Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Ther ; 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38532630

ABSTRACT

Base editing of hematopoietic stem/progenitor cells (HSPCs) is an attractive strategy for treating immunohematologic diseases. However, the feasibility of using adenine-base-edited HSPCs for treating X-linked severe combined immunodeficiency (SCID-X1), the influence of dose-response relationships on immune cell generation, and the potential risks have not been demonstrated in vivo. Here, a humanized SCID-X1 mouse model was established, and 86.67% ± 2.52% (n = 3) of mouse hematopoietic stem cell (HSC) pathogenic mutations were corrected, with no single-guide-RNA (sgRNA)-dependent off-target effects detected. Analysis of peripheral blood over 16 weeks post-transplantation in mice with different immunodeficiency backgrounds revealed efficient immune cell generation following transplantation of different amounts of modified HSCs. Therefore, a large-scale infusion of gene-corrected HSCs within a safe range can achieve rapid, stable, and durable immune cell regeneration. Tissue-section staining further demonstrated the restoration of immune organ tissue structures, with no tumor formation in multiple organs. Collectively, these data suggest that base-edited HSCs are a potential therapeutic approach for SCID-X1 and that a threshold infusion dose of gene-corrected cells is required for immune cell regeneration. This study lays a theoretical foundation for the clinical application of base-edited HSCs in treating SCID-X1.

2.
Nanotechnology ; 35(3)2023 Oct 30.
Article in English | MEDLINE | ID: mdl-37797600

ABSTRACT

The electrolytic water method is an outstanding hydrogen production process because of its high stability and no restriction. A low-priced and efficient catalyst for electro-deposition of Ni-Co microspheres and nanoclusters on carbon steel (Ni-Co/CS) has been prepared by the dynamic hydrogen bubble template. In the 6 M KOH solution, Ni-Co/CS only requires an overpotential of 48 mV to provide a current density of 50 mA cm-2. At the same time, it also has a large electrochemically active specific surface area (ECSA) and a hydrophilic surface. In addition, the study about the influence of carbon steel (CS) on Ni-Co coatings and the comparison experiment for different base materials has been completed. The results prove that CS is an excellent base material for hydrogen production. It can help the Ni-Co catalyst to have a stable electrolysis in 6 M KOH for 500 h. The above properties of Ni-Co/CS catalyst make it a new choice of hydrogen production by electrolysis of water in practical applications.

3.
J Biol Chem ; 299(2): 102881, 2023 02.
Article in English | MEDLINE | ID: mdl-36626986

ABSTRACT

Mutations in genes involved in mitochondrial proline catabolism lead to the rare genetic disorder hyperprolinemia in humans. We have previously reported that mutations of proline catabolic genes in Caenorhabditis elegans impair mitochondrial homeostasis and shorten life span, and that these effects surprisingly occur in a diet type-dependent manner. Therefore, we speculated that a specific dietary component may mitigate the adverse effects of defective proline catabolism. Here, we discovered that high dietary glucose, which is generally detrimental to health, actually improves mitochondrial homeostasis and life span in C. elegans with faulty proline catabolism. Mechanistically, defective proline catabolism results in a shift of glucose catabolism toward the pentose phosphate pathway, which is crucial for cellular redox balance. This shift helps to maintain mitochondrial reactive oxygen species homeostasis and to extend life span, as suppression of the pentose phosphate pathway enzyme GSPD-1 prevents the favorable effects of high glucose. In addition, we demonstrate that this crosstalk between proline and glucose catabolism is mediated by the transcription factor DAF-16. Altogether, these findings suggest that a glucose-rich diet may be advantageous in certain situations and might represent a potentially viable treatment strategy for disorders involving impaired proline catabolism.


Subject(s)
Caenorhabditis elegans , Glucose , Longevity , Animals , Humans , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Glucose/metabolism , Glucose/pharmacology , Longevity/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Proline/metabolism
4.
J Biol Chem ; 298(7): 102118, 2022 07.
Article in English | MEDLINE | ID: mdl-35691340

ABSTRACT

Sphingolipids are a class of bioactive complex lipids that have been closely associated with aging and aging-related diseases. However, the mechanism through which sphingolipids control aging has long been a mystery. Emerging studies reveal that sphingolipids exert tight control over lysosomal homeostasis and function, as evidenced by sphingolipid-related diseases, including but not limited to lysosomal storage disorders. These diseases are defined by primary lysosomal defects and a few secondary defects such as mitochondrial dysfunction. Intriguingly, recent research indicates that the majority of these defects are also associated with aging, implying that sphingolipid-related diseases and aging may share common mechanisms. We propose that the lysosome is a pivotal hub for sphingolipid-mediated aging regulation. This review discusses the critical roles of sphingolipid metabolism in regulating various lysosomal functions, with an emphasis on how such regulation may contribute to aging and aging-related diseases.


Subject(s)
Aging/pathology , Lysosomes/metabolism , Sphingolipids , Aging/metabolism , Humans , Sphingolipids/metabolism
5.
Front Physiol ; 12: 775648, 2021.
Article in English | MEDLINE | ID: mdl-34887779

ABSTRACT

Phospholipids are major membrane lipids that consist of lipid bilayers. This basic cellular structure acts as a barrier to protect the cell against various environmental insults and more importantly, enables multiple cellular processes to occur in subcellular compartments. Numerous studies have linked the complexity of membrane lipids to signal transductions, organelle functions, as well as physiological processes, and human diseases. Recently, crucial roles for membrane lipids in the aging process are beginning to emerge. In this study, we summarized current advances in our understanding of the relationship between membrane lipids and aging with an emphasis on phospholipid species. We surveyed how major phospholipid species change with age in different organisms and tissues, and some common patterns of membrane lipid change during aging were proposed. Further, the functions of different phospholipid molecules in regulating healthspan and lifespan, as well as their potential mechanisms of action, were also discussed.

6.
Mech Ageing Dev ; 200: 111586, 2021 12.
Article in English | MEDLINE | ID: mdl-34655615

ABSTRACT

Metabolic reprogramming is crucial for the adaptation to environmental temperature stress. It is generally accepted that fatty acid (FA) desaturation is suppressed at high temperature, which decreases the ratio of unsaturated FAs to saturated FAs (UFAs/SFAs) to maintain the fluidity of cell membranes and favor cellular survival. Here by working in C. elegans, we found that FA desaturation is essential for longevity in response to temperature upshift at the organismal level, opposite to its role in cellular survival. High temperature unexpectedly increases the contents of total fat and multiple UFA species. Specifically, monounsaturated oleic acid (OA) is required for animal survival at high temperature. Mechanistic study showed that OA acts through HSF-1, which in turn promotes histone acetylation as well as the expression of defense genes that are crucial for longevity at high temperature. Together, our findings reveal an unprecedented role for FA desaturation in organismal fitness to temperature upshift, and implicate divergent metabolic requirements between cellular and organismal survival upon temperature stress.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cell Survival/physiology , Fatty Acids/metabolism , Hot Temperature , Longevity/physiology , Oleic Acid/metabolism , Transcription Factors/metabolism , Adaptation, Physiological , Animals , Caenorhabditis elegans , Fatty Acid Desaturases/metabolism
7.
Nat Commun ; 12(1): 5073, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34417467

ABSTRACT

The contents of numerous membrane lipids change upon ageing. However, it is unknown whether and how any of these changes are causally linked to lifespan regulation. Acyl chains contribute to the functional specificity of membrane lipids. In this study, working with C. elegans, we identified an acyl chain-specific sphingolipid, C22 glucosylceramide, as a longevity metabolite. Germline deficiency, a conserved lifespan-extending paradigm, induces somatic expression of the fatty acid elongase ELO-3, and behenic acid (22:0) generated by ELO-3 is incorporated into glucosylceramide for lifespan regulation. Mechanistically, C22 glucosylceramide is required for the membrane localization of clathrin, a protein that regulates membrane budding. The reduction in C22 glucosylceramide impairs the clathrin-dependent autophagic lysosome reformation, which subsequently leads to TOR activation and longevity suppression. These findings reveal a mechanistic link between membrane lipids and ageing and suggest a model of lifespan regulation by fatty acid-mediated membrane configuration.


Subject(s)
Caenorhabditis elegans/physiology , Fatty Acids, Nonesterified/metabolism , Glycosphingolipids/metabolism , Homeostasis , Longevity/physiology , Lysosomes/metabolism , Animals , Caenorhabditis elegans Proteins/metabolism , Ceramides/metabolism , Cholesterol/metabolism , Clathrin/metabolism , Germ-Line Mutation/genetics , Green Fluorescent Proteins/metabolism , Larva/metabolism , Models, Biological , RNA Interference , Stress, Physiological
8.
PLoS Genet ; 17(5): e1009573, 2021 05.
Article in English | MEDLINE | ID: mdl-34014977

ABSTRACT

Coordinated regulation of stress response pathways is crucial for cellular homeostasis. However, crosstalk between the different stress pathways and the physiological significance of this crosstalk remain poorly understood. In this study, using the model organism C. elegans, we discovered that suppression of the transcription factor LET-607/CREBH, a regulator of cellular defense and proteostatic responses, triggers adaptive induction of DAF-16-dependent stress responses. Suppression of LET-607 improves stress resistance and extends C. elegans lifespan in a DAF-16-dependent manner. We identified the sphingomyelin synthase SMS-5 to be a central mediator in the communication between LET-607 and DAF-16. SMS-5 reduces the contents of unsaturated phosphatidylcholine (PC), which activates DAF-16 through ITR-1-dependent calcium signaling and calcium-sensitive kinase PKC-2. Our data reveal the significance of crosstalk between different stress pathways in animal fitness and identify LET-607/CREBH and specific PC as regulators of DAF-16 and longevity.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Forkhead Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Phosphatidylcholines/metabolism , Stress, Physiological , Transferases (Other Substituted Phosphate Groups)/metabolism , Adaptation, Physiological , Animals , Calcium Signaling , Germ-Line Mutation , Longevity/genetics , Membrane Lipids/metabolism , Protein Kinase C/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics
9.
J Biol Chem ; 296: 100095, 2021.
Article in English | MEDLINE | ID: mdl-33208465

ABSTRACT

DNA damage triggers the cellular adaptive response to arrest proliferation and repair DNA damage; when damage is too severe to be repaired, apoptosis is initiated to prevent the spread of genomic insults. However, how cells endure DNA damage to maintain cell function remains largely unexplored. By using Caenorhabditis elegans as a model, we report that DNA damage elicits cell maintenance programs, including the unfolded protein response of the endoplasmic reticulum (UPRER). Mechanistically, sublethal DNA damage unexpectedly suppresses apoptotic genes in C. elegans, which in turn increases the activity of the inositol-requiring enzyme 1/X-box binding protein 1 (IRE-1/XBP-1) branch of the UPRER by elevating unsaturated phosphatidylcholine. In addition, UPRER activation requires silencing of the lipid regulator skinhead-1 (SKN-1). DNA damage suppresses SKN-1 activity to increase unsaturated phosphatidylcholine and activate UPRER. These findings reveal the UPRER activation as an organismal adaptive response that is important to maintain cell function during DNA damage.


Subject(s)
Caenorhabditis elegans/metabolism , DNA Damage , Endoplasmic Reticulum Stress , Phosphatidylcholines/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Phosphatidylcholines/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Unfolded Protein Response/genetics
10.
G3 (Bethesda) ; 10(5): 1707-1712, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32161088

ABSTRACT

The transcription factor SKN-1, the C. elegans ortholog of mammalian Nrf protein, is a well-known longevity factor, and its activation is observed in several long-lived models. SKN-1 also plays essential roles in xenobiotic and oxidative stress responses. Here, we report deleterious functions of SKN-1 in somatic stress resistance that may impair lifespan. Constitutive SKN-1 activation impairs animal resistance to several stresses, including heat, ER stress and mitochondrial stress, which result from the suppression of DAF-16, another master regulator of longevity. SKN-1 activation abrogates DAF-16 nuclear import and downregulates DAF-16 target genes under stress conditions, while SKN-1 inhibition promotes the expression of DAF-16 targets, even in long-lived mutants. Further, SKN-1 activation induces the expression of vitellogenin proteins, which are required for SKN-1-mediated suppression of DAF-16 and stress resistance. Together, these findings identify detrimental roles for SKN-1 activation in animal health, and more importantly, inspire the rethinking of the complex roles for SKN-1 in aging regulation.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Longevity/genetics , Oxidative Stress , RNA Interference , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Elife ; 92020 02 05.
Article in English | MEDLINE | ID: mdl-32022684

ABSTRACT

Exposure to environmental stress is clinically established to influence male reproductive health, but the impact of normal cellular metabolism on sperm quality is less well-defined. Here we show that impaired mitochondrial proline catabolism, reduces energy-storing flavin adenine dinucleotide (FAD) levels, alters mitochondrial dynamics toward fusion, and leads to age-related loss of sperm quality (size and activity), which diminishes competitive fitness of the animal. Loss of the 1-pyrroline-5-carboxylate dehydrogenase enzyme alh-6 that catalyzes the second step in mitochondrial proline catabolism leads to premature male reproductive senescence. Reducing the expression of the proline catabolism enzyme alh-6 or FAD biosynthesis pathway genes in the germline is sufficient to recapitulate the sperm-related phenotypes observed in alh-6 loss-of-function mutants. These sperm-specific defects are suppressed by feeding diets that restore FAD levels. Our results define a cell autonomous role for mitochondrial proline catabolism and FAD homeostasis on sperm function and specify strategies to pharmacologically reverse these defects.


Subject(s)
Caenorhabditis elegans/physiology , Flavin-Adenine Dinucleotide/metabolism , Spermatozoa/physiology , 1-Pyrroline-5-Carboxylate Dehydrogenase/genetics , Animals , Caenorhabditis elegans/metabolism , Male , Mitochondria/enzymology , Mitochondrial Dynamics , Reproduction , Spermatozoa/metabolism
12.
PLoS Genet ; 15(4): e1008122, 2019 04.
Article in English | MEDLINE | ID: mdl-31034475

ABSTRACT

Early exposure to some mild stresses can slow down the aging process and extend lifespan, raising the question of how early life stress might impact the somatic health of aged animals. Here, we reveal that early life heat experience triggers the establishment of epigenetic memory in soma, which promotes long-lasting stress responses and longevity in C. elegans. Unlike lethal heat shock, mild heat activates a unique transcriptional program mimicking pathogen defense responses, characterized by the enhanced expression of innate immune and detoxification genes. Surprisingly, the expression of defense response genes persists long after heat exposure, conferring enhanced stress resistance even in aged animals. Further studies identify the histone acetyltransferase CBP-1 and the chromatin remodeling SWI/SNF complex as epigenetic modulators of the long-lasting defense responses. Histone acetylation is elevated by heat stress and maintained into agedness thereafter. Accordingly, histone acetylation levels were increased on the promoters of defense genes. Moreover, disruption of epigenetic memory abrogates the longevity response to early hormetic heat stress, indicating that long-lasting defense responses are crucial for the survival of aged animals. Together, our findings provide mechanistic insights into how temperature stress experienced in early life provides animals with lifetime health benefits.


Subject(s)
Heat-Shock Response , Histones/metabolism , Longevity , Acetylation , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Epigenesis, Genetic , Hot Temperature , Immunity, Innate , Metabolic Detoxication, Phase I , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Promoter Regions, Genetic
13.
Cell Rep ; 17(11): 2837-2844, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27974198

ABSTRACT

Metabolic pathways are regulated to fuel or instruct the immune responses to pathogen threats. However, the regulatory roles for amino acid metabolism in innate immune responses remains poorly understood. Here, we report that mitochondrial proline catabolism modulates innate immunity in Caenorhabditis elegans. Modulation of proline catabolic enzymes affects host susceptibility to bacterial pathogen Pseudomonas aeruginosa. Mechanistically, proline catabolism governs reactive oxygen species (ROS) homeostasis and subsequent activation of SKN-1, a critical transcription factor regulating xenobiotic stress response and pathogen defense. Intriguingly, proline catabolism-mediated activation of SKN-1 requires cell-membrane dual-oxidase Ce-Duox1/BLI-3, highlighting the importance of interaction between mitochondrial and cell-membrane components in host defense. Our findings reveal how animals utilize metabolism of a single amino acid to defend against a pathogen and identify proline catabolism as a component of innate immune signaling.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/metabolism , DNA-Binding Proteins/genetics , Dual Oxidases/genetics , Oxidoreductases/genetics , Transcription Factors/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Host-Pathogen Interactions/genetics , Immunity, Innate/genetics , Metabolic Networks and Pathways , Metabolism/genetics , Mitochondria/metabolism , Proline/metabolism , Pseudomonas aeruginosa/pathogenicity , Reactive Oxygen Species/metabolism
14.
Nat Commun ; 5: 5048, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-25284427

ABSTRACT

Mechanisms that coordinate different metabolic pathways, such as glucose and lipid, have been recognized. However, a potential interaction between amino acid and lipid metabolism remains largely elusive. Here we show that during starvation of Caenorhabditis elegans, proline catabolism is coupled with lipid metabolism by SKN-1. Mutation of alh-6, a conserved proline catabolic enzyme, accelerates fat mobilization, enhances the expression of genes involved in fatty acid oxidation and reduces survival in response to fasting. This metabolic coordination is mediated by the activation of the transcription factor SKN-1/Nrf2, possibly due to the accumulation of the alh-6 substrate P5C, and also requires the transcriptional co-regulator MDT-15. Constitutive activation of SKN-1 induces a similar transcriptional response, which protects animals from fat accumulation when fed a high carbohydrate diet. In human cells, an orthologous alh-6 enzyme, ALDH4A1, is also linked to the activity of Nrf2, the human orthologue of SKN-1, and regulates the expression of lipid metabolic genes. Our findings identify a link between proline catabolism and lipid metabolism, and uncover a physiological role for SKN-1 in metabolism.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/metabolism , Lipid Metabolism , NF-E2-Related Factor 2/metabolism , Transcription Factors/metabolism , 1-Pyrroline-5-Carboxylate Dehydrogenase/metabolism , Animals , Caenorhabditis elegans , Fatty Acids/chemistry , Food , HEK293 Cells , Humans , Metabolism , Oxidative Stress , Oxygen/chemistry , Proline/genetics , Proline/metabolism , RNA Interference
15.
Cell Metab ; 19(2): 221-31, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24440036

ABSTRACT

Diet has a substantial impact on cellular metabolism and physiology. Animals must sense different food sources and utilize distinct strategies to adapt to diverse diets. Here we show that Caenorhabditis elegans lifespan is regulated by their adaptive capacity to different diets, which is controlled by alh-6, a conserved proline metabolism gene. alh-6 mutants age prematurely when fed an Escherichia coli OP50 but not HT115 diet. Remarkably, this diet-dependent aging phenotype is determined by exposure to food during development. Mechanistically, the alh-6 mutation triggers diet-induced mitochondrial defects and increased generation of ROS, likely due to accumulation of its substrate 1-pyrroline-5-carboxylate. We also identify that neuromedin U receptor signaling is essential for diet-induced mitochondrial changes and premature aging. Moreover, dietary restriction requires alh-6 to induce longevity. Collectively, our data reveal a homeostatic mechanism that animals employ to cope with potential dietary insults and uncover an example of lifespan regulation by dietary adaptation.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Mitochondria/metabolism , Mutation , Proline/metabolism
16.
Bioessays ; 34(8): 652-4, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22674543

ABSTRACT

A recent study reported that longevity in Caenorhabditits elegans can be inherited over several generations. This is probably achieved through the following epigenetic mechanism: inherited demethylated histones at some central loci, such as miRNA, transcription factors or signaling regulators affect the expression of certain genes leading to the longevity phenotype.


Subject(s)
Epigenesis, Genetic , Genome, Human , Inheritance Patterns , Longevity , Aging , Animals , DNA Methylation , Gene Expression Profiling , Gene Expression Regulation , Genetic Loci , Genotype , Histones/metabolism , Humans , Phenotype , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Int J Biochem Cell Biol ; 44(4): 629-38, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22245600

ABSTRACT

Renal hypertrophy and extracellular matrix accumulation are early features of diabetic nephropathy. Hyperglycemia-induced oxidative stress is implicated in the etiology of diabetic nephropathy. Resveratrol has potent antioxidative and protective effects on diabetic nephropathy. We aimed to examine whether high glucose (HG)-induced NADPH oxidase activation and reactive oxygen species (ROS) production contribute to glomerular mesangial cell proliferation and fibronectin expression and the effect of resveratrol on HG action in mesangial cells. By using rat mesangial cell line and primary mesangial cells, we found that NADPH oxidase inhibitor (apocynin) and ROS inhibitor (N-acetyl cysteine) both inhibited HG-induced mesangial cell proliferation and fibronectin expression. HG-induced elevation of NADPH oxidase activity and production of ROS in mesangial cells was inhibited by apocynin. These results suggest that HG induces mesangial cell proliferation and fibronectin expression through NADPH oxidase-mediated ROS production. Mechanistic studies revealed that HG upregulated NADPH oxidase subunits p22(phox) and p47(phox) expression through JNK/NF-κB pathway, which resulted in elevation of NADPH oxidase activity and consequent ROS production. Resveratrol prevented HG-induced mesangial cell proliferation and fibronectin expression through inhibiting HG-induced JNK and NF-κB activation, NADPH oxidase activity elevation and ROS production. These results demonstrate that HG enhances mesangial cell proliferation and fibronectin expression through JNK/NF-κB/NADPH oxidase/ROS pathway, which was inhibited by resveratrol. Our findings provide novel therapeutic targets for diabetic nephropathy.


Subject(s)
Fibronectins/metabolism , Gene Expression Regulation/drug effects , Glucose/antagonists & inhibitors , Glucose/pharmacology , Mesangial Cells/drug effects , Signal Transduction/drug effects , Stilbenes/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , JNK Mitogen-Activated Protein Kinases/metabolism , Mesangial Cells/cytology , Mesangial Cells/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Subunits/metabolism , Rats , Reactive Oxygen Species/metabolism , Resveratrol
18.
Diabetes ; 59(12): 3041-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20855545

ABSTRACT

OBJECTIVE: Toll-like receptor 4 (TLR4) has been reported to induce insulin resistance through inflammation in high-fat-fed mice. However, the physiological role of TLR4 in metabolism is unknown. Here, we investigated the involvement of TLR4 in fasting metabolism. RESEARCH DESIGN AND METHODS: Wild-type and TLR4 deficient (TLR4(-/-)) mice were either fed or fasted for 24 h. Glucose and lipid levels in circulation and tissues were measured. Glucose and lipid metabolism in tissues, as well as the expression of related enzymes, was examined. RESULTS: Mice lacking TLR4 displayed aggravated fasting hypoglycemia, along with normal hepatic gluconeogenesis, but reversed activity of pyruvate dehydrogenase complex (PDC) in skeletal muscle, which might account for the fasting hypoglycemia. TLR4(-/-) mice also exhibited higher lipid levels in circulation and skeletal muscle after fasting and reversed expression of lipogenic enzymes in skeletal muscle but not liver and adipose tissue. Adipose tissue lipolysis is normal and muscle fatty acid oxidation is increased in TLR4(-/-) mice after fasting. Inhibition of fatty acid synthesis in TLR4(-/-) mice abolished hyperlipidemia, hypoglycemia, and PDC activity increase, suggesting that TLR4-dependent inhibition of muscle lipogenesis may contribute to glucose and lipid homeostasis during fasting. Further studies showed that TLR4 deficiency had no effect on insulin signaling and muscle proinflammatory cytokine production in response to fasting. CONCLUSIONS: These data suggest that TLR4 plays a critical role in glucose and lipid metabolism independent of insulin during fasting and identify a novel physiological role for TLR4 in fuel homeostasis.


Subject(s)
Fasting/physiology , Toll-Like Receptor 4/physiology , Adipose Tissue/physiology , Animals , Carbon Dioxide/analysis , DNA, Complementary/genetics , Fatty Acids/metabolism , Homeostasis , Hypoglycemia/genetics , Lipolysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Oxygen Consumption , Polymerase Chain Reaction , RNA/drug effects , RNA/isolation & purification , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
19.
J Immunol ; 185(4): 2563-9, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20631308

ABSTRACT

IL-17 is a recently identified proinflammatory cytokine that plays pivotal roles in several chronic inflammatory disease models. Its expression was also found to be elevated in the serum of patients with chronic diseases. However, whether elevated systemic IL-17 expression can induce pathophysiological tissue inflammation is unknown. In this study, we demonstrated that systemic overexpression of IL-17 using an adenoviral vector could induce multiple tissue inflammation and wasting in mice. We also found that the expression of TLR4 was increased in tissues of IL-17-overexpressing mice. Moreover, TLR4 activation is required for IL-17-induced tissue inflammation and wasting, as evidenced by the absence of aggressive atrophy in gastrocnemius muscle, neutrophil accumulation, and expression of proinflammatory cytokines downstream of TLR4 in multiple tissues of TLR4-deficient mice. Further investigation revealed that TLR4 endogenous ligands high-mobility group box 1 and heat shock protein 22, were systemically upregulated and might be involved in the IL-17-induced TLR4 activation. Our results suggest that IL-17 may induce disease-associated tissue inflammation and wasting through TLR4 signaling. The study indicates a novel interaction between IL-17 and TLR4 activation and may have implications in the pathogenesis and treatment of chronic diseases.


Subject(s)
Inflammation/metabolism , Interleukin-17/metabolism , Toll-Like Receptor 4/metabolism , Wasting Syndrome/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , Body Weight/genetics , Body Weight/physiology , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Inflammation/blood , Inflammation/genetics , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Interleukin-17/blood , Interleukin-17/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Toll-Like Receptor 4/genetics , Transduction, Genetic , Wasting Syndrome/blood , Wasting Syndrome/genetics
20.
Endocrinology ; 149(2): 672-80, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17962352

ABSTRACT

Pancreatic-derived factor (PANDER) is a cytokine-like peptide highly expressed in pancreatic beta-cells. PANDER was reported to promote apoptosis of pancreatic beta-cells and secrete in response to glucose. Here we explored the effects of glucose on PANDER expression, and the underlying mechanisms in murine pancreatic beta-cell line MIN6 and primary islets. Our results showed that glucose up-regulated PANDER mRNA and protein levels in a time- and dose-dependent manner in MIN6 cells and pancreatic islets. In cells expressing cAMP response element-binding protein (CREB) dominant-negative construct, glucose failed to induce PANDER gene expression and promoter activation. Treatment of the cells with calcium chelator [EGTA, 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetra(acetoxymethyl)ester (BAPTA/AM)], the voltage-dependent Ca(2+) channel inhibitor (nifedipine), the protein kinase A (PKA) inhibitor (H89), the protein kinase C (PKC) inhibitor (Go6976), or the MAPK kinase 1/2 inhibitor (PD98059), all significantly inhibited glucose-induced PANDER gene expression and promoter activation. Further studies showed that glucose induced CREB phosphorylation through Ca(2+)-PKA-ERK1/2 and Ca(2+)-PKC pathways. Thus, the Ca(2+)-PKA-ERK1/2-CREB and Ca(2+)-PKC-CREB signaling pathways are involved in glucose-induced PANDER gene expression. Wortmannin (phosphatidylinositol 3-kinase inhibitor), ammonium pyrrolidinedithiocarbamate (nuclear factor-kappaB inhibitor and nonspecific antioxidant), and N-acetylcysteine (antioxidant) were also found to inhibit glucose-induced PANDER promoter activation and gene expression. Because there is no nuclear factor-kappaB binding site in the promoter region of PANDER gene, these results suggest that phosphatidylinositol 3-kinase and reactive oxygen species be involved in glucose-induced PANDER gene expression. In conclusion, glucose induces PANDER gene expression in pancreatic beta-cells through multiple signaling pathways. Because PANDER is expressed by pancreatic beta-cells and in response to glucose in a similar way to those of insulin, PANDER may be involved in glucose homeostasis.


Subject(s)
Cytokines/genetics , Cytokines/metabolism , Glucose/metabolism , Insulin-Secreting Cells/physiology , Signal Transduction/physiology , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression/drug effects , Gene Expression/physiology , Glucose/pharmacology , Insulin-Secreting Cells/cytology , Luciferases/genetics , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic/physiology , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...