level. Visualization of the integrated dataset on pathways led to new observations on gene-regulation in the response of the gut to starvation. Our methods are generic and can be applied to any multi-omics study. The PathVisio software can be obtained at http://www.pathvisio.org. Supplemental data are available at http://www.bigcat.unimaas.nl/data/jib-supplemental/ , including instructions on reproducing the pathway visualizations of this manuscript.


Subject(s)
Genomics/methods , Intestinal Mucosa/metabolism , Starvation/genetics , Starvation/metabolism , Amino Acids/metabolism , Animals , Intestines/pathology , Male , Metabolic Networks and Pathways , Mice , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
J Biol Chem ; 288(50): 36073-82, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24187137

ABSTRACT

Cells turn on autophagy, an intracellular recycling pathway, when deprived of nutrients. How autophagy is regulated by hormonal signals in response to major changes in metabolic state is not well understood. Here, we provide evidence that myonectin (CTRP15), a skeletal muscle-derived myokine, is a novel regulator of cellular autophagy. Starvation activated liver autophagy, whereas nutrient supplementation following food deprivation suppressed it; the former and latter correlated with reduced and increased expression and circulating levels of myonectin, respectively, suggestive of a causal link. Indeed, recombinant myonectin administration suppressed starvation-induced autophagy in mouse liver and cultured hepatocytes, as indicated by the inhibition of LC3-dependent autophagosome formation, p62 degradation, and expression of critical autophagy-related genes. Reduction in protein degradation is mediated by the PI3K/Akt/mTOR signaling pathway; inhibition of this pathway abrogated the ability of myonectin to suppress autophagy in cultured hepatocytes. Together, our results reveal a novel skeletal muscle-liver axis controlling cellular autophagy, underscoring the importance of hormone-mediated tissue cross-talk in maintaining energy homeostasis.


Subject(s)
Autophagy/drug effects , Cytokines/pharmacology , Liver/cytology , Muscle Proteins/pharmacology , Muscle, Skeletal/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Dietary Supplements , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Humans , Male , Mice , Microtubule-Associated Proteins/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Recombinant Proteins/pharmacology , Starvation/metabolism , Starvation/pathology , Transcription Factor TFIIH , Transcription Factors/metabolism
12.
J Comp Physiol B ; 183(8): 1039-52, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23925409

ABSTRACT

Most fasting animals are believed to sequentially switch from predominantly utilizing one metabolic substrate to another from carbohydrates, to lipids, then to proteins. The timing of these physiological transitions has been estimated using measures of substrate oxidation including changes in respiratory exchange ratios, blood metabolites, nitrogen excretion, or enzyme activities in tissues. Here, we demonstrate how (13)CO2-breath testing can be used to partition among the oxidation of distinct nutrient pools in the body (i.e., carbohydrates, lipids, and proteins) that have become artificially enriched in (13)C. Seventy-two Swiss Webster mice were raised to adulthood on diets supplemented with (13)C-1-L-leucine, (13)C-1-palmitic acid, (13)C-1-D-glucose, or no tracer. Mice were then fasted for 72 h during which [Formula: see text], [Formula: see text], δ(13)C of exhaled CO2, body temperature, body mass, and blood metabolites (i.e., glucose, ketone bodies, and triacylglycerols) were measured. The fasting mice exhibited reductions in body mass (29 %), body temperature (3.3 °C), minimum observed metabolic rates (24 %), and respiratory exchange ratio (0.18), as well as significant changes in blood metabolites; but these responses were not particularly indicative of changes in oxidative fuel mixture. Measurements of endogenous nutrient oxidation by way of (13)CO2-breath testing revealed a decrease in the rate of oxidation of carbohydrates from 61 to 10 % of the total energy expenditure during the first 6 h without food. This response was mirrored by a coincidental increase in rate of endogenous lipid oxidation from 18 to 64 %. A transient peak in carbohydrate oxidation occurred between 8 and 14 h, presumably during increased glycogen mobilization. A well-defined period of protein sparing between 8 and 12 h was observed where endogenous protein oxidation accounted for as little as 8 % of the total energy expenditure. Thereafter, protein oxidation continually increased accounting for as much as 24 % of the total energy expenditure by 72 h. This study demonstrates that (13)CO2-breath testing may provide a complementary approach to characterizing the timing and magnitude of sequential changes in substrate oxidation that occur during prolonged fasting and starvation.


Subject(s)
Carbon Dioxide/metabolism , Carbon Isotopes/metabolism , Energy Metabolism/physiology , Starvation/physiopathology , Analysis of Variance , Animals , Body Temperature , Body Weight , Breath Tests , Carbohydrate Metabolism/physiology , Glucose , Leucine , Lipid Metabolism/physiology , Mice , Oxidation-Reduction , Palmitic Acid , Proteins/metabolism , Starvation/metabolism
13.
Nat Med ; 19(9): 1153-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23933983

ABSTRACT

Preventing reproduction during nutritional deprivation is an adaptive process that is conserved and essential for the survival of species. In mammals, the mechanisms that inhibit fertility during starvation are complex and incompletely understood. Here we show that exposure of female mice to fibroblast growth factor 21 (FGF21), a fasting-induced hepatokine, mimics infertility secondary to starvation. Mechanistically, FGF21 acts on the suprachiasmatic nucleus (SCN) in the hypothalamus to suppress the vasopressin-kisspeptin signaling cascade, thereby inhibiting the proestrus surge in luteinizing hormone. Mice lacking the FGF21 co-receptor, ß-Klotho, in the SCN are refractory to the inhibitory effect of FGF21 on female fertility. Thus, FGF21 defines an important liver-neuroendocrine axis that modulates female reproduction in response to nutritional challenge.


Subject(s)
Fibroblast Growth Factors/metabolism , Infertility, Female/metabolism , Membrane Proteins/metabolism , Reproduction , Starvation/metabolism , Animals , Energy Metabolism , Female , Hypothalamus , Kisspeptins/antagonists & inhibitors , Kisspeptins/metabolism , Klotho Proteins , Luteinizing Hormone/biosynthesis , Luteinizing Hormone/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Proestrus/physiology , Signal Transduction , Suprachiasmatic Nucleus , Vasopressins/antagonists & inhibitors , Vasopressins/metabolism
14.
PLoS One ; 8(5): e63886, 2013.
Article in English | MEDLINE | ID: mdl-23704949

ABSTRACT

Rhodiola rosea has been extensively used to improve physical and mental performance and to protect against stress. We, and others, have reported that R. rosea can extend lifespan in flies, worms, and yeast. However, its molecular mechanism is currently unknown. Here, we tested whether R. rosea might act through a pathway related to dietary restriction (DR) that can extend lifespan in a range of model organisms. While the mechanism of DR itself is also unknown, three molecular pathways have been associated with it: the silent information regulator 2 (SIR2) proteins, insulin and insulin-like growth factor signaling (IIS), and the target of rapamycin (TOR). In flies, DR is implemented through a reduction in dietary yeast content. We found that R. rosea extract extended lifespan in both sexes independent of the yeast content in the diet. We also found that the extract extended lifespan when the SIR2, IIS, or TOR pathways were genetically perturbed. Upon examination of water and fat content, we found that R. rosea decreased water content and elevated fat content in both sexes, but did not sensitize flies to desiccation or protect them against starvation. There were some sex-specific differences in response to R. rosea. In female flies, the expression levels of glycolytic genes and dSir2 were down-regulated, and NADH levels were decreased. In males however, R. rosea provided no protection against heat stress and had no effect on the major heat shock protein HSP70 and actually down-regulated the mitochondrial HSP22. Our findings largely rule out an elevated general resistance to stress and DR-related pathways as mechanistic candidates. The latter conclusion is especially relevant given the limited potential for DR to improve human health and lifespan, and presents R. rosea as a potential viable candidate to treat aging and age-related diseases in humans.


Subject(s)
Caloric Restriction , Drosophila melanogaster/physiology , Longevity/drug effects , Plant Extracts/pharmacology , Rhodiola/chemistry , Adaptation, Physiological/drug effects , Animals , Desiccation , Down-Regulation/drug effects , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Female , Glycolysis/drug effects , Glycolysis/genetics , Hot Temperature , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Lipid Metabolism/drug effects , Male , NAD/metabolism , Peptides/metabolism , Sex Characteristics , Signal Transduction/drug effects , Solubility , Starvation/metabolism , Water/metabolism , Yeasts
15.
Nat Commun ; 4: 1862, 2013.
Article in English | MEDLINE | ID: mdl-23673647

ABSTRACT

Hypothalamic feeding circuits are essential for the maintenance of energy balance. There have been intensive efforts to discover new biological molecules involved in these pathways. Here we report that central administration of clusterin, also called apolipoprotein J, causes anorexia, weight loss and activation of hypothalamic signal transduction-activated transcript-3 in mice. In contrast, inhibition of hypothalamic clusterin action results in increased food intake and body weight, leading to adiposity. These effects are likely mediated through the mutual actions of the low-density lipoprotein receptor-related protein-2, a potential receptor for clusterin, and the long-form leptin receptor. In response to clusterin, the low-density lipoprotein receptor-related protein-2 binding to long-form leptin receptor is greatly enhanced in cultured neuronal cells. Furthermore, long-form leptin receptor deficiency or hypothalamic low-density lipoprotein receptor-related protein-2 suppression in mice leads to impaired hypothalamic clusterin signalling and actions. Our study identifies the hypothalamic clusterin-low-density lipoprotein receptor-related protein-2 axis as a novel anorexigenic signalling pathway that is tightly coupled with long-form leptin receptor-mediated signalling.


Subject(s)
Clusterin/metabolism , Feeding Behavior , Hypothalamus/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Animals , Anorexia/complications , Anorexia/metabolism , Anorexia/pathology , Body Weight/drug effects , Cell Line , Clusterin/administration & dosage , Clusterin/pharmacology , Epididymis/drug effects , Epididymis/metabolism , Feeding Behavior/drug effects , Humans , Hypothalamus/drug effects , Immunohistochemistry , Injections, Intraventricular , Leptin/administration & dosage , Leptin/pharmacology , Male , Mice , Obesity/complications , Obesity/metabolism , Obesity/pathology , Phosphorylation/drug effects , Protein Binding/drug effects , Rats , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Starvation/metabolism
16.
Br J Nutr ; 107(1): 52-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21733331

ABSTRACT

Prolonged starvation changes energy metabolism; therefore, the metabolic response to starvation is divided into three phases according to changes in glucose, lipid and protein utilisation. B-group vitamins are involved in energy metabolism via metabolism of carbohydrates, fatty acids and amino acids. To determine how changes in energy metabolism alter B-group vitamin concentrations during starvation, we measured the concentration of eight kinds of B-group vitamins daily in rat blood, urine and in nine tissues including cerebrum, heart, lung, stomach, kidney, liver, spleen, testis and skeletal muscle during 8 d of starvation. Vitamin B1, vitamin B6, pantothenic acid, folate and biotin concentrations in the blood reduced after 6 or 8 d of starvation, and other vitamins did not change. Urinary excretion was decreased during starvation for all B-group vitamins except pantothenic acid and biotin. Less variation in B-group vitamin concentrations was found in the cerebrum and spleen. Concentrations of vitamin B1, vitamin B6, nicotinamide and pantothenic acid increased in the liver. The skeletal muscle and stomach showed reduced concentrations of five vitamins including vitamin B1, vitamin B2, vitamin B6, pantothenic acid and folate. Concentrations of two or three vitamins decreased in the kidney, testis and heart, and these changes showed different patterns in each tissue and for each vitamin. The concentration of pantothenic acid rapidly decreased in the heart, stomach, kidney and testis, whereas concentrations of nicotinamide were stable in all tissues except the liver. Different variations in B-group vitamin concentrations in the tissues of starved rats were found. The present findings will lead to a suitable supplementation of vitamins for the prevention of the re-feeding syndrome.


Subject(s)
Starvation/metabolism , Vitamin B Complex/metabolism , Animals , Energy Metabolism , Male , Nutritional Status , Organ Size , Organ Specificity , Random Allocation , Rats , Rats, Wistar , Starvation/blood , Starvation/pathology , Starvation/urine , Time Factors , Vitamin B Complex/blood , Vitamin B Complex/urine , Weight Loss
17.
Physiol Res ; 60(Suppl 1): S165-70, 2011.
Article in English | MEDLINE | ID: mdl-21777028

ABSTRACT

Leptin is produced by white adipose tissue and other cell types and is involved in both short- and long-term appetite control. Here we studied effects of starvation on serum, pituitary and hypothalamic levels of leptin during 72 h period. Each of the starved groups was sacrificed simultaneously with the group of ad libitum fed animals. The progression of the discrete starvation response phases was monitored by testing the blood glucose, free fatty acid, urea and corticosterone levels. Starvation caused biphasic increase in corticosterone and free fatty acid levels, and significant but transient decrease in urea and glucose levels. Starvation also abolished diurnal rhythm of changes in leptin concentrations in serum and hypothalamic and pituitary tissues. Only 6 h starving period was sufficient to lock serum leptin at low levels, whereas 12 h were needed to silence leptin production/secretion in hypothalamus for the whole examined period. In contrast, leptin production by pituitary tissues of starved animals required 24 h to reach minimum, followed by full recovery by the end of starvation period. These results indicate the tissue specific pattern of leptin release and suggest that the locally produced leptin could activate its receptor in pituitary cells independently of serum levels of this hormone.


Subject(s)
Energy Metabolism , Hypothalamus/metabolism , Leptin/metabolism , Pituitary Gland/metabolism , Starvation/metabolism , Animals , Biomarkers/blood , Blood Glucose/metabolism , Circadian Rhythm , Corticosterone/blood , Disease Models, Animal , Fatty Acids, Nonesterified/blood , Food Deprivation , Leptin/blood , Male , Rats , Rats, Wistar , Starvation/blood , Starvation/etiology , Time Factors , Urea/blood
18.
J Neurosci ; 31(23): 8373-80, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21653842

ABSTRACT

Nutritional deprivation or malnutrition suppresses immune function in humans and animals, thereby conferring higher susceptibility to infectious diseases. Indeed, nutritional deprivation induces atrophy of lymphoid tissues such as thymus and spleen and decreases the number of circulating lymphocytes. Leptin, a major adipocytokine, is exclusively produced in the adipose tissue in response to the nutritional status and acts on the hypothalamus, thereby regulating energy homeostasis. Although leptin plays a critical role in the starvation-induced T-cell-mediated immunosuppression, little is known about its role in B-cell homeostasis under starvation conditions. Here we show the alteration of B-cell development in the bone marrow of fasted mice, characterized by decrease in pro-B, pre-B, and immature B cells and increase in mature B cells. Interestingly, intracerebroventricular leptin injection was sufficient to prevent the alteration of B-cell development of fasted mice. The alteration of B lineage cells in the bone marrow of fasted mice was markedly prevented by oral administration of glucocorticoid receptor antagonist RU486 (11ß-[p-(dimethylamino)phenyl]-17ß-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one). It was also effectively prevented by intracerebroventricular injection of neuropeptide Y Y(1) receptor antagonist BIBP3226 [(2R)-5-(diaminomethylideneamino)-2-[(2,2-diphenylacetyl)amino]-N-[(4-hydroxyphenyl)methyl]pentanamide], along with suppression of the otherwise increased serum corticosterone concentrations. This study provides the first in vivo evidence for the role of central leptin signaling in the starvation-induced alteration of B-cell development. The data of this study suggest that the CNS, which is inherent to integrate information from throughout the organism, is able to control immune function.


Subject(s)
B-Lymphocytes/metabolism , Cell Differentiation/physiology , Hypothalamus/metabolism , Leptin/metabolism , Signal Transduction/physiology , Starvation/metabolism , Adipose Tissue/drug effects , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adrenalectomy , Animals , B-Lymphocytes/immunology , Blood Glucose , Cell Differentiation/drug effects , Flow Cytometry , Hypothalamus/drug effects , Hypothalamus/immunology , Leptin/pharmacology , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Starvation/immunology
19.
Gene Expr ; 14(5): 279-89, 2009.
Article in English | MEDLINE | ID: mdl-19630271

ABSTRACT

The expression of five genes involved in nitrogen assimilation in cyanobacteria, namely glnA, glsF, icd, ntcA, and glnB, encoding three key enzymes from that pathway (glutamine synthetase, glutamate synthase, isocitrate dehydrogenase) and two regulatory proteins (NtcA and PII), was studied in this work. Their changes under different conditions were analyzed by quantitative real-time RT-PCR. Nutrient limitation induced clear modifications on the expression of most studied genes: lack of nitrogen provoked an initial increase, followed by a marked decrease; in the cases of phosphorus and iron starvation, a general, stronger expression decrease was observed, particularly striking in the case of iron. Darkness and addition of the photosynthethic inhibitors DCMU and DBMIB also had a strong effect on gene expression. Methionine sulfoximine and azaserine, inhibitors of glutamine synthetase and glutamate synthase, respectively, provoked a sharp increase in icd expression. These results, together with previous studies, suggest that 2-oxoglutarate could be the molecule utilized by Prochlorococcus to sense the C/N balance. Besides, our results confirm the different regulation of nitrogen assimilation in Prochlorococcus with regard to other cyanobacteria.


Subject(s)
Carbon/metabolism , Genes, Bacterial , Nitrogen/metabolism , Prochlorococcus/genetics , Electron Transport/genetics , Gene Expression Regulation, Bacterial , Genes, Bacterial/physiology , Iron/metabolism , Iron Deficiencies , Light , Metabolic Networks and Pathways/genetics , Phosphorus/deficiency , Phosphorus/metabolism , Photosynthesis/genetics , Prochlorococcus/metabolism , Quorum Sensing/genetics , Species Specificity , Starvation/genetics , Starvation/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL