Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(39): e2302878120, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37722058

ABSTRACT

Although tumor-intrinsic fatty acid ß-oxidation (FAO) is implicated in multiple aspects of tumorigenesis and progression, the impact of this metabolic pathway on cancer cell susceptibility to immunotherapy remains unknown. Here, we report that cytotoxicity of killer T cells induces activation of FAO and upregulation of carnitine palmitoyltransferase 1A (CPT1A), the rate-limiting enzyme of FAO in cancer cells. The repression of CPT1A activity or expression renders cancer cells more susceptible to destruction by cytotoxic T lymphocytes. Our mechanistic studies reveal that FAO deficiency abrogates the prosurvival signaling in cancer cells under immune cytolytic stress. Furthermore, we identify T cell-derived IFN-γ as a major factor responsible for induction of CPT1A and FAO in an AMPK-dependent manner, indicating a dynamic interplay between immune effector cells and tumor targets. While cancer growth in the absence of CPT1A remains largely unaffected, established tumors upon FAO inhibition become significantly more responsive to cellular immunotherapies including chimeric antigen receptor-engineered human T cells. Together, these findings uncover a mode of cancer resistance and immune editing that can facilitate immune escape and limit the benefits of immunotherapies.


Subject(s)
Carnitine O-Palmitoyltransferase , Neoplasms , Humans , Carnitine O-Palmitoyltransferase/genetics , Cytotoxicity, Immunologic , Fatty Acids , Lipid Metabolism , Neoplasms/therapy , T-Lymphocytes, Cytotoxic
2.
J Biol Chem ; 299(7): 104915, 2023 07.
Article in English | MEDLINE | ID: mdl-37315790

ABSTRACT

Biological functions of the highly conserved ubiquitin-like protein 5 (UBL5) are not well understood. In Caenorhabditis elegans, UBL5 is induced under mitochondrial stress to mount the mitochondrial unfolded protein response (UPR). However, the role of UBL5 in the more prevalent endoplasmic reticulum (ER) stress-UPR in the mammalian system is unknown. In the present work, we demonstrated that UBL5 was an ER stress-responsive protein, undergoing rapid depletion in mammalian cells and livers of mice. The ER stress-induced UBL5 depletion was mediated by proteasome-dependent yet ubiquitin-independent proteolysis. Activation of the protein kinase R-like ER kinase arm of the UPR was essential and sufficient for inducing UBL5 degradation. RNA-Seq analysis of UBL5-regulated transcriptome revealed that multiple death pathways were activated in UBL5-silenced cells. In agreement with this, UBL5 knockdown induced severe apoptosis in culture and suppressed tumorigenicity of cancer cells in vivo. Furthermore, overexpression of UBL5 protected specifically against ER stress-induced apoptosis. These results identify UBL5 as a physiologically relevant survival regulator that is proteolytically depleted by the UPR-protein kinase R-like ER kinase pathway, linking ER stress to cell death.


Subject(s)
Cell Death , Endoplasmic Reticulum Stress , Ubiquitins , eIF-2 Kinase , Animals , Mice , Apoptosis , eIF-2 Kinase/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism , Unfolded Protein Response
4.
Methods Mol Biol ; 2455: 49-62, 2022.
Article in English | MEDLINE | ID: mdl-35212985

ABSTRACT

Fatty acid beta oxidation (FAO) is a predominant bioenergetic pathway in mammals. Substantial investigations have demonstrated that FAO activity is dysregulated in many pathophysiological conditions including nonalcoholic steatohepatitis (NASH). Convenient and quantitative assays of FAO activities are important for studies of cell metabolism and the biological relevance of FAO to health and diseases. However, most current FAO assays are based on non-physiological culture conditions, measure FAO activity indirectly or lack adequate quantification. We herein describe details of practical protocols for measurement of basal and genetically or pharmacologically regulated FAO activities in the mammalian system. We also discuss the advantages and disadvantages of these assays in the context of experimental purposes.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Energy Metabolism , Lipolysis , Liver/metabolism , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/metabolism
5.
FASEB J ; 34(10): 13935-13948, 2020 10.
Article in English | MEDLINE | ID: mdl-32851734

ABSTRACT

Epithelial ovarian carcinoma tissues express high levels of tumor necrosis factor-alpha (TNF-α) and other inflammatory cytokines. The underlying mechanism leading to the abnormal TNF-α expression in ovarian cancer remains poorly understood. In the current study, we demonstrated that lysophosphatidic acid (LPA), a lipid mediator present in ascites of ovarian cancer patients, induced expression of TNF-α mRNA and release of TNF-α protein in ovarian cancer cells. LPA also induced expression of interleukin-1ß (IL-1ß) mRNA but no significant increase in IL-1ß protein was detected. LPA enhanced TNF-α mRNA through NF-κB-mediated transcriptional activation. Inactivation of ADAM17, a disintegrin and metalloproteinase, with a specific inhibitor TMI-1 or by shRNA knockdown prevented ovarian cancer cells from releasing TNF-α protein in response to LPA, indicating that LPA-mediated TNF-α production relies on both transcriptional upregulations of the TNF-α gene and the activity of ADAM17, the membrane-associated TNF-α-converting enzyme. Like many other biological responses to LPA, induction of TNF-α by LPA also depended on the transactivation of the epidermal growth factor receptor (EGFR). Interestingly, our results revealed that ADAM17 was also the shedding protease responsible for the transactivation of EGFR by LPA in ovarian cancer cells. To explore the biological outcomes of LPA-induced TNF-α, we examined the effects of a TNF-α neutralizing antibody and recombinant TNF-α soluble receptor on LPA-stimulated expression of pro-tumorigenic cytokines and chemokines overexpressed in ovarian cancer. Blockade of TNF-α signaling significantly reduced the production of IL-8, IL-6, and CXCL1, suggesting a hierarchy of mechanisms contributing to the robust expression of the inflammatory mediators in response to LPA in ovarian cancer cells. In contrast, TNF-α inhibition did not affect LPA-dependent cell proliferation. Taken together, our results establish that the bioactive lipid LPA drives the expression of TNF-α to regulate an inflammatory network in ovarian cancer.


Subject(s)
Lysophospholipids/pharmacology , Ovarian Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics
6.
Sci Rep ; 10(1): 1450, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31996743

ABSTRACT

Fatty acid oxidation (FAO) is a key bioenergetic pathway often dysregulated in diseases. The current knowledge on FAO regulators in mammalian cells is limited and sometimes controversial. Previous FAO analyses involve nonphysiological culture conditions or lack adequate quantification. We herein described a convenient and quantitative assay to monitor dynamic FAO activities of mammalian cells in physiologically relevant settings. The method enabled us to assess various molecular and pharmacological modulators of the FAO pathway in established cell lines, primary cells and mice. Surprisingly, many previously proposed FAO inhibitors such as ranolazine and trimetazidine lacked FAO-interfering activity. In comparison, etomoxir at low micromolar concentrations was sufficient to saturate its target proteins and to block cellular FAO function. Oxfenicine, on the other hand, acted as a partial inhibitor of FAO. As another class of FAO inhibitors that transcriptionally repress FAO genes, antagonists of peroxisome proliferator-activated receptors (PPARs), particularly that of PPARα, significantly decreased cellular FAO activity. Our assay also had sufficient sensitivity to monitor upregulation of FAO in response to environmental glucose depletion and other energy-demanding cues. Altogether this study provided a reliable FAO assay and a clear picture of biological properties of potential FAO modulators in the mammalian system.


Subject(s)
Fatty Acids/metabolism , Glycine/analogs & derivatives , Mitochondria/metabolism , PPAR alpha/metabolism , Animals , Energy Metabolism , Epoxy Compounds/pharmacology , Female , Glycine/pharmacology , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Oxidation-Reduction , PPAR alpha/antagonists & inhibitors , Ranolazine/pharmacology , Trimetazidine/pharmacology
7.
Adv Cancer Res ; 143: 195-253, 2019.
Article in English | MEDLINE | ID: mdl-31202359

ABSTRACT

Fundamental metabolic pathways are essential for mammalian cells to provide energy, precursors for biosynthesis of macromolecules, and reducing power for redox regulation. While dysregulated metabolism (e.g., aerobic glycolysis also known as the Warburg effect) has long been recognized as a hallmark of cancer, recent discoveries of metabolic reprogramming in immune cells during their activation and differentiation have led to an emerging concept of "immunometabolism." Considering the recent success of cancer immunotherapy in the treatment of several cancer types, increasing research efforts are being made to elucidate alterations in metabolic profiles of cancer and immune cells during their interplays in the setting of cancer progression and immunotherapy. In this review, we summarize recent advances in studies of metabolic reprogramming in cancer as well as differentiation and functionality of various immune cells. In particular, we will elaborate how distinct metabolic pathways in the tumor microenvironment cause functional impairment of immune cells and contribute to immune evasion by cancer. Lastly, we highlight the potential of metabolically reprogramming the tumor microenvironment to promote effective and long-lasting antitumor immunity for improved immunotherapeutic outcomes.


Subject(s)
Antineoplastic Agents/therapeutic use , Immune System/immunology , Immunotherapy/methods , Neoplasms/drug therapy , Signal Transduction/drug effects , T-Lymphocytes/metabolism , Tumor Microenvironment/immunology , Animals , Cellular Reprogramming , Energy Metabolism , Glycolysis , Humans , Immune System/drug effects , Immune System/metabolism , Metabolome , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes/immunology
8.
Cancer Lett ; 435: 92-100, 2018 10 28.
Article in English | MEDLINE | ID: mdl-30102953

ABSTRACT

Cancer cells undergo metabolic reprogramming such as enhanced aerobic glycolysis, mutations in the tricarboxylic acid cycle enzymes, and upregulation of de novo lipid synthesis and glutaminolysis. These alterations are pivotal to the development and maintenance of the malignant phenotype of cancer cells in unfavorable tumor microenvironment or metastatic sites. Although mitochondrial fatty acid ß-oxidation (FAO) is a primary bioenergetic source, it has not been generally recognized as part of the metabolic landscape of cancer. The last few years, however, have seen a dramatic change in the view of cancer relevance of the FAO pathway. Many recent studies have provided significant evidence to support a "lipolytic phenotype" of cancer. FAO, like other well-defined metabolic pathways involved in cancer, is dysregulated in diverse human malignancies. Cancer cells rely on FAO for proliferation, survival, stemness, drug resistance, and metastatic progression. FAO is also reprogrammed in cancer-associated immune and other host cells, which may contribute to immune suppression and tumor-promoting microenvironment. This article reviews and puts into context our current understanding of multi-faceted roles of FAO in oncogenesis as well as anti-cancer therapeutic opportunities posed by the FAO pathway.


Subject(s)
Energy Metabolism , Fatty Acids/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Humans , Lipid Metabolism , Lipolysis , Neoplasms/pathology , Oxidation-Reduction , Tumor Microenvironment
9.
Cancers (Basel) ; 11(1)2018 Dec 31.
Article in English | MEDLINE | ID: mdl-30602670

ABSTRACT

The recent progresses in understanding of cancer glycolytic phenotype have offered new strategies to manage ovarian cancer and other malignancies. However, therapeutic targeting of glycolysis to treat cancer remains unsuccessful due to complex mechanisms of tumor glycolysis and the lack of selective, potent and safe glycolytic inhibitors. Recently, BAY-876 was identified as a new-generation inhibitor of glucose transporter 1 (GLUT1), a GLUT isoform commonly overexpressed but functionally poorly defined in ovarian cancer. Notably, BAY-876 has not been evaluated in any cell or preclinical animal models since its discovery. We herein took advantage of BAY-876 and molecular approaches to study GLUT1 regulation, targetability, and functional relevance to cancer glycolysis. The anti-tumor activity of BAY-876 was evaluated with ovarian cancer cell line- and patient-derived xenograft (PDX) models. Our results show that inhibition of GLUT1 is sufficient to block basal and stress-regulated glycolysis, and anchorage-dependent and independent growth of ovarian cancer cells. BAY-876 dramatically inhibits tumorigenicity of both cell line-derived xenografts and PDXs. These studies provide direct evidence that GLUT1 is causally linked to the glycolytic phenotype in ovarian cancer. BAY-876 is a potent blocker of GLUT1 activity, glycolytic metabolism and ovarian cancer growth, holding promise as a novel glycolysis-targeted anti-cancer agent.

10.
J Biol Chem ; 292(51): 20989-20997, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29046352

ABSTRACT

Dineolignans manassantin A and B from the plant Saururus cernuus are used in traditional medicine to manage a wide range of ailments such as edema, jaundice, and gonorrhea. Cell-based studies have identified several molecular target candidates of manassantin including NF-κB, MAPK, STAT3, and hypoxia-inducible factor 1α (HIF-1α). It is unclear whether or how these structurally diverse proteins or pathways mediate any of the medical benefits of manassantin in vivo Moreover, it has recently been reported that manassantin causes developmental arrest in zebrafish by inhibiting the mitochondrial complex I, but it is unknown whether manassantin inhibits mitochondrial respiration in intact mammalian cells and live animals. Here, we present direct evidence that manassantin potently and specifically inhibits the mitochondrial complex I and bioenergetic activity in mammalian systems. Manassantin had no effect on complex II- or complex IV-mediated respiration. Of note, it decreased NADH-ubiquinone reductase activity but not the activity of NADH-ferricyanide reductase. Treatment with manassantin reduced cellular ATP levels and concomitantly stimulated AMP-activated protein kinase in vitro and in vivo As an adaptive response to manassantin-induced bioenergetic deficiency, mammalian cells up-regulated aerobic glycolysis, a process mediated by AMP-activated protein kinase (AMPK) independently of HIF-1α. Together these results demonstrate a biologically important activity of manassantin in the control of complex I-mediated respiration and its profound effects on oxygen utilization, energy homeostasis, and glucose metabolism in mammalian cells.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Energy Metabolism/drug effects , Furans/pharmacology , Lignans/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Glycolysis/drug effects , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxygen Consumption/drug effects
11.
Peptides ; 92: 31-37, 2017 06.
Article in English | MEDLINE | ID: mdl-28363794

ABSTRACT

Insect selective excitatory ß-type sodium channel neurotoxins from scorpion venom (ß-NaScTxs) are composed of about 70-76 amino acid residues and share a common scaffold stabilized by four unique disulfide bonds. The phylogenetic analysis of these toxins was hindered by limited sequence data. In our recent study, two new insect selective excitatory ß-NaScTxs, LmIT and ImIT, were isolated from Lychas mucronatus and Isometrus maculatus, respectively. With the sequences previously reported, we examined the adaptive molecular evolution of insect selective excitatory ß-NaScTxs by estimating the nonsynonymous-to-synonymous rate ratio (ω=dN/dS). The results revealed 12 positively selected sites in the genes of insect selective excitatory ß-NaScTxs. Moreover, these positively selected sites match well with the sites important for interacting with sodium channels, as demonstrated in previous mutagenesis study. These results reveal that adaptive evolution after gene duplication is one of the most important genetic mechanisms of scorpion neurotoxin diversification.


Subject(s)
Evolution, Molecular , Neurotoxins/genetics , Scorpion Venoms/genetics , Scorpions/genetics , Sodium Channels/genetics , Amino Acid Sequence , Animals , Gene Library , Neurotoxins/chemistry , Neurotoxins/classification , Phylogeny , Scorpion Venoms/chemistry , Scorpion Venoms/classification , Selection, Genetic , Sequence Analysis, DNA , Sodium Channels/chemistry , Sodium Channels/classification
12.
Oncotarget ; 7(4): 3832-46, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26716645

ABSTRACT

Cancer cells rely on hyperactive de novo lipid synthesis for maintaining malignancy. Recent studies suggest involvement in cancer of fatty acid oxidation, a process functionally opposite to lipogenesis. A mechanistic link from lipid catabolism to oncogenic processes is yet to be established. Carnitine palmitoyltransferase 1 (CPT1) is a rate-limiting enzyme of fatty acid ß-oxidation (FAO) that catalyzes the transfer of long-chain acyl group of the acyl-CoA ester to carnitine, thereby shuttling fatty acids into the mitochondrial matrix for ß-oxidation. In the present study, we demonstrated that CPT1A was highly expressed in most ovarian cancer cell lines and primary ovarian serous carcinomas. Overexpression of CPT1A correlated with a poor overall survival of ovarian cancer patients. Inactivation of CPT1A decreased cellular ATP levels and induced cell cycle arrest at G0/G1, suggesting that ovarian cancer cells depend on or are addicted to CPT1A-mediated FAO for cell cycle progression. CPT1A deficiency also suppressed anchorage-independent growth and formation of xenografts from ovarian cancer cell lines. The cyclin-dependent kinase inhibitor p21WAF1 (p21) was identified as most consistently and robustly induced cell cycle regulator upon inactivation of CPT1A. Furthermore, p21 was transcriptionally upregulated by the FoxO transcription factors, which were in turn phosphorylated and activated by AMP-activated protein kinase and the mitogen-activated protein kinases JNK and p38. Our results established the oncogenic relevance of CPT1A and a mechanistic link from lipid catabolism to cell cycle regulation, suggesting that CPT1A could be a prognostic biomarker and rational target for therapeutic intervention of cancer.


Subject(s)
Carnitine O-Palmitoyltransferase/metabolism , Cell Cycle/physiology , Cystadenocarcinoma, Serous/pathology , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/pathology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carnitine O-Palmitoyltransferase/genetics , Cell Proliferation , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Electrophoretic Mobility Shift Assay , Fatty Acids/chemistry , Fatty Acids/metabolism , Female , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Humans , Lipid Metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Oxidation-Reduction , Phosphorylation , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Neoplasia ; 17(9): 723-734, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26476080

ABSTRACT

Lysophosphatidic acid (LPA), a blood-borne lipid mediator, is present in elevated concentrations in ascites of ovarian cancer patients and other malignant effusions. LPA is a potent mitogen in cancer cells. The mechanism linking LPA signal to cancer cell proliferation is not well understood. Little is known about whether LPA affects glucose metabolism to accommodate rapid proliferation of cancer cells. Here we describe that in ovarian cancer cells, LPA enhances glycolytic rate and lactate efflux. A real time PCR-based miniarray showed that hexokinase II (HK2) was the most dramatically induced glycolytic gene to promote glycolysis in LPA-treated cells. Analysis of the human HK2 gene promoter identified the sterol regulatory element-binding protein as the primary mediator of LPA-induced HK2 transcription. The effects of LPA on HK2 and glycolysis rely on LPA2, an LPA receptor subtype overexpressed in ovarian cancer and many other malignancies. We further examined the general role of growth factor-induced glycolysis in cell proliferation. Like LPA, epidermal growth factor (EGF) elicited robust glycolytic and proliferative responses in ovarian cancer cells. Insulin-like growth factor 1 (IGF-1) and insulin, however, potently stimulated cell proliferation but only modestly induced glycolysis. Consistent with their differential effects on glycolysis, LPA and EGF-dependent cell proliferation was highly sensitive to glycolytic inhibition while the growth-promoting effect of IGF-1 or insulin was more resistant. These results indicate that LPA- and EGF-induced cell proliferation selectively involves up-regulation of HK2 and glycolytic metabolism. The work is the first to implicate LPA signaling in promotion of glucose metabolism in cancer cells.


Subject(s)
Cell Proliferation/physiology , Glycolysis/physiology , Hexokinase/biosynthesis , Lysophospholipids/pharmacology , Ovarian Neoplasms/metabolism , Up-Regulation/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Glycolysis/drug effects , Humans , Up-Regulation/drug effects
14.
Anal Bioanal Chem ; 407(22): 6851-63, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26159569

ABSTRACT

Ovarian cancer (OVC) remains the most lethal gynecological malignancy in the world due to the combined lack of early-stage diagnostics and effective therapeutic strategies. The development and application of advanced proteomics technology and new experimental models has created unique opportunities for translational studies. In this study, we investigated the ovarian cancer proteome of the chicken, an emerging experimental model of OVC that develops ovarian tumors spontaneously. Matched plasma, ovary, and oviduct tissue biospecimens derived from healthy, early-stage OVC, and late-stage OVC birds were quantitatively characterized by label-free proteomics. Over 2600 proteins were identified in this study, 348 of which were differentially expressed by more than twofold (p ≤ 0.05) in early- and late-stage ovarian tumor tissue specimens relative to healthy ovarian tissues. Several of the 348 proteins are known to be differentially regulated in human cancers including B2M, CLDN3, EPCAM, PIGR, S100A6, S100A9, S100A11, and TPD52. Of particular interest was ovostatin 2 (OVOS2), a novel 165-kDa protease inhibitor found to be strongly upregulated in chicken ovarian tumors (p = 0.0005) and matched plasma (p = 0.003). Indeed, RT-quantitative PCR and Western blot analysis demonstrated that OVOS2 mRNA and protein were also upregulated in multiple human OVC cell lines compared to normal ovarian epithelia (NOE) cells and immunohistochemical staining confirmed overexpression of OVOS2 in primary human ovarian cancers relative to non-cancerous tissues. Collectively, these data provide the first evidence for involvement of OVOS2 in the pathogenesis of both chicken and human ovarian cancer.


Subject(s)
Chromatography, Liquid/methods , Mass Spectrometry/methods , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Proteome/chemistry , Proteome/metabolism , Amino Acid Sequence , Animals , Chickens , Conserved Sequence , Female , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Species Specificity
15.
PLoS One ; 7(10): e47255, 2012.
Article in English | MEDLINE | ID: mdl-23077578

ABSTRACT

Staphylococcus aureus is a major human pathogen and one of the more prominent pathogens causing biofilm related infections in clinic. Antibiotic resistance in S. aureus such as methicillin resistance is approaching an epidemic level. Antibiotic resistance is widespread among major human pathogens and poses a serious problem for public health. Conventional antibiotics are either bacteriostatic or bacteriocidal, leading to strong selection for antibiotic resistant pathogens. An alternative approach of inhibiting pathogen virulence without inhibiting bacterial growth may minimize the selection pressure for resistance. In previous studies, we identified a chemical series of low molecular weight compounds capable of inhibiting group A streptococcus virulence following this alternative anti-microbial approach. In the current study, we demonstrated that two analogs of this class of novel anti-virulence compounds also inhibited virulence gene expression of S. aureus and exhibited an inhibitory effect on S. aureus biofilm formation. This class of anti-virulence compounds could be a starting point for development of novel anti-microbial agents against S. aureus.


Subject(s)
Biofilms , Gene Expression Regulation, Bacterial/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Quinazolines/pharmacology , Streptokinase/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Humans , Methicillin Resistance/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Microbial Sensitivity Tests , Quinazolines/chemical synthesis , Small Molecule Libraries , Streptokinase/genetics
17.
Antimicrob Agents Chemother ; 56(11): 5923-37, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22964248

ABSTRACT

Biofilm formation on implantable medical devices is a major impediment to the treatment of nosocomial infections and promotes local progressive tissue destruction. Staphylococcus epidermidis infections are the leading cause of biofilm formation on indwelling devices. Bacteria in biofilms are highly resistant to antibiotic treatment, which in combination with the increasing prevalence of antibiotic resistance among human pathogens further complicates treatment of biofilm-related device infections. We have developed a novel plasma coating technology. Trimethylsilane (TMS) was used as a monomer to coat the surfaces of 316L stainless steel and grade 5 titanium alloy, which are widely used in implantable medical devices. The results of biofilm assays demonstrated that this TMS coating markedly decreased S. epidermidis biofilm formation by inhibiting the attachment of bacterial cells to the TMS-coated surfaces during the early phase of biofilm development. We also discovered that bacterial cells on the TMS-coated surfaces were more susceptible to antibiotic treatment than their counterparts in biofilms on uncoated surfaces. These findings suggested that TMS coating could result in a surface that is resistant to biofilm development and also in a bacterial community that is more sensitive to antibiotic therapy than typical biofilms.


Subject(s)
Biofilms/drug effects , Coated Materials, Biocompatible/pharmacology , Cross Infection/prevention & control , Prostheses and Implants/microbiology , Silanes/pharmacology , Staphylococcal Infections/prevention & control , Staphylococcus epidermidis/drug effects , Alloys/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms/growth & development , Ciprofloxacin/pharmacology , Coated Materials, Biocompatible/chemistry , Drug Resistance, Microbial , Humans , Microscopy, Confocal , Plasma Gases , Silanes/chemistry , Stainless Steel/chemistry , Staphylococcus epidermidis/growth & development , Surface Properties , Titanium/chemistry
18.
Proc Natl Acad Sci U S A ; 109(9): 3469-74, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22331877

ABSTRACT

The widespread occurrence of antibiotic resistance among human pathogens is a major public health problem. Conventional antibiotics typically target bacterial killing or growth inhibition, resulting in strong selection for the development of antibiotic resistance. Alternative therapeutic approaches targeting microbial pathogenicity without inhibiting growth might minimize selection for resistant organisms. Compounds inhibiting gene expression of streptokinase (SK), a critical group A streptococcal (GAS) virulence factor, were identified through a high-throughput, growth-based screen on a library of 55,000 small molecules. The lead compound [Center for Chemical Genomics 2979 (CCG-2979)] and an analog (CCG-102487) were confirmed to also inhibit the production of active SK protein. Microarray analysis of GAS grown in the presence of CCG-102487 showed down-regulation of a number of important virulence factors in addition to SK, suggesting disruption of a general virulence gene regulatory network. CCG-2979 and CCG-102487 both enhanced granulocyte phagocytosis and killing of GAS in an in vitro assay, and CCG-2979 also protected mice from GAS-induced mortality in vivo. These data suggest that the class of compounds represented by CCG-2979 may be of therapeutic value for the treatment of GAS and potentially other gram-positive infections in humans.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Gene Expression Regulation, Bacterial/drug effects , Quinazolines/therapeutic use , Streptococcal Infections/drug therapy , Streptococcus pyogenes/drug effects , Streptokinase/antagonists & inhibitors , Animals , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Depression, Chemical , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , High-Throughput Screening Assays , Host Specificity/genetics , Humans , Kanamycin Resistance/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Structure , Phagocytosis/drug effects , Plasminogen/genetics , Promoter Regions, Genetic/genetics , Quinazolines/isolation & purification , Quinazolines/pharmacology , Small Molecule Libraries , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Streptokinase/biosynthesis , Streptokinase/genetics , Virulence/drug effects , Virulence/genetics
19.
J Proteomics ; 75(5): 1563-76, 2012 Feb 16.
Article in English | MEDLINE | ID: mdl-22155128

ABSTRACT

Venom is an important genetic development crucial to the survival of scorpions for over 400 million years. We studied the evolution of the scorpion venom arsenal by means of comparative transcriptome analysis of venom glands and phylogenetic analysis of shared types of venom peptides and proteins between buthids and euscorpiids. Fifteen types of venom peptides and proteins were sequenced during the venom gland transcriptome analyses of two Buthidae species (Lychas mucronatus and Isometrus maculatus) and one Euscorpiidae species (Scorpiops margerisonae). Great diversity has been observed in translated amino acid sequences of these transcripts for venom peptides and proteins. Seven types of venom peptides and proteins were shared between buthids and euscorpiids. Molecular phylogenetic analysis revealed that at least five of the seven common types of venom peptides and proteins were likely recruited into the scorpion venom proteome before the lineage split between Buthidae and Euscorpiidae with their corresponding genes undergoing individual or multiple gene duplication events. These are α-KTxs, ßKSPNs (ß-KTxs and scorpines), anionic peptides, La1-like peptides, and SPSVs (serine proteases from scorpion venom). Multiple types of venom peptides and proteins were demonstrated to be continuously recruited into the venom proteome during the evolution process of individual scorpion lineages. Our results provide an insight into the recruitment pattern of the scorpion venom arsenal for the first time.


Subject(s)
Arthropod Proteins/biosynthesis , Evolution, Molecular , Peptides/metabolism , Scorpion Venoms/biosynthesis , Scorpions/metabolism , Transcription, Genetic/physiology , Animals , Arthropod Proteins/genetics , Peptides/genetics , Scorpion Venoms/genetics , Scorpions/genetics
20.
PLoS One ; 6(11): e27548, 2011.
Article in English | MEDLINE | ID: mdl-22087336

ABSTRACT

BACKGROUND: Kunitz-type venom peptides have been isolated from a wide variety of venomous animals. They usually have protease inhibitory activity or potassium channel blocking activity, which by virtue of the effects on predator animals are essential for the survival of venomous animals. However, no Kunitz-type peptides from scorpion venom have been functionally characterized. PRINCIPAL FINDINGS: A new Kunitz-type venom peptide gene precursor, SdPI, was cloned and characterized from a venom gland cDNA library of the scorpion Lychas mucronatus. It codes for a signal peptide of 21 residues and a mature peptide of 59 residues. The mature SdPI peptide possesses a unique cysteine framework reticulated by three disulfide bridges, different from all reported Kunitz-type proteins. The recombinant SdPI peptide was functionally expressed. It showed trypsin inhibitory activity with high potency (K(i) = 1.6×10(-7) M) and thermostability. CONCLUSIONS: The results illustrated that SdPI is a potent and stable serine protease inhibitor. Further mutagenesis and molecular dynamics simulation revealed that SdPI possesses a serine protease inhibitory active site similar to other Kunitz-type venom peptides. To our knowledge, SdPI is the first functionally characterized Kunitz-type trypsin inhibitor derived from scorpion venom, and it represents a new class of Kunitz-type venom peptides.


Subject(s)
Scorpion Venoms/chemistry , Trypsin Inhibitors/isolation & purification , Animals , Cloning, Molecular , Gene Library , Protein Stability , Serine Proteinase Inhibitors , Trypsin Inhibitors/chemistry , Trypsin Inhibitors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...