Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Hepatol ; 80(3): 467-481, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37972658

ABSTRACT

BACKGROUND & AIMS: Metabolic dysfunction-associated steatohepatitis (MASH) is linked to insulin resistance and type 2 diabetes and marked by hepatic inflammation, microvascular dysfunction, and fibrosis, impairing liver function and aggravating metabolic derangements. The liver homeostatic interactions disrupted in MASH are still poorly understood. We aimed to elucidate the plasticity and changing interactions of non-parenchymal cells associated with advanced MASH. METHODS: We characterized a diet-induced mouse model of advanced MASH at single-cell resolution and validated findings by assaying chromatin accessibility, bioimaging murine and human livers, and via functional experiments in vivo and in vitro. RESULTS: The fibrogenic activation of hepatic stellate cells (HSCs) led to deterioration of a signaling module consisting of the bile acid receptor NR1H4/FXR and HSC-specific GS-protein-coupled receptors (GSPCRs) capable of preserving stellate cell quiescence. Accompanying HSC activation, we further observed the attenuation of HSC Gdf2 expression, and a MASH-associated expansion of a CD207-positive macrophage population likely derived from both incoming monocytes and Kupffer cells. CONCLUSION: We conclude that HSC-expressed NR1H4 and GSPCRs of the healthy liver integrate postprandial cues, which sustain HSC quiescence and, through paracrine signals, overall sinusoidal health. Hence HSC activation in MASH not only drives fibrogenesis but may desensitize the hepatic sinusoid to liver homeostatic signals. IMPACT AND IMPLICATIONS: Homeostatic interactions between hepatic cell types and their deterioration in metabolic dysfunction-associated steatohepatitis are poorly characterized. In our current single cell-resolved study of advanced murine metabolic dysfunction-associated steatohepatitis, we identified a quiescence-associated hepatic stellate cell-signaling module with potential to preserve normal sinusoid function. As expression levels of its constituents are conserved in the human liver, stimulation of the identified signaling module is a promising therapeutic strategy to restore sinusoid function in chronic liver disease.


Subject(s)
Diabetes Mellitus, Type 2 , Fatty Liver , Mice , Humans , Animals , Pericytes/metabolism , Diabetes Mellitus, Type 2/metabolism , Liver/pathology , Signal Transduction , Hepatic Stellate Cells/metabolism , Fatty Liver/metabolism , Liver Cirrhosis/pathology , Growth Differentiation Factor 2/metabolism
2.
Nat Commun ; 13(1): 3956, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35803907

ABSTRACT

ß-Adrenergic signaling is a core regulator of brown adipocyte function stimulating both lipolysis and transcription of thermogenic genes, thereby expanding the capacity for oxidative metabolism. We have used pharmacological inhibitors and a direct activator of lipolysis to acutely modulate the activity of lipases, thereby enabling us to uncover lipolysis-dependent signaling pathways downstream of ß-adrenergic signaling in cultured brown adipocytes. Here we show that induction of lipolysis leads to acute induction of several gene programs and is required for transcriptional regulation by ß-adrenergic signals. Using machine-learning algorithms to infer causal transcription factors, we show that PPARs are key mediators of lipolysis-induced activation of genes involved in lipid metabolism and thermogenesis. Importantly, however, lipolysis also activates the unfolded protein response and regulates the core circadian transcriptional machinery independently of PPARs. Our results demonstrate that lipolysis generates important metabolic signals that exert profound pleiotropic effects on transcription and function of cultured brown adipocytes.


Subject(s)
Adipocytes, Brown , Lipolysis , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Adrenergic Agents/pharmacology , Lipolysis/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Thermogenesis/physiology
3.
J Neurosci ; 41(33): 7148-7159, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34210784

ABSTRACT

Following stroke, the survival of neurons and their ability to reestablish connections is critical to functional recovery. This is strongly influenced by the balance between neuronal excitation and inhibition. In the acute phase of experimental stroke, lethal hyperexcitability can be attenuated by positive allosteric modulation of GABAA receptors (GABAARs). Conversely, in the late phase, negative allosteric modulation of GABAAR can correct the suboptimal excitability and improves both sensory and motor recovery. Here, we hypothesized that octadecaneuropeptide (ODN), an endogenous allosteric modulator of the GABAAR synthesized by astrocytes, influences the outcome of ischemic brain tissue and subsequent functional recovery. We show that ODN boosts the excitability of cortical neurons, which makes it deleterious in the acute phase of stroke. However, if delivered after day 3, ODN is safe and improves motor recovery over the following month in two different paradigms of experimental stroke in mice. Furthermore, we bring evidence that, during the subacute period after stroke, the repairing cortex can be treated with ODN by means of a single hydrogel deposit into the stroke cavity.SIGNIFICANCE STATEMENT Stroke remains a devastating clinical challenge because there is no efficient therapy to either minimize neuronal death with neuroprotective drugs or to enhance spontaneous recovery with neurorepair drugs. Around the brain damage, the peri-infarct cortex can be viewed as a reservoir of plasticity. However, the potential of wiring new circuits in these areas is restrained by a chronic excess of GABAergic inhibition. Here we show that an astrocyte-derived peptide, can be used as a delayed treatment, to safely correct cortical excitability and facilitate sensorimotor recovery after stroke.


Subject(s)
Diazepam Binding Inhibitor/therapeutic use , GABA-A Receptor Agonists/therapeutic use , Neurons/drug effects , Neuropeptides/therapeutic use , Peptide Fragments/therapeutic use , Receptors, GABA-A/drug effects , Stroke/drug therapy , Adult , Animals , Astrocytes/metabolism , Cortical Spreading Depression/physiology , Diazepam Binding Inhibitor/deficiency , Diazepam Binding Inhibitor/physiology , Drug Implants , Evoked Potentials, Somatosensory , Female , GABA-A Receptor Agonists/pharmacology , Humans , Hydrogels , Infarction, Middle Cerebral Artery/drug therapy , Intracranial Thrombosis/drug therapy , Intracranial Thrombosis/etiology , Light , Mice , Mice, Inbred C57BL , N-Methylaspartate/toxicity , Neurons/physiology , Neuropeptides/deficiency , Neuropeptides/physiology , Patch-Clamp Techniques , Peptide Fragments/deficiency , Peptide Fragments/physiology , Rats , Rose Bengal/radiation effects , Rose Bengal/toxicity , Single-Blind Method , Stroke/etiology
4.
Mol Metab ; 44: 101144, 2021 02.
Article in English | MEDLINE | ID: mdl-33346070

ABSTRACT

OBJECTIVES: The skin is the largest sensory organ of the human body and plays a fundamental role in regulating body temperature. However, adaptive alterations in skin functions and morphology have only vaguely been associated with physiological responses to cold stress or sensation of ambient temperatures. We previously found that loss of acyl-CoA-binding protein (ACBP) in keratinocytes upregulates lipolysis in white adipose tissue and alters hepatic lipid metabolism, suggesting a link between epidermal barrier functions and systemic energy metabolism. METHODS: To assess the physiological responses to loss of ACBP in keratinocytes in detail, we used full-body ACBP-/- and skin-specific ACBP-/- knockout mice to clarify how loss of ACBP affects 1) energy expenditure by indirect calorimetry, 2) response to high-fat feeding and a high oral glucose load, and 3) expression of brown-selective gene programs by quantitative PCR in inguinal WAT (iWAT). To further elucidate the role of the epidermal barrier in systemic energy metabolism, we included mice with defects in skin structural proteins (ma/ma Flgft/ft) in these studies. RESULTS: We show that the ACBP-/- mice and skin-specific ACBP-/- knockout mice exhibited increased energy expenditure, increased food intake, browning of the iWAT, and resistance to diet-induced obesity. The metabolic phenotype, including browning of the iWAT, was reversed by housing the mice at thermoneutrality (30 °C) or pharmacological ß-adrenergic blocking. Interestingly, these findings were phenocopied in flaky tail mice (ma/ma Flgft/ft). Taken together, we demonstrate that a compromised epidermal barrier induces a ß-adrenergic response that increases energy expenditure and browning of the white adipose tissue to maintain a normal body temperature. CONCLUSIONS: Our findings show that the epidermal barrier plays a key role in maintaining systemic metabolic homeostasis. Thus, regulation of epidermal barrier functions warrants further attention to understand the regulation of systemic metabolism in further detail.


Subject(s)
Diazepam Binding Inhibitor/genetics , Diazepam Binding Inhibitor/metabolism , Energy Metabolism/physiology , Homeostasis , Skin/metabolism , Adipose Tissue, White/metabolism , Animals , Body Temperature , Energy Metabolism/genetics , Filaggrin Proteins , Intermediate Filament Proteins , Lipid Metabolism , Lipolysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism
5.
Cell Metab ; 33(2): 437-453.e5, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33378646

ABSTRACT

Adipose tissues display a remarkable ability to adapt to the dietary status. Here, we have applied single-nucleus RNA-seq to map the plasticity of mouse epididymal white adipose tissue at single-nucleus resolution in response to high-fat-diet-induced obesity. The single-nucleus approach allowed us to recover all major cell types and to reveal distinct transcriptional stages along the entire adipogenic trajectory from preadipocyte commitment to mature adipocytes. We demonstrate the existence of different adipocyte subpopulations and show that obesity leads to disappearance of the lipogenic subpopulation and increased abundance of the stressed lipid-scavenging subpopulation. Moreover, obesity is associated with major changes in the abundance and gene expression of other cell populations, including a dramatic increase in lipid-handling genes in macrophages at the expense of macrophage-specific genes. The data provide a powerful resource for future hypothesis-driven investigations of the mechanisms of adipocyte differentiation and adipose tissue plasticity.


Subject(s)
Adipose Tissue/metabolism , Obesity/metabolism , Adipogenesis/genetics , Animals , Cell Plasticity , Diet, High-Fat , Mice , Obesity/chemically induced , Obesity/genetics , Sequence Analysis, RNA
6.
Sci Rep ; 9(1): 2324, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30787418

ABSTRACT

Non-alcoholic steatohepatitis (NASH) signified by hepatic steatosis, inflammation, hepatocellular injury, and fibrosis is a growing cause of chronic liver disease, cirrhosis, and hepatocellular carcinoma. Hepatic fibrosis resulting from accumulation of extracellular matrix proteins secreted by hepatic myofibroblasts plays an important role in disease progression. Activated hepatic stellate cells (HSCs) have been identified as the primary source of myofibroblasts in animal models of hepatotoxic liver injury; however, so far HSC activation and plasticity have not been thoroughly investigated in the context of NASH-related fibrogenesis. Here we have determined the time-resolved changes in the HSC transcriptome during development of Western diet- and fructose-induced NASH in mice, a NASH model recapitulating human disease. Intriguingly, HSC transcriptional dynamics are highly similar across disease models pointing to HSC activation as a point of convergence in the development of fibrotic liver disease. Bioinformatic interrogation of the promoter sequences of activated genes combined with loss-of-function experiments indicates that the transcriptional regulators ETS1 and RUNX1 act as drivers of NASH-associated HSC plasticity. Taken together, our results implicate HSC activation and transcriptional plasticity as key aspects of NASH pathophysiology.


Subject(s)
Gene Expression Regulation , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Non-alcoholic Fatty Liver Disease/genetics , Transcription, Genetic , Animals , Cell Plasticity , Core Binding Factor Alpha 2 Subunit/metabolism , Diet, Western , Feeding Behavior , Fructose , Humans , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/pathology , Proto-Oncogene Protein c-ets-1/metabolism , Time Factors , Transcriptome/genetics
7.
Cell Mol Life Sci ; 75(11): 2075-2091, 2018 06.
Article in English | MEDLINE | ID: mdl-29264673

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by a progressive loss of dopamine (DA) neurons through apoptotic, inflammatory and oxidative stress mechanisms. The octadecaneuropeptide (ODN) is a diazepam-binding inhibitor (DBI)-derived peptide, expressed by astrocytes, which protects neurons against oxidative cell damages and apoptosis in an in vitro model of PD. The present study reveals that a single intracerebroventricular injection of 10 ng ODN 1 h after the last administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) prevented the degeneration of DA neurons induced by the toxin in the substantia nigra pars compacta of mice, 7 days after treatment. ODN-mediated neuroprotection was associated with a reduction of the number of glial fibrillary acidic protein-positive reactive astrocytes and a strong inhibition of the expression of pro-inflammatory genes such as interleukins 1ß and 6, and tumor necrosis factor-α. Moreover, ODN blocked the inhibition of the anti-apoptotic gene Bcl-2, and the stimulation of the pro-apoptotic genes Bax and caspase-3, induced by MPTP in the substantia nigra pars compacta. ODN also decreased or even in some cases abolished MPTP-induced oxidative damages, overproduction of reactive oxygen species and accumulation of lipid oxidation products in DA neurons. Furthermore, DBI knockout mice appeared to be more vulnerable than wild-type animals to MPTP neurotoxicity. Taken together, these results show that the gliopeptide ODN exerts a potent neuroprotective effect against MPTP-induced degeneration of nigrostriatal DA neurons in mice, through mechanisms involving downregulation of neuroinflammatory, oxidative and apoptotic processes. ODN may, thus, reduce neuronal damages in PD and other cerebral injuries involving oxidative neurodegeneration.


Subject(s)
Diazepam Binding Inhibitor/therapeutic use , Neuropeptides/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Peptide Fragments/therapeutic use , Animals , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Gene Expression Regulation/drug effects , Humans , Male , Mice, Inbred C57BL , Oxidative Stress/drug effects , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Reactive Oxygen Species/metabolism
8.
Behav Brain Res ; 313: 201-207, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27363924

ABSTRACT

Diazepam is well known for its anxiolytic properties, which are mediated via activation of the GABAA receptor. Diazepam Binding Inhibitor (DBI), also called acyl-CoA binding protein (ACBP), is a ubiquitously expressed protein originally identified based on its ability to displace diazepam from its binding site on the GABAA receptor. Central administration of ACBP or its cleaved fragment, commonly referred to as endozepines, induces proconflict and anxiety-like behaviour in rodents. For this reason, ACBP is known as an anxiogenic peptide. However, the role of endogenous ACBP in anxiety-like behaviour and anxiolytic responses to diazepam has not been investigated. To address this question, we assessed anxiety behaviour and anxiolytic responses to diazepam in two complementary loss-of-function mouse models including astrocyte-specific ACBP KO (ACBP(GFAP) KO) and whole-body KO (ACBP KO) mice. Male and female ACBP(GFAP) KO and ACBP KO mice do not show significant changes in anxiety-like behaviour compared to control littermates during elevated plus maze (EPM) and open field (OF) tests. Surprisingly, ACBP(GFAP) KO and ACBP KO mice were unresponsive to the anxiolytic effect of a low dose of diazepam during EPM tests. In conclusion, our experiments using genetic ACBP loss-of-function models suggest that endozepines deficiency does not affect anxiety-like behaviour in mice and impairs the anxiolytic action of diazepam.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/physiopathology , Behavior, Animal/drug effects , Diazepam Binding Inhibitor/metabolism , Diazepam/pharmacology , Animals , Anxiety/drug therapy , Carrier Proteins/genetics , Mice, Knockout , RNA, Messenger/metabolism , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism
9.
Cell Rep ; 13(9): 2000-13, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26628366

ABSTRACT

Cold exposure greatly alters brown adipose tissue (BAT) gene expression and metabolism to increase thermogenic capacity. Here, we used RNA sequencing and mass-spectrometry-based lipidomics to provide a comprehensive resource describing the molecular signature of cold adaptation at the level of the transcriptome and lipidome. We show that short-term (3-day) cold exposure leads to a robust increase in expression of several brown adipocyte genes related to thermogenesis as well as the gene encoding the hormone irisin. However, pathway analysis shows that the most significantly induced genes are those involved in glycerophospholipid synthesis and fatty acid elongation. This is accompanied by significant changes in the acyl chain composition of triacylglycerols (TAGs) as well as subspecies-selective changes of acyl chains in glycerophospholipids. These results indicate that cold adaptation of BAT is associated with significant and highly species-selective remodeling of both TAGs and glycerophospholipids.


Subject(s)
Adipose Tissue, Brown/metabolism , Glycerophospholipids/metabolism , Animals , Cluster Analysis , Cold Temperature , Fibronectins/metabolism , Glutathione/metabolism , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , RNA/metabolism , Sequence Analysis, RNA , Thermogenesis , Time Factors , Transcriptome
10.
J Lipid Res ; 56(9): 1738-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26142722

ABSTRACT

Acyl-CoA binding protein (ACBP) is a small, ubiquitously expressed intracellular protein that binds C14-C22 acyl-CoA esters with very high affinity and specificity. We have recently shown that targeted disruption of the Acbp gene leads to a compromised epidermal barrier and that this causes delayed adaptation to weaning, including the induction of the hepatic lipogenic and cholesterogenic gene programs. Here we show that ACBP is highly expressed in the Harderian gland, a gland that is located behind the eyeball of rodents and involved in the production of fur lipids and lipids used for lubrication of the eye lid. We show that disruption of the Acbp gene leads to a significant enlargement of this gland with hypertrophy of the acinar cells and increased de novo synthesis of monoalkyl diacylglycerol, the main lipid species produced by the gland. Mice with conditional targeting of the Acbp gene in the epidermis recapitulate this phenotype, whereas generation of an artificial epidermal barrier during gland development reverses the phenotype. Our findings indicate that the Harderian gland is activated by the compromised epidermal barrier as an adaptive and protective mechanism to overcome the barrier defect.


Subject(s)
Acinar Cells/metabolism , Cholesterol/metabolism , Diazepam Binding Inhibitor/genetics , Harderian Gland/metabolism , Animals , Cholesterol/genetics , Diazepam Binding Inhibitor/metabolism , Epidermis/metabolism , Epidermis/pathology , Lipids/biosynthesis , Lipogenesis/genetics , Liver/metabolism , Mice , Monoglycerides/biosynthesis
11.
Cell Rep ; 5(5): 1403-12, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24316079

ABSTRACT

We previously reported that mice deficient in acyl-CoA-binding protein (ACBP) display a delayed metabolic adaptation to weaning. This includes a delayed activation of the hepatic lipogenic gene program, which may result from hepatic accumulation of triacylglycerol and/or cholesteryl esters in the late suckling period. To further investigate the basis for this phenotype, we generated mice deficient in ACBP in hepatocytes (Alb-ACBP(-/-)) and keratinocytes (K14-ACBP(-/-)). Surprisingly, the delayed adaptation to weaning, including hepatic lipid accumulation, is caused by ACBP deficiency in the skin rather than in the liver. Similarly to ACBP(-/-) mice, K14-ACBP(-/-) mice exhibit an increased transepidermal water loss, and we show that the hepatic phenotype is caused specifically by the epidermal barrier defect, which leads to increased lipolysis in white adipose tissue. Our data demonstrate that an imperfect epidermal barrier leads to profound suppression of the hepatic SREBP gene program and lipid accumulation in the liver.


Subject(s)
Adaptation, Physiological , Diazepam Binding Inhibitor/metabolism , Epidermis/metabolism , Hepatocytes/metabolism , Keratinocytes/metabolism , Weaning , Adipose Tissue, White/metabolism , Animals , Body Water/metabolism , Diazepam Binding Inhibitor/genetics , Lipolysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism
12.
J Lipid Res ; 53(10): 2162-2174, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22829653

ABSTRACT

The acyl-CoA binding protein (ACBP) is a 10 kDa intracellular protein expressed in all eukaryotic species. Mice with targeted disruption of Acbp (ACBP(-/-) mice) are viable and fertile but present a visible skin and fur phenotype characterized by greasy fur and development of alopecia and scaling with age. Morphology and development of skin and appendages are normal in ACBP(-/-) mice; however, the stratum corneum display altered biophysical properties with reduced proton activity and decreased water content. Mass spectrometry analyses of lipids from epidermis and stratum corneum of ACBP(+/+) and ACBP(-/-) mice showed very similar composition, except for a significant and specific decrease in the very long chain free fatty acids (VLC-FFA) in stratum corneum of ACBP(-/-) mice. This finding indicates that ACBP is critically involved in the processes that lead to production of stratum corneum VLC-FFAs via complex phospholipids in the lamellar bodies. Importantly, we show that ACBP(-/-) mice display a ∼50% increased transepidermal water loss compared with ACBP(+/+) mice. Furthermore, skin and fur sebum monoalkyl diacylglycerol (MADAG) levels are significantly increased, suggesting that ACBP limits MADAG synthesis in sebaceous glands. In summary, our study shows that ACBP is required for production of VLC-FFA for stratum corneum and for maintaining normal epidermal barrier function.


Subject(s)
Diazepam Binding Inhibitor/genetics , Epidermis/metabolism , Animals , Cholesterol/metabolism , Diazepam Binding Inhibitor/metabolism , Lipid Metabolism , Lipids/analysis , Mass Spectrometry , Mice , Mice, Inbred Strains , Phenotype , Sebaceous Glands/chemistry , Sebaceous Glands/metabolism , Skin/chemistry , Skin/metabolism
13.
Am J Physiol Renal Physiol ; 302(8): F1034-44, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22237802

ABSTRACT

The acyl-CoA binding protein (ACBP) is a small intracellular protein that specifically binds and transports medium to long-chain acyl-CoA esters. Previous studies have shown that ACBP is ubiquitously expressed but found at particularly high levels in lipogenic cell types as well as in many epithelial cells. Here we show that ACBP is widely expressed in human and mouse kidney epithelium, with the highest expression in the proximal convoluted tubules. To elucidate the role of ACBP in the renal epithelium, mice with targeted disruption of the ACBP gene (ACBP(-/-)) were used to study water and NaCl balance as well as urine concentrating ability in metabolic cages. Food intake and urinary excretion of Na(+) and K(+) did not differ between ACBP(-/-) and (+/+) mice. Interestingly, however, water intake and diuresis were significantly higher at baseline in ACBP(-/-) mice compared with that of (+/+) mice. Subsequent to 20-h water deprivation, ACBP(-/-) mice exhibited increased diuresis, reduced urine osmolality, elevated hematocrit, and higher relative weight loss compared with (+/+) mice. There were no significant differences in plasma concentrations of renin, corticosterone, and aldosterone between mice of the two genotypes. After water deprivation, renal medullary interstitial fluid osmolality and concentrations of Na(+), K(+), and urea did not differ between genotypes and cAMP excretion was similar. Renal aquaporin-1 (AQP1), -2, and -4 protein abundances did not differ between water-deprived (+/+) and ACBP(-/-) mice; however, ACBP(-/-) mice displayed increased apical targeting of pS256-AQP2. AQP3 abundance was lower in ACBP(-/-) mice than in (+/+) control animals. Thus we conclude that ACBP is necessary for intact urine concentrating ability. Our data suggest that the deficiency in urine concentrating ability in the ACBP(-/-) may be caused by reduced AQP3, leading to impaired efflux over the basolateral membrane of the collecting duct.


Subject(s)
Aquaporin 3/biosynthesis , Diazepam Binding Inhibitor/physiology , Kidney Concentrating Ability/physiology , Kidney/physiology , Aldosterone/blood , Animals , Aquaporin 3/genetics , Corticosterone/blood , Diuresis/physiology , Drinking/physiology , Gene Deletion , Humans , Kidney/cytology , Kidney/metabolism , Mice , Mice, Inbred C57BL , Osmolar Concentration , Potassium/urine , Renin/blood , Sodium/urine , Urea/analysis , Water Deprivation/physiology
14.
J Biol Chem ; 286(5): 3460-72, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21106527

ABSTRACT

The acyl-CoA-binding protein (ACBP)/diazepam binding inhibitor is an intracellular protein that binds C(14)-C(22) acyl-CoA esters and is thought to act as an acyl-CoA transporter. In vitro analyses have indicated that ACBP can transport acyl-CoA esters between different enzymatic systems; however, little is known about the in vivo function in mammalian cells. We have generated mice with targeted disruption of ACBP (ACBP(-/-)). These mice are viable and fertile and develop normally. However, around weaning, the ACBP(-/-) mice go through a crisis with overall weakness and a slightly decreased growth rate. Using microarray analysis, we show that the liver of ACBP(-/-) mice displays a significantly delayed adaptation to weaning with late induction of target genes of the sterol regulatory element-binding protein (SREBP) family. As a result, hepatic de novo cholesterogenesis is decreased at weaning. The delayed induction of SREBP target genes around weaning is caused by a compromised processing and decreased expression of SREBP precursors, leading to reduced binding of SREBP to target sites in chromatin. In conclusion, lack of ACBP interferes with the normal metabolic adaptation to weaning and leads to delayed induction of the lipogenic gene program in the liver.


Subject(s)
Adaptation, Physiological , Diazepam Binding Inhibitor/metabolism , Liver/metabolism , Weaning , Animals , Animals, Newborn , Cholesterol/biosynthesis , Chromatin/metabolism , Gene Expression Profiling , Liver/physiology , Metabolism , Mice , Mice, Knockout , Sterol Regulatory Element Binding Protein 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...