Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biomedicines ; 11(2)2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36831104

ABSTRACT

Emerging evidence suggests that Gram-negative bacteria release bacterial outer membrane vesicles (OMVs) and that these play an important role in the pathogenesis of bacterial infection-mediated inflammatory responses and organ damage. Despite the fact that scattered reports have shown that OMVs released from Gram-negative bacteria may function via the TLR2/4-signaling pathway or induce pyroptosis in macrophages, our study reveals a more complex role of OMVs in the development of inflammatory lung responses and macrophage pro-inflammatory activation. We first confirmed that various types of Gram-negative bacteria release similar OMVs which prompt pro-inflammatory activation in both bone marrow-derived macrophages and lung alveolar macrophages. We further demonstrated that mice treated with OMVs via intratracheal instillation developed significant inflammatory lung responses. Using mouse inflammation and autoimmune arrays, we identified multiple altered cytokine/chemokines in both bone marrow-derived macrophages and alveolar macrophages, suggesting that OMVs have a broader spectrum of function compared to LPS. Using TLR4 knock-out cells, we found that OMVs exert more robust effects on activating macrophages compared to LPS. We next examined multiple signaling pathways, including not only cell surface antigens, but also intracellular receptors. Our results confirmed that bacterial OMVs trigger both surface protein-mediated signaling and intracellular signaling pathways, such as the S100-A8 protein-mediated pathway. In summary, our studies confirm that bacterial OMVs strongly induced macrophage pro-inflammatory activation and inflammatory lung responses via multi-signaling pathways. Bacterial OMVs should be viewed as a repertoire of pathogen-associated molecular patterns (PAMPs), exerting more robust effects than Gram-negative bacteria-derived LPS.

2.
Am J Infect Control ; 48(8): 883-889, 2020 08.
Article in English | MEDLINE | ID: mdl-32464294

ABSTRACT

OBJECTIVE: The past 4 months, the emergence and spread of novel 2019 SARS-Cov-2 (COVID-19) has led to a global pandemic which is rapidly depleting supplies of personal protective equipment worldwide. There are currently over 1.6 million confirmed cases of COVID-19 worldwide which has resulted in more the 100,000 deaths. As these numbers grow daily, hospitals are being forced to reuse surgical masks in hopes of conserving their dwindling supply. Since COVID-19 will most likely have effects that last for many months, our nationwide shortage of masks poses a long term issue that must be addressed immediately. METHODS: Based on a previous study by Quan et al., a salt-based soaking strategy has been reported to enhance the filtration ability of surgical masks. We propose a similar soaking process which uses materials widely available in anyone's household. We tested this method of pretreating a variety of materials with a salt-based solution by a droplet test using fluorescently stained nanoparticles similar in size to the COVID-19 virus. RESULTS: In this study, we found that paper towels and surgical masks pretreated with the salt-based solution showed a noticeable increase in filtration of nanoparticles similar in size to the COVID-19 virus. We also show that the TWEEN20 used by Quan et al. is not a critical component for the solution, and using salt alone in solution still provides a dramatically increased level of protection. CONCLUSIONS: We believe this method will allow for healthcare workers to create a disposable added layer of protection to their surgical masks, N95s, or homemade masks by using household available products. Adoption of this method may play an essential role in ensuring the safety of healthcare workers during the COVID-19 pandemic and any pandemics that may arise in the future.


Subject(s)
Coronavirus Infections/prevention & control , Filtration/methods , Infectious Disease Transmission, Patient-to-Professional/prevention & control , Masks/virology , Pandemics/prevention & control , Personal Protective Equipment/microbiology , Pneumonia, Viral/prevention & control , Betacoronavirus/pathogenicity , COVID-19 , Health Personnel , Humans , SARS-CoV-2 , Sodium Chloride/chemistry
3.
Int J Mol Sci ; 21(10)2020 May 19.
Article in English | MEDLINE | ID: mdl-32438692

ABSTRACT

Pancreatic cancer is the worst exocrine gastrointestinal cancer leading to the highest mortality. Recent studies reported that aberrant expression of apurinic/apyrimidinic endodeoxyribonuclease 1 (APE1) is involved in uncontrolled cell growth. However, the molecular mechanism of APE1 biological role remains unrevealed in pancreatic cancer progression. Here, we demonstrate that APE1 accelerates pancreatic cancer cell proliferation through glial cell line-derived neurotrophic factor (GDNF)/glial factor receptor α1 (GFRα1)/Src/ERK axis-cascade signaling. The proliferation of endogenous APE1 expressed-MIA PaCa-2, a human pancreatic carcinoma cell line, was increased by treatment with GDNF, a ligand of GFRα1. Either of downregulated APE1 or GFRα1 expression using small interference RNA (siRNA) inhibited GDNF-induced cancer cell proliferation. The MEK-1 inhibitor PD98059 decreased GDNF-induced MIA PaCa-2 cell proliferation. Src inactivation by either its siRNA or Src inhibitor decreased ERK-phosphorylation in response to GDNF in MIA PaCa-2 cells. Overexpression of GFRα1 in APE1-deficient MIA PaCa-2 cells activated the phosphorylation of Src and ERK. The expression of both APE1 and GFRα1 was gradually increased as progressing pancreatic cancer grades. Our results highlight a critical role for APE1 in GDNF-induced pancreatic cancer cell proliferation through APE1/GFRα1/Src/ERK axis-cascade signaling and provide evidence for future potential therapeutic drug targets for the treatment of pancreatic cancer.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , MAP Kinase Signaling System , Pancreatic Neoplasms/pathology , src-Family Kinases/metabolism , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Female , Humans , MAP Kinase Signaling System/drug effects , Male , Middle Aged , Models, Biological , Neoplasm Invasiveness , Phosphorylation/drug effects , Pancreatic Neoplasms
4.
Chonnam Med J ; 56(1): 1-5, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32021835

ABSTRACT

Scavenger receptors typically bind to multiple ligands on a cell surface, including endogenous and modified host-derived molecules and microbial pathogens. They promote the elimination of degraded or harmful substances such as non-self or altered-self targets through endocytosis, phagocytosis, and adhesion. Currently, scavenger receptors are subdivided into eight classes based on several variations in their sequences due to alternative splicing. Since recent studies indicate targeting scavenger receptors has been involved in cancer prognosis and carcinogenesis, we will focus on the current knowledge about the emerging role of scavenger receptor classes A to E in cancer progression.

5.
Chonnam Med J ; 55(2): 75-85, 2019 May.
Article in English | MEDLINE | ID: mdl-31161119

ABSTRACT

Psoriasis is a chronic, recurrent, heterogeneous, cutaneous inflammatory skin disease for which there is no cure. It affects approximately 7.5 million people in the United States. Currently, several biologic agents that target different molecules implicated in the pathogenic processes of psoriasis are being assessed in diverse clinical studies. However, relapse usually occurs within weeks or months, meaning there is currently no cure for psoriasis. Therefore, recent studies have discovered diverse new potential treatments for psoriasis: inhibitors of bacteria such as Staphylococcus aureus, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and neuropilin 1 (NRP1). A promising approach that has recently been described involves modifying antimicrobial peptides to develop new cutaneous anti-bacterial agents that target inflammatory skin disease induced by Staphylococcus. Increased expression of TRAIL and its death receptors DR4 and DR5 has been implicated in the pathogenesis of plaque psoriasis. In addition, TRAIL has the ability to inhibit angiogenesis by inducing endothelial cell death and by negative regulation of VEGF-induced angiogenesis via caspase-8-mediated enzymatic and non-enzymatic functions. Since NRP1 regulates angiogenesis induced by multiple signals, including VEGF, ECM and semaphorins, and also initiates proliferation of keratinocytes through NF-κB signaling pathway in involved psoriatic skin, targeting NRP1 pathways may offer numerous windows for intervention in psoriasis. In this review, we will focus on the current knowledge about the emerging role of synthetic antimicrobial peptides, TRAIL and NRP1 blocking peptides in the pathogenesis and treatment of psoriasis.

6.
Chonnam Med J ; 54(3): 135-142, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30288368

ABSTRACT

Over recent years, several new molecular and immunogenic therapeutic approaches to melanoma treatment have been approved and implemented in clinical practice. Mechanisms of resistance to these new therapies have become a major problem. Mutation-specific pharmacotherapy can result in simultaneous emergence of resistant clones at many separate body sites despite an initially positive therapeutic response. Additionally, treatments aimed at inducing apoptosis are subject to resistance due to escape through other known mechanisms of regulated cell death (RCD). In this review, we discuss the complexity in pharmacological manipulation of melanoma with c-Kit, BRAF, MEK, and/or mTOR mutant cell lines. This study also addresses melanoma evasion of cell death through modalities of RCD such as apoptosis, autophagy, and necroptosis. This study also examines new combination therapies which have been approved to target both cell cycle dysregulation and cell death pathways. Lastly, we recognize the importance of immunomodulation though manipulation of the body's natural killing mechanisms with CTLA4, PD1, and CSF1 inhibition. As we begin to recognize tumor cell activation of alternate pathways, evasion of programmed cell death, and manipulation of the tumor microenvironment, it is increasingly important to grasp the complexity of personalized therapy in melanoma treatment.

7.
BMC Cancer ; 18(1): 432, 2018 04 16.
Article in English | MEDLINE | ID: mdl-29661248

ABSTRACT

BACKGROUND: Multiple trials have attempted to demonstrate the effective induction of cell death in TRAIL-resistant cancer cells, including using a combined treatment of recombinant TRAIL and various proteasome inhibitors. These studies have yielded limited success, as the mechanism of cell death is currently unidentified. Understanding this mechanism's driving forces may facilitate the induction of cell death in TRAIL-resistant cancer cells. METHODS: Three kinds of recombinant soluble TRAIL proteins were treated into TRAIL-resistant cells and TRAIL-susceptible cells, with or without bortezomib, to compare their respective abilities to induce cell death. Recombinant TRAIL was treated with bortezomib to investigate whether this combination treatment could induce tumor regression in a mouse syngeneic tumor model. To understand the mechanism of combined treatment-induced cell death, cells were analyzed by flow cytometry and the effects of various cell death inhibitors on cell death rates were examined. RESULTS: ILz:rhTRAIL, a recombinant human TRAIL containing isoleucine zipper hexamerization domain, showed the highest cell death inducing ability both in single treatment and in combination treatment with bortezomib. In both TRAIL-resistant and TRAIL-susceptible cells treated with the combination treatment, an increase in cell death rates was dependent upon both the dose of TRAIL and its intrinsic properties. When a syngeneic mouse tumor model was treated with the combination of ILz:rhTRAIL and bortezomib, significant tumor regression was seen as a result of the effective induction of cancer cell death. The combination treatment-induced cell death was both inhibited by TRAIL blocking antibody and caspase-dependent. However, it was not inhibited by various ER stress inhibitors and autophagy inhibitors. CONCLUSIONS: The combination treatment with ILz:rhTRAIL and bortezomib was able to induce cell death in both TRAIL-susceptible and TRAIL-resistant cancer cells through the intracellular TRAIL signaling pathway. The efficiency of cell death was dependent on the properties of TRAIL under the environment provided by bortezomib. The combination treatment-induced cell death was not regulated by bortezomib-induced ER stress response or by autophagy.


Subject(s)
Bortezomib/administration & dosage , Cell Proliferation/drug effects , TNF-Related Apoptosis-Inducing Ligand/genetics , Animals , Apoptosis/drug effects , Caspases/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Humans , Mice , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
8.
Chonnam Med J ; 53(3): 173-177, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29026704

ABSTRACT

Melanoma is one of the most aggressive cancers in the world and is responsible for the majority of skin cancer deaths. Recent advances in the field of immunotherapy using active, adoptive, and antigen-specific therapeutic approaches, have generated the expectation that these technologies have the potential to improve the treatment of advanced malignancies, including melanoma. Treatment options for metastatic melanoma patients have been dramatically improved by the FDA approval of new therapeutic agents including vemurafenib, dabrafenib, and sorafenib. These kinase inhibitors have the potential to work in tandem with MEK, PI3K/AKT, and mTOR to inhibit the activity of melanoma inducing BRAF mutations. This review summarizes the effects of the new therapeutic agents against melanoma and the underlying biology of these BRAF inhibitors.

9.
PLoS One ; 10(8): e0136887, 2015.
Article in English | MEDLINE | ID: mdl-26309029

ABSTRACT

Alpha-melanocyte stimulating hormone (α-MSH) is a highly conserved 13-aa neuropeptide derived from pro-opiomelanocortin by post-translational processing, which has been reported to exhibit potent anti-inflammatory activity and a wide range of immunosuppressive activities in the skin. However, the regulatory effect of α-MSH is not completely clear in cutaneous innate immunity. In this study, we investigate the functional regulation of α-MSH in TLR2-mediated inflammatory responses in normal human keratinocytes (HKs). α-MSH pretreatment down-regulated the Staphylococcus aureus LTA-induced expression of both TLR2 and IL-8 as well as NF-κB nuclear translocation in HK cells. The inhibitory effect of α-MSH was blocked by agouti signaling protein (ASP), an α-MSH receptor-1 antagonist. To investigate the mechanism of this response in more detail, siRNA of IRAK-M, a negative regulator of TLR signaling, was utilized in these studies. The α-MSH suppressive effect on IL-8 production and NF-κB transactivation was inhibited by IRAK-M siRNA transfection in HK cells. These results indicate that α-MSH is capable of suppressing keratinocyte TLR2-mediated inflammatory responses induced by S. aureus-LTA, thus demonstrating another novel immunomodulatory activity of α-MSH in normal human keratinocytes.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Toll-Like Receptor 2/metabolism , alpha-MSH/pharmacology , Active Transport, Cell Nucleus/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Silencing , Humans , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-8/biosynthesis , Interleukin-8/genetics , Keratinocytes/cytology , Keratinocytes/microbiology , Lipopolysaccharides/biosynthesis , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Staphylococcus aureus/metabolism , Teichoic Acids/biosynthesis , Teichoic Acids/pharmacology , Toll-Like Receptor 2/genetics
10.
PLoS One ; 10(7): e0132619, 2015.
Article in English | MEDLINE | ID: mdl-26197393

ABSTRACT

The cutaneous inflammation associated with acne vulgaris is caused by the anaerobic bacterium Propionibacterium acnes through activation of the innate immune system in the skin. Current standard treatments for acne have limitations that include adverse effects and poor efficacy in many patients, making development of a more effective therapy highly desirable. In the present study, we demonstrate the protective effects of a novel customized α-helical cationic peptide, P5, against P. acnes-induced inflammatory responses in vitro and in vivo. Application of P5 significantly reduced expression of two inflammatory cytokines IL-8 and TNF-α in P. acnes-treated primary human keratinocytes, where P5 appeared to act in part by binding to bacterial lipoteichoic acid, thereby suppressing TLR2-to-NF-κB signaling. In addition, in a mouse model of acne vulgaris, P5 exerted both anti-inflammatory and antimicrobial effects against P. acnes, but exerted no cytotoxic effects against skin cells. These results demonstrate that P5, and perhaps other cationic antimicrobial peptides, offer the unique ability to reduce numbers P. acnes cells in the skin and to inhibit the inflammation they trigger. This suggests these peptides could potentially be used to effectively treat acne without adversely affecting the skin.


Subject(s)
Acne Vulgaris/drug therapy , Anti-Inflammatory Agents/administration & dosage , Antimicrobial Cationic Peptides/administration & dosage , Gram-Positive Bacterial Infections/drug therapy , Keratinocytes/drug effects , Lipopolysaccharides/metabolism , Teichoic Acids/metabolism , Acne Vulgaris/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Gram-Positive Bacterial Infections/immunology , Humans , Interleukin-8/metabolism , Keratinocytes/cytology , Keratinocytes/immunology , Mice , Propionibacterium acnes/drug effects , Propionibacterium acnes/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
11.
Int J Mol Sci ; 15(9): 16800-15, 2014 Sep 22.
Article in English | MEDLINE | ID: mdl-25247578

ABSTRACT

Recently, various immunosuppressant drugs have been shown to induce hair growth in normal hair as well as in alopecia areata and androgenic alopecia; however, the responsible mechanism has not yet been fully elucidated. In this study, we investigate the influence of mycophenolate (MPA), an immunosuppressant, on the proliferation of human dermal papilla cells (hDPCs) and on the growth of human hair follicles following catagen induction with interferon (IFN)-γ. IFN-γ was found to reduce ß-catenin, an activator of hair follicle growth, and activate glycogen synthase kinase (GSK)-3ß, and enhance expression of the Wnt inhibitor DKK-1 and catagen inducer transforming growth factor (TGF)-ß2. IFN-γ inhibited expression of ALP and other dermal papillar cells (DPCs) markers such as Axin2, IGF-1, and FGF 7 and 10. MPA increased ß-catenin in IFN-γ-treated hDPCs leading to its nuclear accumulation via inhibition of GSK3ß and reduction of DKK-1. Furthermore, MPA significantly increased expression of ALP and other DPC marker genes but inhibited expression of TGF-ß2. Therefore, we demonstrate for the first time that IFN-γ induces catagen-like changes in hDPCs and in hair follicles via inhibition of Wnt/ß-catenin signaling, and that MPA stabilizes ß-catenin by inhibiting GSK3ß leading to increased ß-catenin target gene and DP signature gene expression, which may, in part, counteract IFN-γ-induced catagen in hDPCs.


Subject(s)
Dermis/drug effects , Hair Follicle/drug effects , Immunosuppressive Agents/pharmacology , Mycophenolic Acid/analogs & derivatives , Wnt Signaling Pathway/drug effects , beta Catenin/physiology , Alopecia/drug therapy , Cell Division/drug effects , Cells, Cultured , Dermis/cytology , Dermis/metabolism , Drug Evaluation, Preclinical , Gene Expression Profiling , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Hair Follicle/growth & development , Hair Follicle/metabolism , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interferon-gamma/pharmacology , Mycophenolic Acid/pharmacology , Transforming Growth Factor beta2/biosynthesis , Transforming Growth Factor beta2/genetics
12.
Int J Mol Sci ; 15(5): 8753-72, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24840573

ABSTRACT

Staphylococcus aureus (S. aureus) is a widespread cutaneous pathogen responsible for the great majority of bacterial skin infections in humans. The incidence of skin infections by S. aureus reflects in part the competition between host cutaneous immune defenses and S. aureus virulence factors. As part of the innate immune system in the skin, cationic antimicrobial peptides (CAMPs) such as the ß-defensins and cathelicidin contribute to host cutaneous defense, which prevents harmful microorganisms, like S. aureus, from crossing epithelial barriers. Conversely, S. aureus utilizes evasive mechanisms against host defenses to promote its colonization and infection of the skin. In this review, we focus on host-pathogen interactions during colonization and infection of the skin by S. aureus and methicillin-resistant Staphylococcus aureus (MRSA). We will discuss the peptides (defensins, cathelicidins, RNase7, dermcidin) and other mediators (toll-like receptor, IL-1 and IL-17) that comprise the host defense against S. aureus skin infection, as well as the various mechanisms by which S. aureus evades host defenses. It is anticipated that greater understanding of these mechanisms will enable development of more sustainable antimicrobial compounds and new therapeutic approaches to the treatment of S. aureus skin infection and colonization.


Subject(s)
Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Skin/metabolism , Staphylococcus aureus/drug effects , Host-Pathogen Interactions , Humans , Immune System/microbiology , Interleukins/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Skin/immunology , Skin/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Toll-Like Receptors/metabolism
13.
J Invest Dermatol ; 131(8): 1677-83, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21593770

ABSTRACT

The lipophilic fungus Malassezia furfur (M. furfur) is a commensal microbe associated with several chronic diseases such as pityriasis versicolor, folliculitis, and seborrheic dermatitis. Because M. furfur-related diseases are difficult to treat and require prolonged use of medications, the treatment for M. furfur-related skin diseases is supposed to gain control over M. furfur growth and the inflammation associated with it, as well as to prevent secondary infections. In this study, we investigated the antifungal and anti-inflammatory effects of cecropin A(1-8)-magainin 2(1-12) hybrid peptide analog P5 on M. furfur. The minimal inhibitory concentration of P5 against M. furfur was 0.39 µM, making it 3-4 times more potent than commonly used antifungal agents such as ketoconazole (1.5 µM) or itraconazole (1.14 µM). P5 efficiently inhibited the expression of IL-8 and Toll-like receptor 2 in M. furfur-infected human keratinocytes without eukaryotic cytotoxicity at its fungicidal concentration. Moreover, P5 significantly downregulated NF-κB activation and intracellular calcium fluctuation, which are closely related with enhanced responses of keratinocyte inflammation induced by M. furfur infection. Taken together, these observations suggest P5 may be a potential therapeutic agent for M. furfur-associated human skin diseases because of its distinct antifungal and anti-inflammatory action.


Subject(s)
Antifungal Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Dermatomycoses/drug therapy , Keratinocytes/microbiology , Malassezia/drug effects , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/immunology , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cells, Cultured , Dermatomycoses/immunology , Dose-Response Relationship, Drug , Humans , Interleukin-8/metabolism , Keratinocytes/cytology , Keratinocytes/immunology , Malassezia/immunology , Microbial Sensitivity Tests , NF-kappa B/metabolism
14.
Cancer Sci ; 101(9): 1990-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20550525

ABSTRACT

Oncogenic Ras proteins transform cells by way of multiple downstream signaling pathways that promote the genesis of human cancers. However, the exact cellular mechanisms by which downstream targets are regulated are not fully understood. Here, we show that oncogenic Ras reduced Clast1/LR8 transcript levels in mouse NIH3T3 fibroblasts and human WI38 fibroblasts. Clast1/LR8 transcript was undetectable in H460, A549, and H1299 cells showing high Ras activity, but was relatively abundant in DMS53 cells displaying low Ras activity. We also showed that K-Ras siRNA restored Clast1/LR8 expression in H460 and A549 cells, and that inhibitors of DNA methylation and histone deacetylation reversed oncogenic H-Ras-mediated suppression of Clast1/LR8 transcription. Additionally, ectopic expression of Clast1/LR8 inhibited serum-stimulated phosphorylation of ERK1/2 and Akt in H-RasV12-transformed NIH3T3 cells. We further showed that the expression of Clast1/LR8 interfered with oncogenic Ras-induced NIH3T3 cell transformation and invasion. Finally, our results showed that Clast1/LR8 inhibited Ras-induced proliferation of, and tumor formation by, oncogenic H-RasV12-transformed NIH3T3 cells in vivo. This study identifies the downregulation of Clast1/LR8 as a potentially important mechanism by which oncogenic Ras-mediated neoplastic transformation occurs.


Subject(s)
Cell Transformation, Neoplastic/genetics , Down-Regulation , Neoplasms, Experimental/genetics , ras Proteins/genetics , Acetylation , Animals , Base Sequence , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA Methylation , Female , Histones/metabolism , Humans , Mice , Mice, Nude , Molecular Sequence Data , NIH 3T3 Cells , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transfection , ras Proteins/metabolism
15.
J Cardiovasc Pharmacol ; 55(4): 417-24, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20422739

ABSTRACT

BACKGROUND: Curcumin, a natural polyphenolic compound, has been shown to reduce cardiomyocyte growth. Angiotensin II type 1 receptor (AT1R) and lectin-like oxidized low density lipoprotein (ox-LDL) receptor-1 (LOX-1) are major stimuli for cardiomyocyte growth via activation of oxidant signals. We postulated that curcumin may reduce Ang II-mediated cardiomyocyte growth via AT1R and LOX-1 inhibition. METHODS: Adult mouse cardiomyocytes (HL-1) were incubated overnight in serum-free medium, and then treated with solvents or curcumin, the AT1R inhibitor losartan or anti-LOX-1 antibody for 3 hours, and the cells were then stimulated with Ang II. We measured cardiomyocyte growth, and associated intracellular redox signals using reverse transcriptase-polymerase chain reaction and quantitative real-time RT-PCR. We also examined the effect of curcumin on cardiomyocyte biology with forced overexpression of LOX-1 gene. RESULTS: Curcumin (5-10 microM), losartan, and anti-LOX-1 antibody markedly attenuated Ang II-mediated oxidant stress, and the expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and nuclear factor-kappaB (NF-kappaB). Attenuation of redox state by curcumin resulted in abrogation of Ang II-mediated cardiomyocyte growth and atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) genes. Curcumin also reduced Ang II-mediated upregulation of AT1R and LOX-1. The forced upregulation of LOX-1 enhanced the expression of genes for AT1R, ANP, and BNP, and curcumin pretreatment reduced LOX-1 and AT1R expression and LOX-1-mediated increase in hypertrophy markers. CONCLUSIONS: Curcumin attenuates Ang II-mediated cardiomyocyte growth by inhibiting LOX-1 and AT1R expression and suppressing the heightened intracellular redox state.


Subject(s)
Angiotensin II/pharmacology , Cell Enlargement/drug effects , Cell Proliferation/drug effects , Curcumin/pharmacology , Myocytes, Cardiac/cytology , Scavenger Receptors, Class E/genetics , Angiotensin II/administration & dosage , Animals , Antibodies/immunology , Antibodies/pharmacology , Atrial Natriuretic Factor/genetics , Cell Line, Transformed , Cell Survival/drug effects , Curcumin/administration & dosage , Gene Expression/drug effects , Gene Expression/genetics , Losartan/pharmacology , Membrane Glycoproteins/genetics , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NADPH Oxidase 2 , NADPH Oxidases/genetics , NF-kappa B/genetics , Natriuretic Peptide, Brain/genetics , Oxidative Stress/drug effects , Protein Subunits/genetics , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/genetics , Scavenger Receptors, Class E/immunology , Signal Transduction/genetics , Transfection
16.
Physiol Genomics ; 42(1): 42-54, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20332185

ABSTRACT

We studied the gene expression profile during cardiac hypertrophy induced by angiotensin (ANG) II in wild-type mice and the influence of LOX-1 deletion on the gene expression profile. Wild-type and LOX-1 knockout mice were given saline or ANG II infusion for 4 wk. The saline-treated LOX-1 knockout mice showed upregulation of several genes including Ddx3y and Eif2s3y. ANG II infusion enhanced expression of genes known to be associated with cardiac remodeling, such as Agt, Ace, Timp4, Fstl, and Tnfrst12a, as well as oxidant stress-related genes Gnaq, Sos1, and Rac1. Some other strongly upregulated genes identified in this study have not been previously associated with LOX-1 deletion and/or hypertension. To confirm these observations with ANG II infusion and LOX-1 deletion, cultured HL-1 mouse cardiomyocytes were exposed to ANG II or transfected with pCI-neo/LOX-1, which resulted in severalfold increase in reactive oxygen species generation, upregulation of ANG II type 1 (AT(1)) receptor, and cardiomyocyte growth. Quantitative PCR analysis of these treated cardiomyocytes confirmed upregulation of many of the genes identified in the in vivo study. This study provides the first set of data on the gene expression profiling of cardiac tissue treated with ANG II and expands on the important role of LOX-1 in cardiac response to ANG II.


Subject(s)
Angiotensin II/pharmacology , Genomics , Heart/drug effects , Myocardium/metabolism , Scavenger Receptors, Class E/deficiency , Animals , Blood Pressure/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Size/drug effects , Gene Expression Profiling , Heart/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oligonucleotide Array Sequence Analysis , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Scavenger Receptors, Class E/genetics , Vasoconstrictor Agents/pharmacology
17.
J Cardiovasc Pharmacol ; 55(2): 176-83, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19935077

ABSTRACT

BACKGROUND: Curcumin, a natural polyphenolic compound, has been shown to reduce cardiomyocyte growth. Angiotensin II type 1 receptor (AT1R) and lectin-like oxidized low density lipoprotein (ox-LDL) receptor-1 (LOX-1) are major stimuli for cardiomyocyte growth via activation of oxidant signals. We postulated that curcumin may reduce Ang II-mediated cardiomyocyte growth via AT1R and LOX-1 inhibition. METHODS: Adult mouse cardiomyocytes (HL-1) were incubated overnight in serum-free medium, and then treated with solvents or curcumin, the AT1R inhibitor losartan or anti-LOX-1 antibody for 3 hours, and the cells were then stimulated with Ang II. We measured cardiomyocyte growth, and associated intracellular redox signals using reverse transcriptase-polymerase chain reaction and quantitative real-time RT-PCR. We also examined the effect of curcumin on cardiomyocyte biology with forced overexpression of LOX-1 gene. RESULTS: Curcumin (5-10 microM), losartan, and anti-LOX-1 antibody markedly attenuated Ang II-mediated oxidant stress, and the expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and nuclear factor-kappaB (NF-kappaB). Attenuation of redox state by curcumin resulted in abrogation of Ang II-mediated cardiomyocyte growth and atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) genes. Curcumin also reduced Ang II-mediated upregulation of AT1R and LOX-1. The forced upregulation of LOX-1 enhanced the expression of genes for AT1R, ANP, and BNP, and curcumin pretreatment reduced LOX-1 and AT1R expression and LOX-1-mediated increase in hypertrophy markers. CONCLUSIONS: Curcumin attenuates Ang II-mediated cardiomyocyte growth by inhibiting LOX-1 and AT1R expression and suppressing the heightened intracellular redox state.


Subject(s)
Angiotensin II/physiology , Cell Enlargement/drug effects , Curcumin/pharmacology , Growth Inhibitors/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Scavenger Receptors, Class E/antagonists & inhibitors , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cells, Cultured , Mice , Myocytes, Cardiac/cytology , Receptor, Angiotensin, Type 1/physiology , Scavenger Receptors, Class E/biosynthesis
18.
J Exp Bot ; 58(10): 2471-8, 2007.
Article in English | MEDLINE | ID: mdl-17545222

ABSTRACT

The intensity of flower colour, mainly determined by the amount of anthocyanin, is an important horticultural trait. To modulate flower colour intensity, post-transcriptional gene silencing (PTGS)-based technology has been widely used. The constraint of PTGS, however, is that it requires a high degree of conservation in the nucleotide sequences of the target and the silencer. Further, it is difficult to restrict PTGS to the desired tissue or organ due to its systemic spread. To overcome these problems, dominant-negative chalcone synthase (CHS) enzymes have been developed by mutating a cysteine that is essential for the catalytic activity and a methionine that protrudes into the adjoining CHS monomer, as shown through crystallography. The dominant-negative action of mutated CHS enzymes from Mazus japonicus are demonstrated using transgenic Arabidopsis. Also, the modulation of Petunia flower colour intensity by the dominant-negative CHS is shown. The data support the crystallography result showing the importance of the protruding methionine for the function of the adjoining CHS monomer. Furthermore, the modulation of anthocyanin production by the mutated Mazus CHS in Arabidopsis and petunia suggests that the dominant-negative CHS can be used even in distantly related species.


Subject(s)
Acyltransferases/genetics , Arabidopsis/genetics , Color , Flowers/genetics , Plant Proteins/genetics , Acyltransferases/chemistry , Acyltransferases/physiology , Amino Acid Sequence , Arabidopsis/anatomy & histology , Arabidopsis/classification , Cloning, Molecular , Flowers/anatomy & histology , Genes, Dominant , Magnoliopsida/classification , Magnoliopsida/enzymology , Magnoliopsida/genetics , Methionine , Molecular Sequence Data , Mutagenesis, Site-Directed , Phylogeny , Plant Proteins/chemistry , Plant Proteins/physiology , Plants, Genetically Modified/anatomy & histology , Plants, Genetically Modified/metabolism , Protein Engineering , Sequence Alignment
19.
J Korean Med Sci ; 22(2): 305-10, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17449941

ABSTRACT

Claudin-7 has recently been suggested to be a distal nephron marker. We tested the possibility that expression of claudin-7 could be used as a marker of renal tumors originating from the distal nephron. We examined the immunohistochemical expression of claudin-7 and parvalbumin in 239 renal tumors, including 179 clear cell renal cell carcinoma (RCC)s, 29 papillary RCCs, 20 chromophobe RCCs, and 11 renal oncocytomas. In addition, the methylation specific-PCR (MSP) of claudin-7 was performed. Claudin-7 and parvalbumin immunostains were positive in 3.4%, 7.8% of clear cell RCCs, 34.5%, 31.0% of papillary RCCs, 95.0%, 80.0% of chromophobe RCCs, and 72.7%, 81.8% of renal oncocytomas, respectively. The sensitivity and specificity of claudin-7 in diagnosing chromophobe RCC among subtypes of RCC were 95.0% and 92.3%. Those of parvalbumin were 80.0% and 88.9%. The expression pattern of claudin-7 was mostly diffuse in chromophobe RCC and was either focal or diffuse in oncocytoma. All of the cases examined in the MSP revealed the presence of unmethylated promoter of claudin-7 without regard to claudin-7 immunoreactivity. Hypermethylation of the promoter might not be the underlying mechanism for loss of its expression in RCC. Claudin-7 can be used as a useful diagnostic marker in diagnosing chromophobe RCC and oncocytoma.


Subject(s)
Adenoma, Oxyphilic/diagnosis , Adenoma, Oxyphilic/metabolism , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/diagnosis , Kidney Neoplasms/metabolism , Membrane Proteins/metabolism , Biomarkers, Tumor/metabolism , Claudins , Humans , Membrane Proteins/analysis , Neoplasm Proteins/metabolism , Nephrons/metabolism , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...