Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; 10(5): e0170022, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36102652

ABSTRACT

The greenhouse gas methane (CH4) is of pivotal importance for Earth's climate system and as a human energy source. A significant fraction of this CH4 is produced by anaerobic Archaea. Here, we describe the first CH4 production by facultative anaerobic wood-rot fungi during growth on hydroxylated/carboxylated aromatic compounds, including lignin and lignite. The amount of CH4 produced by fungi is positively correlated with the amount of CH3Cl produced during the rapid growth period of the fungus. Biochemical, genetic, and stable isotopic tracer analyses reveal the existence of a novel halomethane-dependent fungal CH4 production pathway during the degradation of phenol and benzoic acid monomers and polymers and utilization of cyclic sugars. Even though this halomethane-dependent pathway may only play a side role in anaerobic fungal activity, it could represent a globally significant, previously overlooked source of biogenic CH4 in natural ecosystems. IMPORTANCE Here, we demonstrate that wood-rot fungi produce methane anaerobically without the involvement of methanogenic archaea via a new, halomethane-dependent pathway. These findings of an anaerobic fungal methane formation pathway open another avenue in methane research and will further assist with current efforts in the identification of the processes involved and their ecological implications.


Subject(s)
Greenhouse Gases , Wood , Humans , Wood/chemistry , Wood/metabolism , Wood/microbiology , Lignin/metabolism , Anaerobiosis , Ecosystem , Greenhouse Gases/analysis , Greenhouse Gases/metabolism , Methane/analysis , Methane/metabolism , Archaea/metabolism , Fungi/genetics , Fungi/metabolism , Coal/analysis , Sugars/metabolism , Phenols/analysis , Phenols/metabolism , Benzoic Acid/analysis , Benzoic Acid/metabolism
4.
Ecotoxicology ; 30(2): 231-239, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33483874

ABSTRACT

Chromium (Cr) pollution in farmlands is a common environmental issue, that can seriously inhibit plant growth, damage plant cells, and even cause plant death. In this study, bok choy (Brassica campestris L. ssp. chinensis Makino (var. communis Tsen et Lee)) was selected as a model plant to investigate the metabolic response to Cr stress at concentrations of 2.0 mg/L and 8.0 mg/L. Metabolites were identified using gas chromatography-mass spectrometry. Principal component analysis and orthogonal projections to latent structure discriminant analysis revealed the notable effect of Cr stress on the metabolites of bok choy. Under Cr stress, 145 metabolites were identified in the bok choy leaves. At 2.0 mg/L Cr stress, 10 and 26 metabolites changed compared to the control after 7 d and 14 d, respectively. At 8.0 mg/L Cr stress, 24 and 24 metabolites changed significantly after 7 and 14 d, respectively. The data showed that metabolism was affected by the Cr stress concentration and exposure time. Specifically, under the Cr stress, the tricarboxylic acid cycle, glutamine synthetase/glutamate synthase cycle, and partial amino acid metabolic pathways were blocked, inhibiting the normal growth and development of bok choy. The change of citric acid content was the most significant, and the accumulation of citric acid indicated the degree of plant Cr toxicity and resistance. These results would facilitate further dissection of the mechanisms of heavy metal accumulation/tolerance in plants and the effective management of such contamination in vegetable crops by genetic manipulation.


Subject(s)
Brassica , Metals, Heavy , Soil Pollutants , Chromium/toxicity , Environmental Pollution , Metals, Heavy/analysis , Plant Leaves/chemistry , Soil Pollutants/analysis
5.
Microbiome ; 7(1): 98, 2019 06 29.
Article in English | MEDLINE | ID: mdl-31255176

ABSTRACT

BACKGROUND: Western-style diets arouse neuroinflammation and impair emotional and cognitive behavior in humans and animals. Our previous study showed that a high-fructose diet caused the hippocampal neuroinflammatory response and neuronal loss in animals, but the underlying mechanisms remained elusive. Here, alterations in the gut microbiota and intestinal epithelial barrier were investigated as the causes of hippocampal neuroinflammation induced by high-fructose diet. RESULTS: A high-fructose diet caused the hippocampal neuroinflammatory response, reactive gliosis, and neuronal loss in C57BL/6N mice. Depletion of the gut microbiota using broad-spectrum antibiotics suppressed the hippocampal neuroinflammatory response in fructose-fed mice, but these animals still exhibited neuronal loss. Gut microbiota compositional alteration, short-chain fatty acids (SCFAs) reduction, intestinal epithelial barrier impairment, NOD-like receptor family pyrin domain-containing 6 (NLRP6) inflammasome dysfunction, high levels of serum endotoxin, and FITC-dextran were observed in fructose-fed mice. Of note, SCFAs, as well as pioglitazone (a selective peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist), shaped the gut microbiota and ameliorated intestinal epithelial barrier impairment and NLRP6 inflammasome dysfunction in fructose-fed mice. Moreover, SCFAs-mediated NLRP6 inflammasome activation was inhibited by histamine (a bacterial metabolite) in ex vivo colonic explants and suppressed in murine CT26 colon carcinoma cells transfected with NLRP6 siRNA. However, pioglitazone and GW9662 (a PPAR-γ antagonist) exerted no impact on SCFAs-mediated NLRP6 inflammasome activation in ex vivo colonic explants, suggesting that SCFAs may stimulate NLRP6 inflammasome independently of PPAR-γ activation. SCFAs and pioglitazone prevented fructose-induced hippocampal neuroinflammatory response and neuronal loss in mice. Additionally, SCFAs activated colonic NLRP6 inflammasome and increased DCX+ newborn neurons in the hippocampal DG of control mice. CONCLUSIONS: Our findings reveal that gut dysbiosis is a critical factor for a high-fructose diet-induced hippocampal neuroinflammation in C57BL/6N mice possibly mediated by impairing intestinal epithelial barrier. Mechanistically, the defective colonic NLRP6 inflammasome is responsible for intestinal epithelial barrier impairment. SCFAs can stimulate NLRP6 inflammasome and ameliorate the impairment of intestinal epithelial barrier, resulting in the protection against a high-fructose diet-induced hippocampal neuroinflammation and neuronal loss. This study addresses a gap in the understanding of neuronal injury associated with Western-style diets. A new intervention strategy for reducing the risk of neurodegenerative diseases through SCFAs supplementation or dietary fiber consumption is emphasized.


Subject(s)
Dysbiosis/chemically induced , Fatty Acids, Volatile/administration & dosage , Fructose/adverse effects , Hippocampus/drug effects , Inflammation/chemically induced , Animals , Doublecortin Protein , Gastrointestinal Microbiome , Hippocampus/pathology , Inflammasomes , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Neuroimmunomodulation/drug effects , Pioglitazone/administration & dosage
6.
Acta Pharmacol Sin ; 40(7): 879-894, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30568253

ABSTRACT

Increasing evidence has demonstrated that excessive fructose intake induces liver fibrosis. Epithelial-mesenchymal transition (EMT) driven by transforming growth factor-ß1 (TGF-ß1)/mothers against decapentaplegic homolog (Smad) signaling activation promotes the occurrence and development of liver fibrosis. Magnesium isoglycyrrhizinate is clinically used as a hepatoprotective agent to treat liver fibrosis, but its underlying molecular mechanism has not been identified. Using a rat model, we found that high fructose intake reduced microRNA (miR)-375-3p expression and activated the janus-activating kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) cascade and TGF-ß1/Smad signaling, which is consistent with the EMT and liver fibrosis. To further verify these observations, BRL-3A cells and/or primary rat hepatocytes were exposed to high fructose and/or transfected with a miR-375-3p mimic or inhibitor or treated with a JAK2 inhibitor, and we found that the low expression of miR-375-3p could induce the JAK2/STAT3 pathway to activate TGF-ß1/Smad signaling and promote the EMT. Magnesium isoglycyrrhizinate was found to ameliorate high fructose-induced EMT and liver fibrosis in rats. More importantly, magnesium isoglycyrrhizinate increased miR-375-3p expression to suppress the JAK2/STAT3 pathway and TGF-ß1/Smad signaling in these animal and cell models. This study provides evidence showing that magnesium isoglycyrrhizinate attenuates liver fibrosis associated with a high fructose diet.


Subject(s)
Liver Cirrhosis/drug therapy , MicroRNAs/metabolism , Saponins/therapeutic use , Signal Transduction/drug effects , Triterpenes/therapeutic use , Animals , Cell Line , Epithelial-Mesenchymal Transition/drug effects , Fructose , Janus Kinase 2/metabolism , Liver Cirrhosis/chemically induced , Male , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Saponins/pharmacology , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta1/metabolism , Triterpenes/pharmacology
7.
Data Brief ; 18: 69-75, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29896493

ABSTRACT

The data presented herein are related to the research article entitled "Magnesium isoglycyrrhizinate blocks fructose-induced hepatic NF-κB/NLRP3 inflammasome activation and lipid metabolism disorder" (Zhao et al., 2017) [1]. This article describes the effects of magnesium isoglycyrrhizinate on 24-h food or water intake in fructose-fed rats at 15-week. In addition, this article expands the effect of magnesium isoglycyrrhizinate on the animal body weight change during 1-17 week. The field dataset is made publicly available to enable critical or extended analyzes.

8.
Biomed Pharmacother ; 103: 1415-1428, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29864926

ABSTRACT

Depression is a mental illness comorbid risk factor for glucose intolerance worldwide. Chaihu-shugan san, a 'Shu-Gan' formula in traditional Chinese medicine, is clinically used in the treatment of depression. The aim of this study was to investigate whether Chaihu-shugan san improved glucose tolerance with its antidepressant activity in rat model of depression and explore the mechanisms underlying its action on liver-brain inflammation axis. After 6 weeks of chronic unpredictable mild stress (CUMS) procedure, male Wistar rats were given Chaihu-shugan san water extract (925 and 1850 mg/kg) by gavage for the next 6 consecutive weeks. Sucrose consumption test was used to assess animal depressive-like behaviors. Oral glucose tolerance test (OGTT) was employed to define the status of glucose tolerance in rats. Serum alanine aminotransferase (ALT) and interleukin-1 beta (IL-1ß) were measured by commercial kits, respectively. Western blot was used to detect the expression of key proteins in inflammatory signaling cascades including toll-like receptor 4 (TLR4), myeloid differentiation protein 88 (MyD88), nuclear factor-kappa B (NF-κB), Nod-like receptor family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing CARD (ASC), cysteinyl aspartate specific proteinase-1 (Caspase-1) and IL-1ß, as well as insulin signaling in liver and prefrontal cortex of rats. Immunohistochemical staining or immunofluorescence staining of NF-κB, and nuclear/cytoplasmic ratio of NF-κB by Western blot were used to describe its nuclear entry in liver and prefrontal cortex of rats. RT-qPCR and Western blot analysis, as well as microRNA-155 (miR-155) mimic or inhibitor transfection were used to explore possible association of MyD88 and miR-155. In this study, Chaihu-shugan san increased sucrose consumption and reduced serum glucose levels in CUMS rats, showing its antidepressant activity with glucose tolerance improvement. Chaihu-shugan san reduced serum levels of ALT and IL-1ß in this animal model. Furthermore, this formula inhibited hepatic and prefrontal cortical inflammatory response by suppressing TLR4/MyD88/NF-κB pathway and NLRP3 inflammasome activation, and improved insulin signaling in CUMS rats. More importantly, Chaihu-shugan san up-regulated miR-155 expression in liver and prefrontal cortex of CUMS rats. These results provide direct evidence that Chaihushugan San can ameliorate depressive-like behaviors by inhibiting liver-brain inflammation axis.


Subject(s)
Glucose Intolerance/complications , Inflammation/drug therapy , Insulin/metabolism , Liver/metabolism , Plant Extracts/therapeutic use , Prefrontal Cortex/metabolism , Signal Transduction , Stress, Psychological/metabolism , Alanine Transaminase/metabolism , Animals , Behavior, Animal/drug effects , Blood Glucose/metabolism , Cell Line , Chronic Disease , Depression/drug therapy , Depression/etiology , Glucose Intolerance/blood , Glucose Intolerance/genetics , Inflammasomes/metabolism , Inflammation/blood , Inflammation/complications , Inflammation/pathology , Interleukin-1beta/metabolism , Liver/drug effects , Liver/physiopathology , Male , MicroRNAs/genetics , MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Plant Extracts/pharmacology , Prefrontal Cortex/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Signal Transduction/drug effects , Stress, Psychological/blood , Stress, Psychological/complications , Stress, Psychological/genetics , Water/chemistry
9.
Eur J Pharmacol ; 809: 141-150, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28526339

ABSTRACT

Magnesium isoglycyrrhizinate as a hepatoprotective agent possesses immune modulation and anti-inflammation, and treats liver diseases. But its effects on immunological-inflammatory and metabolic profiles for metabolic syndrome with liver injury and underlying potential mechanisms are not fully understood. In this study, magnesium isoglycyrrhizinate alleviated liver inflammation and lipid accumulation in fructose-fed rats with metabolic syndrome. It also suppressed hepatic inflammatory signaling activation by reducing protein levels of phosphorylation of nuclear factor-kappa B p65 (p-NF-κB p65), inhibitor of nuclear factor kappa-B kinase α/ß (p-IKKα/ß) and inhibitor of NF-κB α (p-IκBα) as well as nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC) and Caspase-1 in rats, being consistent with its reduction of interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α) and IL-6 levels. Furthermore, magnesium isoglycyrrhizinate modulated lipid metabolism-related genes characterized by up-regulating peroxisome proliferator-activated receptor-α (PPAR-α) and carnitine palmitoyl transferase-1 (CPT-1), and down-regulating sensor for fatty acids to control-1 (SREBP-1) and stearoyl-CoA desaturase 1 (SCD-1) in the liver of fructose-fed rats, resulting in the reduction of triglyceride and total cholesterol levels. These effective actions were further confirmed in fructose-exposed BRL-3A and HepG2 cells. The molecular mechanisms underpinning these observations suggest that magnesium isoglycyrrhizinate may inhibit NF-κB/NLRP3 inflammasome activation to reduce immunological-inflammatory response, which in turn may prevent liver lipid metabolic disorder and accumulation under high fructose condition. Thus, blockade of NF-κB/NLRP3 inflammasome activation and lipid metabolism disorder by magnesium isoglycyrrhizinate may be the potential therapeutic approach for improving fructose-induced liver injury with metabolic syndrome in clinic.


Subject(s)
Fructose/adverse effects , Inflammasomes/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Down-Regulation/drug effects , Hep G2 Cells , Humans , Liver/metabolism , Liver/pathology , Male , PPAR alpha/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism
10.
Planta Med ; 80(1): 39-47, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24338552

ABSTRACT

Betaine as a dietary alkaloid has attracted the attention of patients with kidney diseases. This study aimed to investigate the effects of betaine on serum uric acid levels and kidney function, and explore their underlying mechanisms in potassium oxonate-induced hyperuricemic mice. Betaine at 5, 10, 20, and 40 mg/kg was orally administered to hyperuricemic mice for 7 days and found to significantly reduce serum uric acid levels and increase fractional excretion of uric acid in hyperuricemic mice in a dose-dependent manner. It effectively restored renal protein level alterations of urate transport-related molecular proteins urate transporter 1, glucose transporter 9, organic anion transporter 1, and ATP-binding cassette subfamily G member 2 in this model, possibly resulting in the enhancement of kidney urate excretion. Moreover, betaine reduced serum creatinine and blood urea nitrogen levels and affected urinary levels of beta-2-microglobulin and N-acetyl-beta-D-glucosaminidase as well as upregulated renal protein levels of organic cation/carnitine transporters OCT1, OCTN1, and OCTN2, resulting in kidney function improvement in hyperuricemic mice. The findings from this study provide evidence that betaine has anti-hyperuricemic and nephroprotective actions by regulating protein levels of these renal organic ion transporters in hyperuricemic mice.


Subject(s)
Betaine/pharmacology , Hyperuricemia/drug therapy , Kidney/drug effects , Kidney/physiology , Uric Acid/blood , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Acetylglucosaminidase/metabolism , Animals , Blood Urea Nitrogen , Carrier Proteins/metabolism , Creatinine/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Glucose Transport Proteins, Facilitative/metabolism , Hyperuricemia/physiopathology , Male , Membrane Proteins/metabolism , Mice , Octamer Transcription Factor-1/metabolism , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Solute Carrier Family 22 Member 5 , Symporters , beta 2-Microglobulin/urine
11.
Br J Pharmacol ; 169(6): 1352-71, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23647015

ABSTRACT

BACKGROUND AND PURPOSE: Thioredoxin-interacting protein (TXNIP), a regulator of cellular oxidative stress, has been associated with activation of NOD-like receptor 3 (NLRP3) inflammasome, inflammation and lipid metabolism, suggesting it has a role in the pathogenesis of non-alcoholic fatty liver disease (NAFLD) in diabetes. In this study we investigated whether TXNIP is involved in type 1 diabetes-associated NAFLD and whether antioxidants, quercetin and allopurinol, alleviate NAFLD by targeting TXNIP. EXPERIMENTAL APPROACH: Diabetes was induced in male Sprague-Dawley rats by a single i.p. injection of 55 mg · kg⁻¹ streptozotocin. Quercetin and allopurinol were given p.o. to diabetic rats for 7 weeks. Hepatic function, oxidative stress, inflammation and lipid levels were determined. Rat BRL-3A and human HepG2 cells were exposed to high glucose (30 mM) in the presence and absence of antioxidants, TXNIP siRNA transfection or caspase-1 inhibitor, Ac-YVAD-CMK. KEY RESULTS: Quercetin and allopurinol significantly inhibited the TXNIP overexpression, activation of NLRP3 inflammasome, down-regulation of PPARα and up-regulation of sterol regulatory element binding protein-1c (SREBP-1c), SREBP-2, fatty acid synthase and liver X receptor α, as well as elevation of ROS and IL-1ß in diabetic rat liver. These effects were confirmed in hepatocytes in vitro and it was further shown that TXNIP down-regulation contributed to the suppression of NLRP3 inflammasome activation, inflammation and changes in PPARα and SREBPs. CONCLUSIONS AND IMPLICATIONS: Inhibition of hepatic TXNIP by quercetin and allopurinol contributes to the reduction in liver inflammation and lipid accumulation under hyperglycaemic conditions. The targeting of hepatic TXNIP by quercetin and allopurinol may have therapeutic implications for prevention of type 1 diabetes-associated NAFLD.


Subject(s)
Allopurinol/therapeutic use , Antioxidants/therapeutic use , Carrier Proteins/antagonists & inhibitors , Diabetes Mellitus, Type 1/complications , Fatty Liver/prevention & control , Liver/drug effects , Quercetin/therapeutic use , Allopurinol/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/administration & dosage , Antioxidants/metabolism , Antioxidants/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line , Dietary Supplements , Fatty Liver/complications , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Silencing , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Lipid Metabolism/drug effects , Liver/immunology , Liver/metabolism , Liver/pathology , Male , Molecular Targeted Therapy , Non-alcoholic Fatty Liver Disease , Oxidative Stress/drug effects , Quercetin/administration & dosage , Quercetin/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism
12.
Toxicol Lett ; 201(1): 72-9, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21167265

ABSTRACT

Aristolochic acid (AA) nephropathy exhibits early proximal tubular injury and fatty acid metabolic disorder. In order to study the unrecognized abnormalities of organic ion transporters and fatty acid metabolism indicators in AA nephropathy, Wistar rats were orally administrated with vehicle, 10 and 20mg/kg AA once daily for 7 days, respectively. At day 8, significant reduction of body weight and right kidney weight, as well as elevation of plasma blood urea nitrogen (BUN) levels, renal long-chain fatty acids (LCFAs), non-esterified fatty acids (NEFA) and triglycerides (TG) contents were observed in AA-treated rats, accompanying with down-regulation of renal rOAT1/3, rOCT1/2 and rOCTN1/2 expressions. OCTN2 particularly transports l-carnitine through cell membrane. AA treatment also induced a significant decrease of L-carnitine levels in renal cortex of rats. Down-regulation of peroxisome proliferator-activated receptor alpha (rPPARα) and carnitine acyltransferase 1 (rCPT1), and up-regulation of acetyl coenzyme A carboxylase 1/2 (rACC1/2) in renal cortex were detected in AA-treated rats. These results indicate that alterations of organic ion transportation and fatty acid metabolism are part of AA-induced nephropathy (AAN), contribute to the altered urinary metabolic profile and may lead to further proximal tubule injury in rats.


Subject(s)
Aristolochic Acids/toxicity , Fatty Acids/metabolism , Kidney/drug effects , Membrane Transport Proteins/genetics , Acetyltransferases/genetics , Animals , Blood Urea Nitrogen , Carnitine/analysis , Catecholamine Plasma Membrane Transport Proteins/genetics , Creatinine/blood , Kidney/metabolism , Male , Membrane Transport Proteins/analysis , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 2 , Rats , Rats, Wistar , Solute Carrier Family 22 Member 5
SELECTION OF CITATIONS
SEARCH DETAIL
...