Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 22357, 2023 12 15.
Article in English | MEDLINE | ID: mdl-38102220

ABSTRACT

Solar ultraviolet B (UVB) radiation triggers excessive inflammation, disrupting the epidermal barrier, and can eventually cause skin cancer. A previous study reported that under UVB irradiation, epidermal keratinocytes synthesize the proopiomelanocortin-derived peptide ß-endorphin, which is known for its analgesic effect. However, little is known about the role of ß-endorphin in UVB-exposed skin. Therefore, in this study, we aimed to explore the protective role of ß-endorphin against UVB irradiation-induced damage to the skin barrier in normal human keratinocytes (NHKs) and on a human skin equivalent model. Treatment with ß-endorphin reduced inflammatory responses in UVB-irradiated NHKs by inactivating the NF-κB signaling pathway. Additionally, we found that ß-endorphin treatment reversed UVB-induced abnormal epidermal proliferation and differentiation in NHKs and, thus, repaired the skin barrier in UVB-treated skin equivalents. The observed effects of ß-endorphin on UVB-irradiated NHKs were mediated via blockade of the Akt/mTOR signaling pathway. These results reveal that ß-endorphin might be useful against UVB-induced skin injury, including the disruption of the skin barrier function.


Subject(s)
Epidermis , beta-Endorphin , Humans , beta-Endorphin/metabolism , Epidermis/metabolism , Keratinocytes/metabolism , Signal Transduction , Inflammation/prevention & control , Inflammation/metabolism , Ultraviolet Rays/adverse effects , Mechanistic Target of Rapamycin Complex 1/metabolism
2.
Chemistry ; 29(71): e202302897, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-37864280

ABSTRACT

Contamination of water supplies by polyfluoroalkyl substances, notably perfluorooctanesulfonate (PFOS) and perfluorooctanoic acid (PFOA), has serious health and environmental consequences. Therefore, the development of straightforward and effective means of monitoring and removing PFASs is urgently required. In this study, we report a rapid and sensitive method for the detection of PFOS and PFOA in water that rely on the J-aggregate formation of meso-ester-BODIPY dyes. The dye C10-mim, which contains a hydrophilic methylimidazolium group and a hydrophobic alkylated BODIPY, self-assembles in water into weakly green-emissive micellar assemblies. Upon binding to PFOS or PFOA, a spontaneous disassembly and reorganization forms orange-emissive J-aggregates. The rapid formation (≤5 s) of J-aggregates and the accompanying spectral shifts provide a superior sensing performance, with excellent sensitivity (limit of detection=0.18 ppb for PFOS) and distinct chromogenic and fluorogenic "turn-on" responses.

3.
Chemistry ; 29(23): e202203739, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-36734188

ABSTRACT

Urokinase-type plasminogen activator receptor (uPAR) is a glycolipid-anchored protein located on the cell surface that is implicated in the promotion of metastasis. New fluorescent probes for the detection of uPAR expression that feature a rapid "turn-on" response are reported here. They consist of a donor-π-acceptor-based fluorophore conjugated with a uPAR-binding AE105 peptide. The resulting AE105-coupled uPAR-targeting probes are weakly emissive in aqueous buffer solutions; however, a fluorescence "turn-on" signal is instantly triggered upon specific binding to uPAR (KD =63.2 nM for P1 and 49.5 nM for P2), which restricts the rotational deactivation of the fluorophore. Applications of the probes were demonstrated in the imaging of uPAR overexpressed on the membrane of cancer cell and in a cell-based uPAR inhibitor assay.


Subject(s)
Fluorescent Dyes , Receptors, Urokinase Plasminogen Activator , Receptors, Urokinase Plasminogen Activator/metabolism , Cell Membrane/metabolism
4.
Front Neurogenom ; 4: 1136362, 2023.
Article in English | MEDLINE | ID: mdl-38234497

ABSTRACT

Objective: The benefits of long-term consumption of green tea on the brain are well known. However, among many ingredients of green tea, the acute effects of (-)-gallocatechin gallate-rich green tea extract (GCG-GTE), have received comparatively less attention. Herein, we investigated the acute effects of oral ingestion of green tea with GCG-GTE, which contains close replicas of the ingredients of hot green tea, on task-dependent hemodynamics in the prefrontal cortex of healthy adult human brains. Methods: In this randomized, double-blind, placebo-controlled, parallel group trial, 35 healthy adults completed computerized cognitive tasks that demand activation of the prefrontal cortex at baseline and 1 h after consumption of placebo and 900 mg of GCG-GTE extract supplement. During cognitive testing, hemodynamic responses (change in HbO2 concentration) in the prefrontal cortex were assessed using functional near-infrared spectroscopy (fNIRS). Results: In fNIRS data, significant group x session interactions were found in the left (p = 0.035) and right (p = 0.036) dorsolateral prefrontal cortex (DLPFC). In behavioral data, despite the numerical increase in the GCG-GTE group and the numerical decrease in the Placebo group, no significant differences were observed in the cognitive performance measure between the groups. Conclusion: The result suggests a single dose of orally administered GCG-GTE can reduce DLPFC activation in healthy humans even with increased task demand. GCG-GTE is a promising functional material that can affect neural efficiency to lower mental workload during cognitively demanding tasks. However, further studies are needed to verify this.

5.
Biomed Pharmacother ; 156: 113864, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36252351

ABSTRACT

Skin aging is a major risk factor for the dermal diseases, and interventions to attenuate cellular senescence are expected to reduce the risk for age-related diseases involving skin atrophy. However, blocking cell death or extending proliferation causally results in side effects and an increased cancer risk. For identification of a safer approach, we focused on PDK1 inhibition, which could revert cellular senescence and reduce senescence factors in skin in vitro, in a human skin equivalent model and in an exploratory, placebo-controlled, interventional trial. Natural phytochemical kaempferol tetrasaccharides resulted in a significant reduction in cellular senescence, and an increase in collagen fiber was observed in the skin cell and human skin equivalent. Clinical enhancement in skin appearance was noted in multiple participants, and an immunohistochemical study revealed improvement in the histological appearance of skin tissue and extracellular matrix. This change was associated with relative improvement in histological markers of senescence and clinical appearance of the aged skin and an increase in collagen fiber, an essential factor for preventing skin atrophy and consistency of the basement membrane. These results indicate that PDK1 inhibition is a potentially effective antiaging intervention, suggesting a diagnostic role and preventive actions of PDK1 in senescence-associated skin atrophy.


Subject(s)
Fibroblasts , Kaempferols , Humans , Aged , Kaempferols/pharmacology , Kaempferols/therapeutic use , Skin , Cellular Senescence , Collagen/metabolism , Atrophy/drug therapy , Atrophy/metabolism
6.
Biomaterials ; 289: 121749, 2022 10.
Article in English | MEDLINE | ID: mdl-36055174

ABSTRACT

We report that the electrostatic association between a cyanovinylene (CV)-based cationic dye (CV-N+) and anionic surfactants rapidly induced the formation of self-assembled aggregates of CV-N+ molecules in an aqueous solution, displaying a red-shifted (Δλ = 100 nm), broad excimer emission at 650 nm. The anionic-surfactant-mediated excimer-forming properties of CV-N+ were further exploited as a versatile platform for the design of simple fluorescent "light-up" sensing systems for the specific and efficient detection of enzymatic activity (i.e., phospholipase D) and small molecules (i.e., bisulfite ions), respectively, without the complicated functionalization of fluorophores with suitable recognition groups. In these systems, which can be ensembled using various substrates, the cationic CV-N+ dye interacts with an anionic surfactant, which is the product of an analyte-induced specific reaction, thus triggering the formation of emissive aggregates.


Subject(s)
Phospholipase D , Surface-Active Agents , Cations , Fluorescent Dyes/chemistry , Surface-Active Agents/chemistry , Water/chemistry
7.
Chemistry ; 27(49): 12545-12551, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34132430

ABSTRACT

meso-Carboxyl-BODIPY responds to small electronic changes resulting from acyl substitution reactions with a marked change in fluorescence. Herein, the minute changes that accompany the thioester to amide conversion encountered in native chemical ligation (NCL) are exploited in the construction of fluorescent "turn-on" probes. Two fluorogenic probes, 1 a and 4, derived from a meso-thioester-BODIPY scaffold, were designed for the selective detection of cysteine (1 a) and aminopeptidase N (4), respectively. The aromatic (1 a) and aliphatic (4) thioesters of meso-carboxyl-BODIPY are nonfluorescent. However, specific analyte-induced conversion to the meso-amide derivative caused significant spectral changes and a dramatic fluorescence enhancement. Probe 1 a exhibited a large fluorescence "turn-on" response with high selectivity toward cysteine via a tandem NCL reaction. Probe 4 was successfully applied to the monitoring and imaging of endogenous aminopeptidase N in live cancer cells.


Subject(s)
Cysteine , Fluorescent Dyes , Boron Compounds , CD13 Antigens
8.
Proc Natl Acad Sci U S A ; 117(49): 31535-31546, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229519

ABSTRACT

Cellular senescence is defined as a stable, persistent arrest of cell proliferation. Here, we examine whether senescent cells can lose senescence hallmarks and reenter a reversible state of cell-cycle arrest (quiescence). We constructed a molecular regulatory network of cellular senescence based on previous experimental evidence. To infer the regulatory logic of the network, we performed phosphoprotein array experiments with normal human dermal fibroblasts and used the data to optimize the regulatory relationships between molecules with an evolutionary algorithm. From ensemble analysis of network models, we identified 3-phosphoinositide-dependent protein kinase 1 (PDK1) as a promising target for inhibitors to convert the senescent state to the quiescent state. We showed that inhibition of PDK1 in senescent human dermal fibroblasts eradicates senescence hallmarks and restores entry into the cell cycle by suppressing both nuclear factor κB and mTOR signaling, resulting in restored skin regeneration capacity. Our findings provide insight into a potential therapeutic strategy to treat age-related diseases associated with the accumulation of senescent cells.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , Cellular Senescence , Dermis/cytology , Fibroblasts/cytology , Fibroblasts/enzymology , Protein Kinase Inhibitors/pharmacology , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Adult , Cell Cycle/drug effects , Cellular Senescence/drug effects , Computer Simulation , Female , Fibroblasts/drug effects , Humans , Middle Aged , Models, Biological , Phenotype , Phosphoproteins/metabolism , Regeneration/drug effects , Skin Aging/drug effects , Young Adult
9.
Adv Biosyst ; 4(7): e1900248, 2020 07.
Article in English | MEDLINE | ID: mdl-32558394

ABSTRACT

Many studies utilizing animal models have revealed the genetic and pharmacogenetic modulators of the rate of organismal aging. However, finding routes for healthy aging during extended life remains one of the largest questions. With regards to an antiaging reagent, it has been shown that natural phytochemical syringaresinol (SYR) delays cellular senescence by activating sirtuin1 (SIRT1). Here, it is found that SYR treatment results in metabolic changes similar to those observed during dietary restriction (DR). The DR mimetic effects are mediated by FoxO3a-dependent SIRT1 activation and insulin/insuline growth factor-1 signaling modulation. The direct binding of SYR-FoxO3a is identified and this could partially explain the DR-like phenotype. The report gives a clue as to how the longevity gene involves the DR pathway and suggests that natural phytochemicals applied as a geroprotector mimics DR effects.


Subject(s)
Biomimetic Materials/pharmacology , Caloric Restriction , Forkhead Box Protein O3/metabolism , Phytochemicals/pharmacology , Animals , Cellular Reprogramming , Mice , Sirtuin 1/metabolism
10.
Exp Dermatol ; 28(11): 1270-1278, 2019 11.
Article in English | MEDLINE | ID: mdl-31461579

ABSTRACT

Ageing is characterized by the accumulation of chronic and irreversible oxidative damage, chronic inflammation and organ dysfunction. To attenuate these ageing-related changes, various natural phytochemicals are often applied. Trans-communic acid (TCA), an active component of brown pine leaf extract, has antimicrobial and cancer chemopreventive activity and inhibits ultraviolet B (UVB)-induced MMP-1 expression. To determine whether the phytochemical TCA could affect the lifespan of an ageing model, Caenorhabditis elegans prevent ageing-related phenotypes of the skin. Caenorhabditis elegans (C. elegans) wild-type N2 and mutant strains were used in this study to explore the lifespan extension effect of TCA and its mechanism. We estimated lipofuscin accumulation and melanin levels, which are closely associated with skin senescence. Moreover, we explored the mechanism of action associated with ageing attenuation. We performed oxidative stress resistance and thermotolerance assays in C. elegans and surface plasmon resonance analysis of TCA binding with the forkhead box-O3a (FoxO3a) protein. TCA, which is the active component in Korean red pine (Pinus densiflora), attenuated ageing-related changes in skin cells. TCA lowered lipofuscin accumulation in fibroblasts and decreased melanin levels in melanocytes. These protective effects were mediated by activation of the representative longevity gene FoxO3a, which was induced by direct binding with TCA. Interestingly, TCA extended the lifespan of C. elegans, although it did not affect stress resistance, oxidative stress or thermotolerance. These results strongly suggest that TCA prevents the senescent phenotype of model organisms and exhibits beneficial effects on ageing-related skin phenotypes through direct FoxO3a activation.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Diterpenes/pharmacology , Forkhead Transcription Factors/metabolism , Longevity/drug effects , Animals , Caenorhabditis elegans , Cell Line, Tumor , Drug Evaluation, Preclinical , Feasibility Studies , Fibroblasts/drug effects , Humans , Melanocytes/drug effects , Phytotherapy , Pinus
11.
J Invest Dermatol ; 139(3): 648-655, 2019 03.
Article in English | MEDLINE | ID: mdl-30798853

ABSTRACT

Aging is characterized by accumulation of chronic and irreversible oxidative damage, chronic inflammation, and organ dysfunction. Superoxide dismutase (SOD) serves as a major enzyme for cellular superoxide radical metabolism and physiologically regulates cellular redox balance throughout the body. Copper/zinc superoxide dismutase-deficient (SOD1-/-) mice showed diverse phenotypes associated with enhanced oxidative damage in whole organs. Here, we found that oral treatment with syringaresinol (also known as lirioresinol B), which is the active component in the berries of Korean ginseng (Panax ginseng C.A. Meyer), attenuated the age-related changes in Sod1-/- skin. Interestingly, syringaresinol morphologically normalized skin atrophy in Sod1-/- mice and promoted fibroblast outgrowth from Sod1-/- skin in vitro. These protective effects were mediated by the suppression of matrix metalloproteinase-2 overproduction in Sod1-/- skin, but not by increased collagen expression. Syringaresinol also decreased the oxidative damage and the phosphorylation of FoxO3a protein, which was a transcriptional factor of matrix metalloproteinase-2, in Sod1-/- skin. These results strongly suggest that syringaresinol regulates the FoxO3-matrix metalloproteinase-2 axis in oxidative damaged skin and exhibits beneficial effects on age-related skin involution in Sod1-/- mice.


Subject(s)
Forkhead Box Protein O3/genetics , Furans/pharmacology , Lignans/pharmacology , Matrix Metalloproteinase 2/metabolism , Skin Aging/drug effects , Skin Aging/genetics , Superoxide Dismutase-1/genetics , Animals , Atrophy/genetics , Biopsy, Needle , Blotting, Western/methods , Copper/deficiency , Immunohistochemistry , Mice , Mice, Hairless , Polymerase Chain Reaction/methods , Skin Aging/pathology , Superoxide Dismutase/deficiency , Treatment Outcome
13.
Sci Rep ; 6: 39026, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27976725

ABSTRACT

Age-associated immunological dysfunction (immunosenescence) is closely linked to perturbation of the gut microbiota. Here, we investigated whether syringaresinol (SYR), a polyphenolic lignan, modulates immune aging and the gut microbiota associated with this effect in middle-aged mice. Compared with age-matched control mice, SYR treatment delayed immunosenescence by enhancing the numbers of total CD3+ T cells and naïve T cells. SYR treatment induced the expression of Bim as well as activation of FOXO3 in Foxp3+ regulatory T cells (Tregs). Furthermore, SYR treatment significantly enhanced the Firmicutes/Bacteroidetes ratio compared with that in age-matched controls by increasing beneficial bacteria, Lactobacillus and Bifidobacterium, while reducing the opportunistic pathogenic genus, Akkermansia. In addition, SYR treatment reduced the serum level of lipopolysaccharide-binding protein, an inflammatory marker, and enhanced humoral immunity against influenza vaccination to the level of young control mice. Taken together, these findings suggest that SYR may rejuvenate the immune system through modulation of gut integrity and microbiota diversity as well as composition in middle-aged mice, which may delay the immunosenescence associated with aging.


Subject(s)
Aging/immunology , Furans/pharmacology , Gastrointestinal Microbiome/drug effects , Immunosenescence/drug effects , Lignans/pharmacology , Animals , Area Under Curve , Bifidobacterium/drug effects , Bifidobacterium/immunology , Bifidobacterium/physiology , CD3 Complex/immunology , CD3 Complex/metabolism , Female , Forkhead Box Protein O3/immunology , Forkhead Box Protein O3/metabolism , Furans/pharmacokinetics , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Immunosenescence/immunology , Lactobacillus/drug effects , Lactobacillus/immunology , Lactobacillus/physiology , Lignans/pharmacokinetics , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Rats, Sprague-Dawley , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Verrucomicrobia/drug effects , Verrucomicrobia/immunology , Verrucomicrobia/physiology
14.
Nutrients ; 8(4): 199, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27058552

ABSTRACT

This study investigates the in vivo functions of ginseng berry extract (GB) as a therapy for dextran sodium sulfate (DSS)-induced colitis. C57BL/6 mice were given drinking water containing DSS (3%) for eight days to induce acute colitis. At the same time, the mice received an oral dose of GB (50 mg/kg) once daily. The GB-treated mice were less susceptible to the development of acute colitis than were control mice treated with saline, as determined by weight loss, disease activity, and colon histology. The administration of GB to DSS-treated mice also reduced the numbers and inhibited the activation of colon-infiltrating T cells, neutrophils, intestinal CD103(-)CD11c⁺ dendritic cells (cDCs), and macrophages. In addition, GB treatment promoted the migration of CD103⁺CD11c⁺ cDCs and expansion of Foxp3⁺ regulatory T cells in the colons of DSS-treated mice. Similarly, in the DSS-induced chronic colitis model, GB treatment improved the macroscopic and histological appearance of the colon wall when compared to untreated control mice, as indicated by longer colon length and lower histological scores. This is the first report to show that oral administration of GB suppresses immune activation and protects against experimentally induced colitis.


Subject(s)
Colitis/chemically induced , Dextran Sulfate/toxicity , Fruit/chemistry , Panax/chemistry , Plant Extracts/pharmacology , Administration, Oral , Animals , Cell Differentiation/drug effects , Colitis/prevention & control , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/drug effects , Intestines/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Plant Extracts/chemistry , Th1 Cells , Th17 Cells
15.
PLoS One ; 10(6): e0130926, 2015.
Article in English | MEDLINE | ID: mdl-26090808

ABSTRACT

Ginseng extract has been shown to possess certain anti-virus, anti-tumor and immune-activating effects. However, the immunostimulatory effect of ginseng berry extract (GB) has been less well characterized. In this study, we investigated the effect of GB on the activation of mouse dendritic cells (DCs) in vitro and in vivo. GB treatment induced up-regulation of co-stimulatory molecules in bone marrow-derived DCs (BMDCs). Interestingly, GB induced a higher degree of co-stimulatory molecule up-regulation than ginseng root extract (GR) at the same concentrations. Moreover, in vivo administration of GB promoted up-regulation of CD86, MHC class I and MHC class II and production of IL-6, IL-12 and TNF-α in spleen DCs. GB also promoted the generation of Th1 and Tc1 cells. Furthermore, Toll like receptor 4 (TLR4) and myeloid differentiation primary response 88 (MyD88) signaling pathway were essential for DC activation induced by GB. In addition, GB strongly prompted the proliferation of ovalbumin (OVA)-specific CD4 and CD8 T cells. Finally, GB induced DC activation in tumor-bearing mice and the combination of OVA and GB treatment inhibited B16-OVA tumor cell growth in C57BL/6 mice. These results demonstrate that GB is a novel tumor therapeutic vaccine adjuvant by promoting DC and T cell activation.


Subject(s)
Cell Differentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Fruit/chemistry , Panax/chemistry , Plant Extracts/pharmacology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction/drug effects , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism , Toll-Like Receptor 4/metabolism , Xenograft Model Antitumor Assays
16.
Oncotarget ; 6(1): 43-55, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25415049

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) is a master regulator of hypoxic response and has been a prime therapeutic target for ischemia/reperfusion (I/R)-derived myocardial dysfunction and tissue damage. There is also increasing evidence that HIF-1 plays a central role in regulating aging, both through interactions with key longevity factors including Sirtuins and mTOR, as well as by directly promoting longevity in Caenorhabditis elegans.We investigated a novel function and the underlying mechanism of syringaresinol, a lignan compound, in modulation of HIF-1 and protection against cellular damage and death in a cardiomyocyte model of I/R injury. Syringaresinol caused destabilization of HIF-1α following H/R and then protected against hypoxia/reoxygenation (H/R)-induced cellular damage, apoptosis, and mitochondrial dysfunction in a dose-dependent manner. Knock-down of FOXO3 by specific siRNAs completely abolished the ability of syringaresinol to inhibit HIF-1 stabilization and apoptosis caused by H/R. Syringaresinol stimulated the nuclear localization and activity of FOXO3 leading to increased expression of antioxidant genes and decreased levels of reactive oxygen species (ROS) following H/R. Our results provide a new mechanistic insight into a functional role of syringaresinol against H/R-induced cardiomyocyte injury and death. The degradation of HIF-1α through activation of FOXO3 is a potential therapeutic strategy for ischemia-related diseases.


Subject(s)
Forkhead Transcription Factors/metabolism , Furans/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lignans/chemistry , Myocytes, Cardiac/cytology , Animals , Antioxidants/metabolism , Apoptosis , Caspase 3/metabolism , Cell Death , Cell Hypoxia , Cell Line , Cell Survival , Flow Cytometry , Forkhead Box Protein O3 , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondria/pathology , Myocardium/metabolism , Oxygen/chemistry , RNA, Small Interfering/metabolism , Rats , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Up-Regulation
17.
Aging (Albany NY) ; 5(3): 174-82, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23525956

ABSTRACT

Increased SIRT1 expression exerts beneficial effects in transgenic animal models, ameliorating the onset and progression of aging-related disease phenotypes in various organs including the heart. The potential beneficial effects of SIRT1 have made SIRT1 a prime therapeutic target for age-related diseases and considerable efforts led to the identification of small molecule activator of SIRT1 protein. Thus far, however, a small molecule activator of SIRT1 gene expression has not been reported. Here, we report that syringaresinol, isolated from Panax ginseng berry pulp, is an activator of SIRT1 gene expression. Using human umbilical endothelial cells (HUVECs), we show that syringaresinol treatment induced binding of FOXO3 to the SIRT1 promoter in a sequence-specific manner, leading to induction of SIRT1 expression. Increased SIRT1 expression in HUVECs by syringaresinol treatment delayed cellular senescence and improved various markers of endothelial functions in a FOXO3 dependent manner. Collectively, these findings bring to light a new transcription activator of SIRT1 that may have therapeutic potential.


Subject(s)
Forkhead Transcription Factors/genetics , Furans/pharmacology , Lignans/pharmacology , Sirtuin 1/genetics , Cells, Cultured , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Promoter Regions, Genetic/drug effects , Sirtuin 1/metabolism
18.
J Surg Res ; 159(1): 497-502, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19321178

ABSTRACT

BACKGROUND: Curcumin is a nontoxic, hepatoprotective antioxidant. It has been shown to efficiently scavenge oxygen free radicals, increase intracellular glutathione concentrations, and prevent lipid peroxidation in rat hepatocytes. Moreover, it has strong anti-inflammatory effects. In the present study we assessed its effect in a model of liver regeneration impaired by bacterial infections. MATERIAL AND METHODS: Male Sprague-Dawley rats underwent sham operation, cecal ligation and puncture (CLP), synchronous partial hepatectomy (PH), and CLP or synchronous PH+CLP with perioperative application of curcumin (100 mg per kg bodyweight per d) 48 h before surgery. Rats were sacrificed 24 h after surgery. Liver function was analyzed by measuring the serum albumin, serum bilirubin, and bile production. The local inflammatory response in the liver tissue was evaluated by quantification of TNF-alpha, IL-6 mRNA, and quantification of IL-1beta by ELISA. In addition, hepatic concentrations of reduced glutathione (GSH) and the oxidized disulfide dimer of glutathione (GSSG) were measured for determination of the redox state. RESULTS: After simultaneous PH+CLP curcumin significantly reduced the expression of TNF-alpha and IL-6 mRNA in the liver tissue. The IL-1beta concentration in the liver was also slightly, but not significantly, lower in the curcumin group. A severe depletion of hepatic glutathione was found in the PH+CLP group. This was reversed by curcumin application, after which the GSH to GSSG ratio increased markedly. The hepatocellular damage, measured by ALT liberation, was significantly lower in the curcumin treated group. The relative liver weight in the curcumin group was significantly higher 24 h after PH+CLP. However, hepatocellular proliferation parameters were not significantly improved by antioxidative treatment with curcumin. Only the Ki-67 index was slightly higher in the curcumin treated PH+CLP group (14+/-3%) than in the untreated PH+CLP group (7%+/-3%). The hepatocyte density was significantly lower in the curcumin group than in the corresponding untreated group. CONCLUSION: In the present model, curcumin revealed significant hepatoprotective effects with stabilization of redox state, reduced liberation of liver enzymes, and attenuated expression of pro-inflammatory cytokines. However, the hepatocellular proliferation was not significantly influenced.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Curcumin/therapeutic use , Inflammation/drug therapy , Liver Regeneration/drug effects , Oxidative Stress/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bacterial Infections/drug therapy , Curcumin/pharmacology , Glutathione/metabolism , Hepatectomy , Inflammation/metabolism , Liver/metabolism , Liver/pathology , Liver Function Tests , Male , Rats , Rats, Sprague-Dawley
20.
Exp Anim ; 57(5): 461-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18946183

ABSTRACT

Adiponectin, which is expressed exclusively in adipose tissue, has been shown to increase fatty acid oxidation via activation of AMP-activated kinase (AMPK) and phosphorylation of acetyl CoA carboxylase (ACC). ACC phosphorylation and carnitine palmitoyl-transferase-1 (CPT1) activity have been shown to be rate controlling factors in fatty acid oxidation. In high fat diet (HFD)-induced obese mice, we analyzed the time-course of changes in the expression of adiponectin and lipid oxidative enzymes induced by treatment with bisphenol A diglycidyl ether (BADGE) or caffeine for 8 weeks, and investigated whether the changes of adiponectin and lipid oxidative enzymes expression correlated with reduced adiposity or steatosis after 8 weeks of treatment. After 8 weeks of treatment, BADGE and caffeine had reduced body weight and epididymal adipose tissue weight in mice fed HFD, and markedly reduced the number of fatty droplets in the liver. Interestingly, the expression of adiponectin and lipid oxidative enzymes significantly increased after 2 weeks of treatment. These results indicate that the expression of adiponectin and lipid oxidative enzymes in the early stages of BADGE or caffeine treatment correlated well with the long-term anti-obesity effects.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Adiponectin/analysis , Biphenyl Compounds/pharmacology , Caffeine/pharmacology , Carnitine O-Palmitoyltransferase/metabolism , Dietary Fats/administration & dosage , Fatty Liver/metabolism , Obesity/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...