Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
J Agric Food Chem ; 72(13): 7167-7178, 2024 Apr 03.
Article En | MEDLINE | ID: mdl-38511978

IAVPGEVA, an octapeptide derived from soybean 11S globulin hydrolysis, also known as SGP8, has exhibited regulatory effects on lipid metabolism, inflammation, and fibrosis in vitro. Studies using MCD and HFD-induced nonalcoholic steatohepatitis (NASH) models in mice show that SGP8 attenuates hepatic injury and metabolic disorders. Mechanistic studies suggest that SGP8 inhibits the JNK-c-Jun pathway in L02 cells and liver tissue under metabolic stress and targets DPP4 with DPP4 inhibitory activity. In conclusion, the results suggest that SGP8 is an orally available DPP4-targeting peptide with therapeutic potential in NASH.


Globulins , Non-alcoholic Fatty Liver Disease , Soybean Proteins , Mice , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Dipeptidyl Peptidase 4/metabolism , Liver/metabolism , Globulins/metabolism , Mice, Inbred C57BL , Disease Models, Animal
2.
Food Funct ; 14(17): 8018-8029, 2023 Aug 29.
Article En | MEDLINE | ID: mdl-37593938

The lacto-ghrestatin derived nonapeptide (LGP9), a bioactive peptide derived from lacto-ghrestatin in bovine milk with the sequence of LIVTQTMKG, was investigated to determine its effects on islet ß-cell dedifferentiation and associated mechanisms in type 2 diabetes mellitus (T2DM). On the animal level, type-2-diabetic (T2D) mice were generated by high-fat-diet (HFD) and streptozocin (STZ). LGP9 was given to T2D mice for four weeks at doses of 1 mg kg-1, 3 mg kg-1, and 9 mg kg-1. A variety of techniques (immunohistochemistry, western blot, QPCR, and ELISA) were employed to evaluate the impact of LGP9 on the diabetic injury. On the cellular level, the pancreatic cell lines, Rin-m5f cells and Min6 cells, were treated with high-glucose (HG) and high-glucose-high-lipid (HG/PA), respectively. The cell models were established to investigate the mechanism of LGP9 treatment on the islet ß-cell dedifferentiation. For the mechanism study, the PI3K/Akt/FOXO1 pathway was investigated by inhibiting FOXO1 with its inhibitor and siRNA. Results showed that LGP9 improved the ß-cell dedifferentiation, prevented the EMT process, and upregulated the PI3K/Akt/FOXO1 signaling in the pancreas of T2D mice. In addition, LGP9 promoted the structural and functional recovery of pancreatic islets and shielded the liver tissue in T2D mice. From the cellular level data, LGP9 prevented ß-cell dedifferentiation and EMT occurrence. To a certain extent, the inhibition of FOXO1 restored PI3K/Akt/FOXO1 pathway activation and prevented ß-cell dedifferentiation. In conclusion, these findings suggest that LGP9 ameliorated pancreatic ß-cell dedifferentiation via PI3k/Akt/FOXO1 signaling in vivo and in vitro.


Diabetes Mellitus, Type 2 , Animals , Mice , Diabetes Mellitus, Type 2/drug therapy , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Milk , Cell Dedifferentiation , Peptides , Glucose
3.
Int J Biol Macromol ; 250: 126080, 2023 Oct 01.
Article En | MEDLINE | ID: mdl-37536405

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant is a heavily mutated virus and designated as a variant of concern. To investigate the codon usage pattern of this new variant, we performed mutation and codon bias analysis for Omicron as well as for its sub-lineages BA.1 and BA.2 and compared them with the original SARS-CoV-2 and the Delta variant sequences obtained in this study. Our results indicate that the sub-lineage BA.1 and BA.2 have up to 23 sites of difference on the spike protein, which have minimal impact on function. The Omicron variant and its sub-lineages have similar codon usage patterns and A/U ending codons appear to be preferred over G/C ending codons. The Omicron has a lower degree of codon usage bias in spite of evidence that natural selection, mutation pressure and dinucleotide abundance shape the codon usage bias of Omicron, with natural selection being more significant on BA.2 than the other sub-lineages of Omicron. The codon usage pattern of Omicron variant that we explored provides valid information for a clearer understanding of Omicron and its sub-lineages, which could find application in vaccine development and optimization.


COVID-19 , Codon Usage , Humans , Codon Usage/genetics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , COVID-19/genetics , Mutation
4.
Life Sci ; 308: 120907, 2022 Nov 01.
Article En | MEDLINE | ID: mdl-36037870

BACKGROUND AND AIMS: Lacto-ghrestatin is abovine milk-derived peptide with the sequence of LIVTQTMKG, named LGP9 here. This study aimed to investigate protective effects of LGP9 on diabetic ß cells in vivo and in vitro. METHODS AND RESULTS: Type-1-diabetic (T1D) mice were generated by alloxan (ALX; 50 mg/kg, i.v.) and received a four-week treatment schedule of LGP9 at 0.3 mg/kg and 1 mg/kg. Related biochemical parameters were analyzed, and the protein expression was evaluated by Western blotting. The results showed that LGP9 decreased body weight, water consumption and blood glucose, improved oxygen stress and upregulated IRS2/PI3K/Akt signaling in the pancreas of T1D mice. To further investigate the mechanism of LGP9 on the preventive effect of the pancreas, Rin-m5f cells were treated with 15 mM alloxan and followed with LGP9 at 30 µM and 90 µM. The results indicated that LGP9 rebalanced oxygen stress levels, increased cell proliferation, decreased cell apoptosis and activated IRS2/PI3K/Akt signaling. CONCLUSION: LGP9 ameliorated alloxan-injured pancreatic ß cells through IRS2/PI3K/Akt signaling. The finding provides important help for the research and development of LGP9 in therapeutics of diabetes.


Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Alloxan , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Secreting Cells/metabolism , Mice , Milk/metabolism , Oxygen/metabolism , Peptides/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
5.
Circulation ; 136(21): 2051-2067, 2017 Nov 21.
Article En | MEDLINE | ID: mdl-28947430

BACKGROUND: Pathological cardiac hypertrophy induced by stresses such as aging and neurohumoral activation is an independent risk factor for heart failure and is considered a target for the treatment of heart failure. However, the mechanisms underlying pathological cardiac hypertrophy remain largely unknown. We aimed to investigate the roles of SIRT2 in aging-related and angiotensin II (Ang II)-induced pathological cardiac hypertrophy. METHODS: Male C57BL/6J wild-type and Sirt2 knockout mice were subjected to the investigation of aging-related cardiac hypertrophy. Cardiac hypertrophy was also induced by Ang II (1.3 mg/kg/d for 4 weeks) in male C57BL/6J Sirt2 knockout mice, cardiac-specific SIRT2 transgenic (SIRT2-Tg) mice, and their respective littermates (8 to ≈12 weeks old). Metformin (200 mg/kg/d) was used to treat wild-type and Sirt2 knockout mice infused with Ang II. Cardiac hypertrophy, fibrosis, and cardiac function were examined in these mice. RESULTS: SIRT2 protein expression levels were downregulated in hypertrophic hearts from mice. Sirt2 knockout markedly exaggerated cardiac hypertrophy and fibrosis and decreased cardiac ejection fraction and fractional shortening in aged (24-month-old) mice and Ang II-infused mice. Conversely, cardiac-specific SIRT2 overexpression protected the hearts against Ang II-induced cardiac hypertrophy and fibrosis and rescued cardiac function. Mechanistically, SIRT2 maintained the activity of AMP-activated protein kinase (AMPK) in aged and Ang II-induced hypertrophic hearts in vivo as well as in cardiomyocytes in vitro. We identified the liver kinase B1 (LKB1), the major upstream kinase of AMPK, as the direct target of SIRT2. SIRT2 bound to LKB1 and deacetylated it at lysine 48, which promoted the phosphorylation of LKB1 and the subsequent activation of LKB1-AMPK signaling. Remarkably, the loss of SIRT2 blunted the response of AMPK to metformin treatment in mice infused with Ang II and repressed the metformin-mediated reduction of cardiac hypertrophy and protection of cardiac function. CONCLUSIONS: SIRT2 promotes AMPK activation by deacetylating the kinase LKB1. Loss of SIRT2 reduces AMPK activation, promotes aging-related and Ang II-induced cardiac hypertrophy, and blunts metformin-mediated cardioprotective effects. These findings indicate that SIRT2 will be a potential target for therapeutic interventions in aging- and stress-induced cardiac hypertrophy.


Cardiomegaly/prevention & control , Metformin/pharmacology , Myocardium/enzymology , Sirtuin 2/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Acetylation , Age Factors , Aging/metabolism , Angiotensin II , Animals , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Cells, Cultured , Disease Models, Animal , Fibrosis , Genetic Predisposition to Disease , Lysine , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocardium/pathology , Phenotype , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/pharmacology , Rats , Signal Transduction/drug effects , Sirtuin 2/deficiency , Sirtuin 2/genetics , Stroke Volume/drug effects , Ventricular Remodeling/drug effects
6.
Sci Rep ; 6: 36576, 2016 11 07.
Article En | MEDLINE | ID: mdl-27819261

The homeodomain transcription factor Nkx2.5/Csx is critically essential for heart specification, morphogenesis, and homeostasis. Acetylation/deacetylation is important for the localization, stability and activation of transcription factors. It remains unknown how Nkx2.5 is deacetylated and how Nkx2.5 acetylation determines its activity. In this study, we provide evidence that the NAD+-dependent class III protein deacetylase SIRT1 deacetylates Nkx2.5 in cardiomyocytes and represses the transcriptional activity of Nkx2.5. We show that SIRT1 interacts with the C-terminus of Nkx2.5 and deacetylates Nkx2.5 at lysine 182 in the homeodomain. The mutation of Nkx2.5 at lysine 182 reduces its transcriptional activity. Furthermore, SIRT1 inhibits the transcriptional activity of Nkx2.5 and represses the expression of its target genes partly by reducing Nkx2.5 binding to its co-factors, including SRF and TBX5. Taken together, these findings demonstrate that SIRT1 deacetylates Nkx2.5 and inhibits the transcriptional activity of Nkx2.5.


Homeobox Protein Nkx-2.5/genetics , Homeodomain Proteins/genetics , Sirtuin 1/genetics , Transcription, Genetic/genetics , Acetylation , Animals , HEK293 Cells , Humans , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/genetics
7.
Cancer Genet ; 207(9): 403-11, 2014 Sep.
Article En | MEDLINE | ID: mdl-25441685

Novel treatment approaches are desperately needed for malignant rhabdoid tumor (MRT). Telomerase is an attractive therapeutic target because it is specific to cancer and critical for cancer cell immortality. We evaluated the effect of the telomerase inhibitor imetelstat in preclinical models of MRT. Three MRT cell lines, BT-12, G401, and RT-peri, were treated with the telomerase inhibitor imetelstat. The effects of imetelstat on telomere length, DNA damage response, and cell proliferation were assessed. The efficacy of imetelstat in vivo was evaluated in subcutaneous xenografts derived from each of the cell lines. Treatment with imetelstat resulted in inhibition of telomerase activity, marked telomere shortening, and activation of the DNA damage response pathway, as measured by formation of γ-H2AX nuclear foci, phosphorylation of ATM, and phosphorylation of TP53. Imetelstat-treated G401 cells underwent complete growth arrest after 16 passages. The other two cell lines exhibited growth inhibition. Imetelstat resulted in 40-50% growth inhibition compared to placebo-treated controls in all three xenograft models. The activity of imetelstat as a single agent suggests that further studies of telomerase inhibitors in combination with other agents may be warranted.


DNA Repair/drug effects , Indoles/therapeutic use , Niacinamide/analogs & derivatives , Rhabdoid Tumor/drug therapy , Telomerase/antagonists & inhibitors , Telomere Shortening/drug effects , Animals , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chromosomal Proteins, Non-Histone/biosynthesis , Chromosomal Proteins, Non-Histone/genetics , DNA Breaks, Double-Stranded/drug effects , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Female , Histones/biosynthesis , Humans , Mice , Mice, Inbred C57BL , Mice, SCID , Niacinamide/therapeutic use , Oligonucleotides , Phosphorylation , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , SMARCB1 Protein , Telomere Homeostasis/drug effects , Transcription Factors/biosynthesis , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
8.
Sci China Life Sci ; 57(9): 915-24, 2014 Sep.
Article En | MEDLINE | ID: mdl-25104317

SIRT1, a mammalian ortholog of yeast silent information regulator 2 (Sir2), is an NAD(+)-dependent protein deacetylase that plays a critical role in the regulation of vascular function. The current study aims to investigate the functional significance of deacetylase activity of SIRT1 in heart. Here we show that the early postnatal hearts expressed the highest level of SIRT1 deacetylase activity compared to adult and aged hearts. We generated transgenic mice with cardiac-specific expression of a dominant-negative form of the human SIRT1 (SIRT1H363Y), which represses endogenous SIRT1 activity. The transgenic mice displayed dilated atrial and ventricular chambers, and died early in the postnatal period. Pathological, echocardiographic and molecular phenotype confirmed the presence of dilated cardiomyopathy. Terminal deoxynucleotidyl transferase-mediated dUTP nick-end-labeling analysis revealed a greater abundance of apoptotic nuclei in the hearts of transgenic mice. Furthermore, we show that cardiomyocyte apoptosis caused by suppression of SIRT1 activity is, at least in part, due to increased p53 acetylation and upregulated Bax expression. These results indicate that dominant negative form of SIRT1 (SIRT1H363Y) overexpression in mouse hearts causes cardiomyocyte apoptosis and early-onset heart failure, suggesting a critical role of SIRT1 in preserving normal cardiac development during the early postnatal period.


Apoptosis/genetics , Genes, Dominant , Heart Failure/metabolism , Myocardium/metabolism , Myocytes, Cardiac/cytology , Sirtuin 1/genetics , Acetylation , Age of Onset , Animals , Cells, Cultured , Heart Failure/pathology , Mice , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
9.
Basic Res Cardiol ; 108(4): 364, 2013 Jul.
Article En | MEDLINE | ID: mdl-23744058

Nkx2.5 plays protective roles in cardiac homeostasis and survival in the postnatal hearts. However, the underlying molecular mechanisms that mediate the protective functions of Nkx2.5 remain unknown. Here, we showed that Nkx2.5 was downregulated in response to various stresses and was required for protection against the stress-induced apoptosis of cardiomyocytes. SIRT1, a member of the sirtuin family of proteins, was found to be a direct transcriptional target of Nkx2.5 and was required for the Nkx2.5-mediated protection of cardiomyocytes from doxorubicin (DOX)-induced apoptosis. Moreover, using chromatin immunoprecipitation assays, we found that Nkx2.5 was able to bind to the SIRT1 promoter and that this binding was significantly decreased in DOX-treated mouse hearts. Furthermore, the cardiac-specific overexpression of SIRT1 decreased the DOX-induced apoptosis of cardiomyocytes in SIRT1 transgenic mouse hearts compared with the hearts of their wild-type littermates. These findings demonstrate that SIRT1 acts as a direct transcriptional target of Nkx2.5 that maintains cardiomyocyte homeostasis and survival.


Homeodomain Proteins/physiology , Myocytes, Cardiac/physiology , Sirtuin 1/physiology , Stress, Physiological/physiology , Transcription Factors/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Survival/physiology , Cells, Cultured , Doxorubicin/pharmacology , Homeobox Protein Nkx-2.5 , Homeostasis/physiology , Mice , Mice, Transgenic , Models, Animal , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Sirtuin 1/genetics , Up-Regulation/physiology
10.
Sci China Life Sci ; 55(6): 467-73, 2012 Jun.
Article En | MEDLINE | ID: mdl-22744176

The rapidly increasing prevalence of diabetes mellitus worldwide is one of the most serious and challenging health problems in the 21st century. Mammalian sirtuin 1 (SIRT1) has been shown to decrease high-glucose-induced endothelial cell senescence in vitro and prevent hyperglycemia-induced vascular dysfunction. However, a role for SIRT1 in prevention of hyperglycemia-induced vascular cell senescence in vivo remains unclear. We used endothelium-specific SIRT1 transgenic (SIRT1-Tg) mice and wild-type (WT) mice to construct a 40-week streptozotocin (STZ)-induced diabetic mouse model. In this mode, 42.9% of wild-type (WT) mice and 38.5% of SIRT1-Tg mice were successfully established as diabetic. Forty weeks of hyperglycemia induced significant vascular cell senescence in aortas of mice, as indicated by upregulation of expression of senescence-associated markers including p53, p21 and plasminogen activator inhibitor-1 (PAI-1). However, SIRT1-Tg diabetic mice displayed dramatically decreased expression of p53, p21 and PAI-1 compared with diabetic WT mice. Moreover, manganese superoxide dismutase expression (MnSOD) was significantly downregulated in the aortas of diabetic WT mice, but was preserved in diabetic SIRT1-Tg mice. Furthermore, expression of the oxidative stress adaptor p66Shc was significantly decreased in aortas of SIRT1-Tg diabetic mice compared with WT diabetic mice. Overall, these findings suggest that SIRT1-mediated inhibition of hyperglycemia-induced vascular cell senescence is mediated at least partly through the reduction of oxidative stress.


Cellular Senescence , Endothelial Cells/metabolism , Hyperglycemia/metabolism , Sirtuin 1/metabolism , Animals , Blotting, Western , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Down-Regulation , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Hyperglycemia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Plasminogen Activator Inhibitor 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Shc Signaling Adaptor Proteins/metabolism , Sirtuin 1/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1 , Superoxide Dismutase/metabolism , Time Factors , Tumor Suppressor Protein p53/metabolism , Up-Regulation
11.
Circ Res ; 109(6): 639-48, 2011 Sep 02.
Article En | MEDLINE | ID: mdl-21778425

RATIONALE: Inactivation of the p66Shc adaptor protein confers resistance to oxidative stress and protects mice from aging-associated vascular diseases. However, there is limited information about the negative regulating mechanisms of p66Shc expression in the vascular system. OBJECTIVE: In this study, we investigated the role of SIRT1, a class III histone deacetylase, in the regulation of p66Shc expression and hyperglycemia-induced endothelial dysfunction. METHODS AND RESULTS: Expressions of p66Shc gene transcript and protein were significantly increased by different kinds of class III histone deacetylase (sirtuin) inhibitors in human umbilical vein endothelial cells and 293A cells. Adenoviral overexpression of SIRT1 inhibited high-glucose-induced p66Shc upregulation in human umbilical vein endothelial cells. Knockdown of SIRT1 increased p66Shc expression and also increased the expression levels of plasminogen activator inhibitor-1 expression, but decreased manganese superoxide dismutase expression in high-glucose conditions. However, knockdown of p66Shc significantly reversed the effects of SIRT1 knockdown. In addition, p66Shc overexpression significantly decreased manganese superoxide dismutase expression and increased plasminogen activator inhibitor-1 expression in high-glucose conditions, which were recovered by SIRT1 overexpression. Moreover, compared to streptozotocin-induced wild-type diabetic mice, endothelium-specific SIRT1 transgenic diabetic mice had decreased p66Shc expression at both the mRNA and the protein levels, improved endothelial function, and reduced accumulation of nitrotyrosine and 8-OHdG (markers of oxidative stress). We further found that SIRT1 was able to bind to the p66Shc promoter (-508 bp to -250 bp), resulting in a decrease in the acetylation of histone H3 bound to the p66Shc promoter region. CONCLUSION: Our findings indicate that repression of p66Shc expression by SIRT1 contributes to the protection of hyperglycemia-induced endothelial dysfunction.


Down-Regulation/genetics , Endothelium, Vascular/metabolism , Hyperglycemia/genetics , Shc Signaling Adaptor Proteins/antagonists & inhibitors , Sirtuin 1/physiology , Aging/genetics , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Endothelium, Vascular/pathology , HEK293 Cells , Humans , Hyperglycemia/pathology , Hyperglycemia/prevention & control , Immunity, Innate/genetics , Male , Mice , Mice, Transgenic , Oxidative Stress/genetics , Protein Stability , Shc Signaling Adaptor Proteins/biosynthesis , Src Homology 2 Domain-Containing, Transforming Protein 1
12.
J Biol Chem ; 286(25): 22219-26, 2011 Jun 24.
Article En | MEDLINE | ID: mdl-21531726

RGS13, a member of the regulator of G protein signaling (RGS) family, inhibits G protein-coupled receptor signaling in B cells and mast cells (MCs) and suppresses IgE-antigen-induced MC degranulation and anaphylaxis. Although RGS13 expression is induced by immune receptor and chemokine receptor stimulation, the molecular regulation of RGS13 transcription is unknown. Here, we investigated the role of two p53 response elements (REs) in the regulation of RGS13 promoter activity and expression. We found that a 1000-bp DNA fragment upstream of the ATG translation start site (TSS) had promoter activity in reporter gene assays, and deletion or mutation of a p53-binding motif nearest the TSS abolished promoter activity. Notably, p53 bound to both REs in the RGS13 promoter in vivo as assessed by chromatin immunoprecipitation, suggesting that the p53 RE most distal to the TSS is physiologically inactive. We detected reduced RGS13 expression in MCs exogenously expressing p53 or treated with doxorubicin, which induces genotoxic stress and leads to p53 accumulation. RNA silencing of p53 up-regulated RGS13 expression in B lymphocytes, and bone marrow-derived MCs from p53(-/-) mice had increased RGS13 expression. Finally, p53-depleted B cells with increased RGS13 expression had reduced Ca(2+) mobilization in response to sphingosine 1-phosphate. These studies indicate that p53 may modulate immune responses through suppression of RGS13 transcription in MCs and B cells.


B-Lymphocytes/metabolism , Gene Expression Regulation/immunology , Mast Cells/metabolism , RGS Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Animals , B-Lymphocytes/immunology , Base Sequence , Binding Sites , DNA Damage/genetics , DNA Damage/immunology , HEK293 Cells , HeLa Cells , Humans , Mast Cells/immunology , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Transcription, Genetic
13.
J Immunol ; 175(8): 5423-9, 2005 Oct 15.
Article En | MEDLINE | ID: mdl-16210649

The excessive production of reactive oxidative species (ROS) associated with inflammation leads to a condition of oxidative stress. Cyclooxygenase-2 (COX-2), PGE(2), and matrix metalloproteinases (MMPs) are important mediators during the process of inflammation. In this paper we report on studies examining how the ROS hydrogen peroxide (H(2)O(2)) affects the production of MMP-1, COX-2, and PGE(2). Addition of H(2)O(2) to LPS-activated monocytes, but not naive monocytes, caused a significant enhancement of the LPS-induced production of MMP-1, COX-2, and PGE(2). The mechanism by which H(2)O(2) increased these mediators was through enhancement of IkappaBalpha degradation, with subsequent increases in NF-kappaB activation and NF-kappaB p50 translocation to the nucleus. The effects of H(2)O(2) on IkappaBalpha degradation, NF-kappaB activation, and NF-kappaB p50 localization to the nucleus were demonstrated through studies of coimmunoprecipitation of IkappaBalpha with p50, ELISA of NF-kappaB p65 activity, and Western blot analysis of the nuclear fraction extract for p50. The key role for NF-kappaB in this process was demonstrated by the ability of MG-132 or lactacystin (proteasome inhibitors) to block the enhanced production of MMP-1, COX-2, and PGE(2). In contrast, indomethacin, which inhibited PGE(2) production, partially blocked the enhanced MMP-1 production. Moreover, although PGE(2) restored MMP-1 production in indomethacin-treated monocyte cultures; it failed to significantly restore MMP-1 production in proteasome inhibitor-treated cultures. Thus, in the presence of LPS and H(2)O(2), NF-kappaB plays a dominate role in the regulation of MMP-1, COX-2, and PGE(2) expression.


Cyclooxygenase 2/biosynthesis , Dinoprostone/biosynthesis , Lipopolysaccharides/immunology , Matrix Metalloproteinase 1/biosynthesis , Monocytes/enzymology , NF-kappa B/metabolism , Oxidative Stress/physiology , Cells, Cultured , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Hydrogen Peroxide/metabolism , Indomethacin/pharmacology , Macrophage Activation/immunology , Monocytes/drug effects , Monocytes/metabolism , NF-kappa B/antagonists & inhibitors , Proteasome Inhibitors
14.
J Leukoc Biol ; 78(1): 259-65, 2005 Jul.
Article En | MEDLINE | ID: mdl-15800029

Matrix metalloproteinase-9 (MMP-9) is considered to be an important component in the progression of inflammation. Monocytes/macrophages are prominent at inflammation sites, and activation of these cells by stimulants, such as lipopolysaccharide (LPS) or tumor necrosis factor alpha and granulocyte macrophage-colony stimulating factor, leads to the production of significant amounts of MMP-9. Here, we show that LPS stimulation of monocytes results in MMP-9 production through a phosphatidylinositol-3 kinase (PI-3K)/Akt/inhibitor of kappaB (IkappaB) kinase-alpha (IKKalpha)/nuclear factor (NF)-kappaB pathway. This new role for Akt in signaling leading to MMP-9 production was demonstrated by inhibitor and immunoprecipitation studies. LY294002 or wortmannin, inhibitors of PI-3K, suppressed LPS-induced Akt activity and MMP-9 production. Evidence for the participation of Akt in monocyte MMP-9 synthesis was demonstrated by the inhibition of MMP-9 by SH-5, a specific inhibitor of Akt. The mechanism by which Akt regulates MMP-9 is through the activation of NF-kappaB, as shown by coimmunoprecipitation of the phosphorylated form of IKKalpha and Akt as well as the SH-5 suppression of the dissociation of IkappaB from NF-kappaB and the activation of NF-kappaB p65. The role of NF-kappaB in regulation of MMP-9 was demonstrated further by the inhibition of MMP-9 production by proteasome inhibitors, lactacystin and MG-132, which prevented the ubiquitination and dissociation of IkappaB from NF-kappaB. This is the first demonstration that Akt is involved in the signaling pathway leading to the production of monocyte MMP-9 and provides an additional approach in the regulation of this enzyme in human primary monocytes.


Matrix Metalloproteinase 9/metabolism , Monocytes/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Cells, Cultured , Humans , I-kappa B Kinase , Lipopolysaccharides/pharmacology , Monocytes/cytology , Monocytes/drug effects , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Signal Transduction/immunology , Transcription Factor RelA
15.
Proc Natl Acad Sci U S A ; 100(10): 6098-103, 2003 May 13.
Article En | MEDLINE | ID: mdl-12732722

Oxidant-induced injury to the lung is associated with extensive damage to the lung epithelium. Instillation of keratinocyte growth factor (KGF) in the lungs of animals protects animals from oxidant-induced injury but the mechanism of protection is not well understood. An inherent problem in studying KGF function in vivo has been that constitutive overexpression of KGF in the lung causes embryonic lethality with extensive pulmonary malformation. Here we report the development of a stringently regulated, tetracycline-inducible, lung-specific transgenic system that allows regulated expression of KGF in the lung without causing developmental abnormalities from leaky KGF expression. By using this system, we show that exposure of KGF-expressing mice to hyperoxia protects the lung epithelium but not the endothelium from cell death in accordance with the selective expression of KGF receptor on epithelial and not on endothelial cells. Investigations of KGF-induced cell survival pathways revealed KGF-induced activation of the multifunctional pro-survival Akt signaling axis both in vitro and in vivo. Inhibition of KGF-induced Akt activation by a dominant-negative mutant of Akt blocked the KGF-mediated protection of epithelial cells exposed to hyperoxia. KGF-induced Akt activation may play an important role in inhibiting lung alveolar cell death thereby preserving the lung architecture and function during oxidative stress.


Fibroblast Growth Factors/genetics , Hyperoxia/prevention & control , Hyperoxia/physiopathology , Lung/physiopathology , Respiratory Mucosa/physiopathology , Animals , Apoptosis , Cell Death , Cell Line , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/physiology , Humans , In Situ Nick-End Labeling , Mice , Mice, Transgenic , Oxidants/toxicity , Respiratory Mucosa/pathology
16.
J Biol Chem ; 278(12): 10374-80, 2003 Mar 21.
Article En | MEDLINE | ID: mdl-12529371

Keratinocyte growth factor (KGF), a member of the fibroblast growth factor (FGF) family (also known as FGF-7), is an important protective factor for epithelial cells. The receptor for KGF (also called FGFR2-IIIb), which has intrinsic tyrosine kinase activity, is expressed specifically on epithelial cells and in the lung epithelium. Administration of KGF has been shown to protect the lung from various insults, but the mechanism of protection is not well understood. To understand the mechanism by which KGF exerts protective functions on epithelial cells, we used the yeast two-hybrid assay to identify proteins that interact with the KGF receptor (KGFR). Here we show that the cytoplasmic domain of KGFR interacts with p21-activated protein kinase (PAK) 4, which is a new member of the PAK family. The PAKs are regulated by the Rho-family GTPases Rac and Cdc42. PAK4 is the most divergent member of the PAK family of proteins and may have distinct functions. However, stimuli that regulate PAK4 activity are not known. Our data show that PAK4 can associate with the KGFR, which is dependent on KGFR tyrosine kinase activity. We show that a dominant negative mutant of PAK4 blocks KGF-mediated inhibition of caspase-3 activation in epithelial cells subjected to oxidant stress. Our data demonstrate that PAK4 is an important mediator of the anti-apoptotic effects of KGF on epithelial cells.


Adaptor Proteins, Signal Transducing , Apoptosis/drug effects , Fibroblast Growth Factors/physiology , Protein Serine-Threonine Kinases/physiology , Receptors, Fibroblast Growth Factor/physiology , Amino Acid Sequence , Animals , Base Sequence , Caspase 3 , Caspase Inhibitors , Cloning, Molecular , Fibroblast Growth Factor 7 , GRB2 Adaptor Protein , Humans , Hydrogen Peroxide/pharmacology , Mice , Molecular Sequence Data , Oxidants/pharmacology , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/genetics , Proteins/physiology , Receptor, Fibroblast Growth Factor, Type 2 , p21-Activated Kinases
...