Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38463975

ABSTRACT

Previous studies have implicated persistent innate immune signaling in the pathogenesis of arrhythmogenic cardiomyopathy (ACM), a familial non-ischemic heart muscle disease characterized by life-threatening arrhythmias and progressive myocardial injury. Here, we provide new evidence implicating inflammatory lipid autocoids in ACM. We show that specialized pro-resolving lipid mediators are reduced in hearts of Dsg2mut/mut mice, a well characterized mouse model of ACM. We also found that ACM disease features can be reversed in rat ventricular myocytes expressing mutant JUP by the pro-resolving epoxy fatty acid (EpFA) 14,15-eicosatrienoic acid (14-15-EET), whereas 14,15-EE-5(Z)E which antagonizes actions of the putative 14,15-EET receptor, intensified nuclear accumulation of the desmosomal protein plakoglobin. Soluble epoxide hydrolase (sEH), an enzyme that rapidly converts pro-resolving EpFAs into polar, far less active or even pro-inflammatory diols, is highly expressed in cardiac myocytes in Dsg2mut/mut mice. Inhibition of sEH prevented progression of myocardial injury in Dsg2mut/mut mice and led to recovery of contractile function. This was associated with reduced myocardial expression of genes involved in the innate immune response and fewer pro-inflammatory macrophages expressing CCR2, which mediate myocardial injury in Dsg2mut/mut mice. These results suggest that pro-inflammatory eicosanoids contribute to the pathogenesis of ACM and, further, that inhibition of sEH may be an effective, mechanism-based therapy for ACM patients.

2.
Proc Natl Acad Sci U S A ; 121(7): e2314085121, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38330013

ABSTRACT

Cancer therapy, including immunotherapy, is inherently limited by chronic inflammation-induced tumorigenesis and toxicity within the tumor microenvironment. Thus, stimulating the resolution of inflammation may enhance immunotherapy and improve the toxicity of immune checkpoint inhibition (ICI). As epoxy-fatty acids (EpFAs) are degraded by the enzyme soluble epoxide hydrolase (sEH), the inhibition of sEH increases endogenous EpFA levels to promote the resolution of cancer-associated inflammation. Here, we demonstrate that systemic treatment with ICI induces sEH expression in multiple murine cancer models. Dietary omega-3 polyunsaturated fatty acid supplementation and pharmacologic sEH inhibition, both alone and in combination, significantly enhance anti-tumor activity of ICI in these models. Notably, pharmacological abrogation of the sEH pathway alone or in combination with ICI counter-regulates an ICI-induced pro-inflammatory and pro-tumorigenic cytokine storm. Thus, modulating endogenous EpFA levels through dietary supplementation or sEH inhibition may represent a unique strategy to enhance the anti-tumor activity of paradigm cancer therapies.


Subject(s)
Epoxide Hydrolases , Neoplasms , Mice , Humans , Animals , Epoxide Hydrolases/metabolism , Fatty Acids/metabolism , Inflammation/metabolism , Neoplasms/therapy , Immunotherapy , Tumor Microenvironment
3.
Int J Mol Sci ; 24(12)2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37373086

ABSTRACT

Parkinson's disease (PD) is an increasingly common neurodegenerative movement disorder with contributing factors that are still largely unexplored and currently no effective intervention strategy. Epidemiological and pre-clinical studies support the close association between environmental toxicant exposure and PD incidence. Aflatoxin B1 (AFB1), a hazardous mycotoxin commonly present in food and environment, is alarmingly high in many areas of the world. Previous evidence suggests that chronic exposure to AFB1 leads to neurological disorders as well as cancer. However, whether and how aflatoxin B1 contributes to the pathogenesis of PD is poorly understood. Here, oral exposure to AFB1 is shown to induce neuroinflammation, trigger the α-synuclein pathology, and cause dopaminergic neurotoxicity. This was accompanied by the increased expression and enzymatic activity of soluble epoxide hydrolase (sEH) in the mouse brain. Importantly, genetic deletion or pharmacological inhibition of sEH alleviated the AFB1-induced neuroinflammation by reducing microglia activation and suppressing pro-inflammatory factors in the brain. Furthermore, blocking the action of sEH attenuated dopaminergic neuron dysfunction caused by AFB1 in vivo and in vitro. Together, our findings suggest a contributing role of AFB1 to PD etiology and highlight sEH as a potential pharmacological target for alleviating PD-related neuronal disorders caused by AFB1 exposure.


Subject(s)
Neurodegenerative Diseases , Neurotoxicity Syndromes , Parkinson Disease , Mice , Animals , Aflatoxin B1/toxicity , Epoxide Hydrolases/genetics , Epoxide Hydrolases/metabolism , Neuroinflammatory Diseases , Parkinson Disease/metabolism , Brain/metabolism
4.
Int J Mol Sci ; 24(5)2023 Feb 26.
Article in English | MEDLINE | ID: mdl-36901999

ABSTRACT

Aging, which is characterized by enhanced cell senescence and functional decline of tissues, is a major risk factor for many chronic diseases. Accumulating evidence shows that age-related dysfunction in the colon leads to disorders in multiple organs and systemic inflammation. However, the detailed pathological mechanisms and endogenous regulators underlying colon aging are still largely unknown. Here, we report that the expression and activity of the soluble epoxide hydrolase (sEH) enzyme are increased in the colon of aged mice. Importantly, genetic knockout of sEH attenuated the age-related upregulation of senescent markers p21, p16, Tp53, and ß-galactosidase in the colon. Moreover, sEH deficiency alleviated aging-associated endoplasmic reticulum (ER) stress in the colon by reducing both the upstream regulators Perk and Ire1 as well as the downstream pro-apoptotic effectors Chop and Gadd34. Furthermore, treatment with sEH-derived linoleic acid metabolites, dihydroxy-octadecenoic acids (DiHOMEs), decreased cell viability and increased ER stress in human colon CCD-18Co cells in vitro. Together, these results support that the sEH is a key regulator of the aging colon, which highlights its potential application as a therapeutic target for reducing or treating age-related diseases in the colon.


Subject(s)
Cellular Senescence , Endoplasmic Reticulum Stress , Epoxide Hydrolases , Animals , Humans , Mice , Aging , Colon/metabolism , Epoxide Hydrolases/metabolism , Inflammation , Mice, Inbred C57BL
5.
Ecotoxicol Environ Saf ; 249: 114417, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36525946

ABSTRACT

Aflatoxin B1 (AFB1) contamination in food and feed leads to severe global health problems. Acting as the frontier immunological barrier, the intestinal mucosa is constantly challenged by exposure to foodborne toxins such as AFB1 via contaminated diets, but the detailed toxic mechanism and endogenous regulators of AFB1 toxicity are still unclear. Here, we showed that AFB1 disrupted intestinal immune function by suppressing macrophages, especially M2 macrophages, and antimicrobial peptide-secreting Paneth cells. Using an oxylipinomics approach, we identified that AFB1 immunotoxicity is associated with decreased epoxy fatty acids, notably epoxyeicosatrienoic acids, and increased soluble epoxide hydrolase (sEH) levels in the intestine. Furthermore, sEH deficiency or inhibition rescued the AFB1-compromised intestinal immunity by restoring M2 macrophages as well as Paneth cells and their-derived lysozyme and α-defensin-3 in mice. Altogether, our study demonstrates that AFB1 exposure impairs intestinal immunity, at least in part, in a sEH-mediated way. Moreover, the present study supports the potential application of pharmacological intervention by inhibiting the sEH enzyme in alleviating intestinal immunotoxicity and associated complications caused by AFB1 global contamination.


Subject(s)
Aflatoxin B1 , Epoxide Hydrolases , Animals , Mice , Aflatoxin B1/toxicity , Diet , Immunity , Intestines
6.
Nutrients ; 12(11)2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33126566

ABSTRACT

Substantial human and animal studies support the beneficial effects of ω-3 polyunsaturated fatty acids (PUFAs) on colonic inflammation and colorectal cancer (CRC). However, there are inconsistent results, which have shown that ω-3 PUFAs have no effect or even detrimental effects, making it difficult to effectively implement ω-3 PUFAs for disease prevention. A better understanding of the molecular mechanisms for the anti-inflammatory and anticancer effects of ω-3 PUFAs will help to clarify their potential health-promoting effects, provide a scientific base for cautions for their use, and establish dietary recommendations. In this review, we summarize recent studies of ω-3 PUFAs on colonic inflammation and CRC and discuss the potential roles of ω-3 PUFA-metabolizing enzymes, notably the cytochrome P450 monooxygenases, in mediating the actions of ω-3 PUFAs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Colitis/prevention & control , Colorectal Neoplasms/prevention & control , Fatty Acids, Omega-3/pharmacology , Animals , Colon/drug effects , Cytochrome P-450 Enzyme System/metabolism , Epoxide Hydrolases/metabolism , Humans , Mixed Function Oxygenases/metabolism
7.
Proc Natl Acad Sci U S A ; 117(35): 21576-21587, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32801214

ABSTRACT

Toxic environmental carcinogens promote cancer via genotoxic and nongenotoxic pathways, but nongenetic mechanisms remain poorly characterized. Carcinogen-induced apoptosis may trigger escape from dormancy of microtumors by interfering with inflammation resolution and triggering an endoplasmic reticulum (ER) stress response. While eicosanoid and cytokine storms are well-characterized in infection and inflammation, they are poorly characterized in cancer. Here, we demonstrate that carcinogens, such as aflatoxin B1 (AFB1), induce apoptotic cell death and the resulting cell debris stimulates hepatocellular carcinoma (HCC) tumor growth via an "eicosanoid and cytokine storm." AFB1-generated debris up-regulates cyclooxygenase-2 (COX-2), soluble epoxide hydrolase (sEH), ER stress-response genes including BiP, CHOP, and PDI in macrophages. Thus, selective cytokine or eicosanoid blockade is unlikely to prevent carcinogen-induced cancer progression. Pharmacological abrogation of both the COX-2 and sEH pathways by PTUPB prevented the debris-stimulated eicosanoid and cytokine storm, down-regulated ER stress genes, and promoted macrophage phagocytosis of debris, resulting in suppression of HCC tumor growth. Thus, inflammation resolution via dual COX-2/sEH inhibition is an approach to prevent carcinogen-induced cancer.


Subject(s)
Cytokines/metabolism , Eicosanoids/metabolism , Liver Neoplasms/metabolism , Aflatoxin B1/adverse effects , Animals , Apoptosis , Carcinogenesis/metabolism , Carcinogens/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line , Cyclooxygenase 2/metabolism , Cytokines/immunology , Disease Progression , Eicosanoids/immunology , Epoxide Hydrolases/metabolism , Hep G2 Cells , Humans , Inflammation/metabolism , Liver Neoplasms/physiopathology , Macrophages/metabolism , Mice , Neoplastic Processes
8.
Am J Pathol ; 190(9): 1782-1788, 2020 09.
Article in English | MEDLINE | ID: mdl-32650004

ABSTRACT

Severe coronavirus disease 2019 (COVID-19) symptoms, including systemic inflammatory response and multisystem organ failure, are now affecting thousands of infected patients and causing widespread mortality. Coronavirus infection causes tissue damage, which triggers the endoplasmic reticulum stress response and subsequent eicosanoid and cytokine storms. Although proinflammatory eicosanoids, including prostaglandins, thromboxanes, and leukotrienes, are critical mediators of physiological processes, such as inflammation, fever, allergy, and pain, their roles in COVID-19 are poorly characterized. Arachidonic acid-derived epoxyeicosatrienoic acids could alleviate the systemic hyperinflammatory response in COVID-19 infection by modulating endoplasmic reticulum stress and stimulating the resolution of inflammation. Soluble epoxide hydrolase (sEH) inhibitors, which increase endogenous epoxyeicosatrienoic acid levels, exhibit potent anti-inflammatory activity and inhibit various pathologic processes in preclinical disease models, including pulmonary fibrosis, thrombosis, and acute respiratory distress syndrome. Therefore, targeting eicosanoids and sEH could be a novel therapeutic approach in combating COVID-19. In this review, we discuss the predominant role of eicosanoids in regulating the inflammatory cascade and propose the potential application of sEH inhibitors in alleviating COVID-19 symptoms. The host-protective action of omega-3 fatty acid-derived epoxyeicosanoids and specialized proresolving mediators in regulating anti-inflammation and antiviral response is also discussed. Future studies determining the eicosanoid profile in COVID-19 patients or preclinical models are pivotal in providing novel insights into coronavirus-host interaction and inflammation modulation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Animals , Betacoronavirus/pathogenicity , COVID-19 , Eicosanoids/pharmacology , Eicosanoids/therapeutic use , Endoplasmic Reticulum Stress/drug effects , Humans , Pandemics , SARS-CoV-2
9.
Small ; 16(36): e2001858, 2020 09.
Article in English | MEDLINE | ID: mdl-32519440

ABSTRACT

The recent ban of titanium dioxide (TiO2 ) as a food additive (E171) in France intensified the controversy on safety of foodborne-TiO2 nanoparticles (NPs). This study determines the biological effects of TiO2 NPs and TiO2 (E171) in obese and non-obese mice. Oral consumption (0.1 wt% in diet for 8 weeks) of TiO2 (E171, 112 nm) and TiO2 NPs (33 nm) does not cause severe toxicity in mice, but significantly alters composition of gut microbiota, for example, increased abundance of Firmicutes phylum and decreased abundance of Bacteroidetes phylum and Bifidobacterium and Lactobacillus genera, which are accompanied by decreased cecal levels of short-chain fatty acids. Both TiO2 (E171) and TiO2 NPs increase abundance of pro-inflammatory immune cells and cytokines in the colonic mucosa, indicating an inflammatory state. Importantly, TiO2 NPs cause stronger colonic inflammation than TiO2 (E171), and obese mice are more susceptible to the effects. A microbiota transplant study demonstrates that altered fecal microbiota by TiO2 NPs directly mediate inflammatory responses in the mouse colon. Furthermore, proteomic analysis shows that TiO2 NPs cause more alterations in multiple pathways in the liver and colon of obese mice than non-obese mice. This study provides important information on the health effects of foodborne inorganic nanoparticles.


Subject(s)
Colon , Dysbiosis , Gastrointestinal Microbiome , Metal Nanoparticles , Proteome , Titanium , Animals , Colon/drug effects , Dysbiosis/chemically induced , Food Contamination , Gastrointestinal Microbiome/drug effects , Inflammation/chemically induced , Metal Nanoparticles/toxicity , Mice , Mice, Obese , Proteome/drug effects , Proteomics , Titanium/toxicity
10.
Proc Natl Acad Sci U S A ; 117(15): 8431-8436, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32220957

ABSTRACT

Intestinal barrier dysfunction, which leads to translocation of bacteria or toxic bacterial products from the gut into bloodstream and results in systemic inflammation, is a key pathogenic factor in many human diseases. However, the molecular mechanisms leading to intestinal barrier defects are not well understood, and there are currently no available therapeutic approaches to target intestinal barrier function. Here we show that soluble epoxide hydrolase (sEH) is an endogenous regulator of obesity-induced intestinal barrier dysfunction. We find that sEH is overexpressed in the colons of obese mice. In addition, pharmacologic inhibition or genetic ablation of sEH abolishes obesity-induced gut leakage, translocation of endotoxin lipopolysaccharide or bacteria, and bacterial invasion-induced adipose inflammation. Furthermore, systematic treatment with sEH-produced lipid metabolites, dihydroxyeicosatrienoic acids, induces bacterial translocation and colonic inflammation in mice. The actions of sEH are mediated by gut bacteria-dependent mechanisms, since inhibition or genetic ablation of sEH fails to attenuate obesity-induced gut leakage and adipose inflammation in mice lacking gut bacteria. Overall, these results support that sEH is a potential therapeutic target for obesity-induced intestinal barrier dysfunction, and that sEH inhibitors, which have been evaluated in human clinical trials targeting other human disorders, could be promising agents for prevention and/or treatment.


Subject(s)
Bacterial Translocation , Epoxide Hydrolases/immunology , Intestinal Diseases/enzymology , Intestines/enzymology , Obesity/complications , Adipose Tissue/immunology , Animals , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Physiological Phenomena , Epoxide Hydrolases/genetics , Gastrointestinal Microbiome , Humans , Intestinal Diseases/etiology , Intestinal Diseases/immunology , Intestinal Diseases/microbiology , Intestines/immunology , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/enzymology , Obesity/genetics
11.
Food Funct ; 11(2): 1684-1691, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32037431

ABSTRACT

Previous studies have shown that curcumin, a bioactive dietary compound with a thiol-reactive α,ß-unsaturated carbonyl moiety, can covalently modify protein thiols. However, most of the previous studies were performed in cultured cells or cell-free enzyme systems, and so it remains unknown whether curcumin could covalently modify proteins after oral administration in vivo. Using click chemistry-based fluorescence imaging, here we show that oral administration of dialkyne-curcumin (Di-Cur), a "click" probe mimicking curcumin, results in covalent modifications of cellular proteins in colon and liver tissues, but not in other tissues, in mice. This result suggests that oral administration of curcumin leads to the formation of the curcumin-protein complex in a tissue-specific manner, which could contribute to the biological effects and/or pharmacokinetics of curcumin. Further studies to elucidate the identities of curcumin-binding proteins could greatly help us to better understand the molecular mechanisms of curcumin, and develop novel strategies for disease prevention.


Subject(s)
Click Chemistry/methods , Curcumin , Administration, Oral , Animals , Cell Line, Tumor , Colon/metabolism , Curcumin/administration & dosage , Curcumin/chemistry , Curcumin/metabolism , Curcumin/pharmacokinetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Probes , Protein Binding , Tissue Distribution
12.
J Nutr Biochem ; 76: 108286, 2020 02.
Article in English | MEDLINE | ID: mdl-31918337

ABSTRACT

Peroxidation of polyunsaturated fatty acids leads to the formation of a large array of lipid-derived electrophiles (LDEs), many of which are important signaling molecules involved in the pathogenesis of human diseases. Previous research has shown that one of such LDEs, trans, trans-2,4-decadienal (tt-DDE), increases inflammation, however, the underlying mechanisms are not well understood. Here we used click chemistry-based proteomics to identify the cellular targets which are required for the pro-inflammatory effects of tt-DDE. We found that treatment with tt-DDE increased cytokine production, JNK phosphorylation, and activation of NF-κB signaling in macrophage cells, and increased severity of dextran sulfate sodium (DSS)-induced colonic inflammation in mice, demonstrating its pro-inflammatory effects in vitro and in vivo. Using click chemistry-based proteomics, we found that tt-DDE directly interacts with Hsp90 and 14-3-3ζ, which are two important proteins involved in inflammation and tumorigenesis. Furthermore, siRNA knockdown of Hsp90 or 14-3-3ζ abolished the pro-inflammatory effects of tt-DDE in macrophage cells. Together, our results support that tt-DDE increases inflammatory responses via Hsp90- and 14-3-3ζ-dependent mechanisms.


Subject(s)
14-3-3 Proteins/metabolism , Aldehydes/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Inflammation/metabolism , Lipid Peroxidation , Macrophages/metabolism , Animals , Cell Survival/drug effects , Colon/metabolism , Cytokines/metabolism , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Proteomics , RAW 264.7 Cells , RNA, Small Interfering/metabolism , Signal Transduction
13.
Cell Mol Immunol ; 17(11): 1136-1147, 2020 11.
Article in English | MEDLINE | ID: mdl-31511643

ABSTRACT

Interleukin-17A (IL-17A)-producing helper T (Th17) cells are a subset of CD4+ T cells that play important pathological roles in autoimmune diseases. Although the intrinsic pathways of Th17 cell differentiation have been well described, how instructive signals derived from the innate immune system trigger the Th17 response and inflammation remains poorly understood. Here, we report that mice deficient in REGγ, a proteasome activator belonging to the 11S family, exhibit significantly deteriorated autoimmune neuroinflammation in an experimental autoimmune encephalomyelitis (EAE) model with augmented Th17 cell polarization in vivo. The results of the adoptive transfer of CD4+ T cells or dendritic cells (DCs) suggest that this phenotype is driven by DCs rather than T cells. Furthermore, REGγ deficiency promotes the expression of integrin αvß8 on DCs, which activates the maturation of TGF-ß1 to enhance Th17 cell development. Mechanistically, this process is mediated by the REGγ-proteasome-dependent degradation of IRF8, a transcription factor for αvß8. Collectively, our findings delineate a previously unknown mechanism by which REGγ-mediated protein degradation in DCs controls the differentiation of Th17 cells and the onset of an experimental autoimmune disease.


Subject(s)
Autoantigens/metabolism , Autoimmunity , Cell Differentiation , Dendritic Cells/immunology , Inflammation/immunology , Proteasome Endopeptidase Complex/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Animals , Cell Polarity , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interferon Regulatory Factors/metabolism , Interleukin-6/metabolism , Mice, Inbred C57BL , Models, Biological , Proteasome Endopeptidase Complex/deficiency , Transforming Growth Factor beta1/metabolism
14.
Gut Microbes ; 12(1): 1690364, 2020 11 09.
Article in English | MEDLINE | ID: mdl-31760871

ABSTRACT

Triclocarban (TCC) is a widely used antimicrobial ingredient in consumer products and is a ubiquitous contaminant in the environment. In 2016, the FDA removed TCC from over-the-counter handwashing products, but this compound is still approved for use in many other personal care products. A better understanding of its impact on human health could lead to significant impact for public health and regulatory policies. Here we show that exposure to low-dose TCC exaggerated the severity of colitis and exacerbated the development of colitis-associated colon tumorigenesis, via gut microbiota-dependent mechanisms. Exposure to TCC increased dextran sodium sulfate (DSS)- and interleukin 10 (IL-10) knockout-induced colitis, and exaggerated azoxymethane (AOM)/DSS-induced colon tumorigenesis in mice. Regarding the mechanisms, TCC exposure reduced the diversity and altered the composition of gut microbiota and failed to promote DSS-induced colitis in mice lacking the microbiota, supporting that the presence of the microbiota is critical for the pro-colitis effects of TCC. Together, these results support TCC could be a novel risk factor for colitis and colitis-associated colon cancer, and further regulatory policies on this compound could be needed.


Subject(s)
Anti-Infective Agents, Local/adverse effects , Carbanilides/adverse effects , Cell Transformation, Neoplastic/drug effects , Colitis/chemically induced , Colonic Neoplasms/chemically induced , Gastrointestinal Microbiome/drug effects , Animals , Anti-Infective Agents, Local/pharmacology , Bifidobacterium longum subspecies infantis/growth & development , Carbanilides/pharmacology , Colitis/microbiology , Colitis/pathology , Colonic Neoplasms/pathology , Dextran Sulfate , Humans , Inflammation/chemically induced , Interleukin-10/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Water Pollutants, Chemical/adverse effects
15.
Toxicol Sci ; 174(1): 92-99, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31868902

ABSTRACT

Triclocarban (3,4,4'-trichlorocarbanilide, TCC) is a high-volume chemical used as an antimicrobial ingredient in many consumer and personal care products. In 2016, the Food and Drug Administration removed TCC from over-the-counter hand washing products. However, TCC remains approved to use in many other products and is a ubiquitous contaminant in the environment; furthermore, many common food crops can efficiently accumulate environmental TCC, resulting in potential human exposure through oral ingestion of contaminated food products. Therefore, human exposure to TCC could be a long-lasting and serious problem. A better understanding of its impact on human health could lead to important impact for public health and regulatory policy. Using a spontaneous colonic inflammation model in Il-10-/- mice, here we demonstrate that exposure to TCC, at doses relevant to human exposure, exaggerates spontaneous colonic inflammation in Il-10-/- mice, with reduced colon length, increase fecal concentration of lipocalin 2, enhanced gene expression of Il-6 and Ifn-γ in the colon, and exaggerated crypt damage in the colon. Collectively, these results support that TCC could be a potential environmental risk factor of colitis and associated gut diseases.


Subject(s)
Anti-Infective Agents/toxicity , Carbanilides/toxicity , Colitis/chemically induced , Colon/drug effects , Interleukin-10/deficiency , Animals , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Lipocalin-2/metabolism , Male , Mice, Knockout , Risk Assessment
16.
ACS Appl Mater Interfaces ; 11(47): 44894-44904, 2019 Nov 27.
Article in English | MEDLINE | ID: mdl-31657898

ABSTRACT

We presented an improved surface-enhanced Raman scattering (SERS) mapping technique for the imaging of pesticides on biological samples including tomato leaves, fruits, and mouse skin using a gold nanoparticle mirror as the SERS substrate. The gold nanoparticle mirror was fabricated using 50 nm commercial citrate-capped gold nanoparticles upon the interface of water and a mediating solvent that was prepared using acetonitrile and hexane. The properties of the gold nanoparticle mirror were compared with gold nanoparticles, and the mirror displayed higher sensitivity with a limit of detection of 0.07 µg/cm2 and better reproducibility with a relative standard deviation of 5.48% for the SERS mapping of pesticide (ferbam) on biological samples. The gold mirror-based SERS mapping technique was also used to investigate pesticide transmission from tomato fruit surfaces to mouse skin after 1 mg/cm2 of pesticides was administered upon the fruit, and the results showed that about 23% of the pesticide was transmitted from the fruit to the mouse skin. We also found that pesticides on the contaminated hand could not be completely removed by routine rinsing with tap water for 2 min. This study provides an effective approach for the imaging of pesticides on biological tissues that would facilitate research on pesticide behaviors both on and in biological systems.


Subject(s)
Gold/chemistry , Pesticides/chemistry , Skin/chemistry , Solanum lycopersicum/chemistry , Spectrum Analysis, Raman/methods , Animals , Fruit/chemistry , Male , Metal Nanoparticles/chemistry , Mice , Mice, Inbred C57BL , Pesticide Residues , Plant Leaves/chemistry , Spectrum Analysis, Raman/instrumentation
17.
Adv Exp Med Biol ; 1161: 115-123, 2019.
Article in English | MEDLINE | ID: mdl-31562626

ABSTRACT

Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer-related death in the United States. It is important to discover novel cellular targets which are crucial in the pathogenesis of CRC, which could facilitate development of mechanism-based strategies to reduce the risks of CRC. Emerging studies support that the cytochrome P450 (CYP) monooxygenase/soluble epoxide hydrolase (sEH) pathway and their eicosanoid metabolites play critical roles in colonic inflammation and CRC, and could be therapeutically explored for treating or preventing CRC. Here in this review, we discuss recent studies about the roles of the CYP/sEH eicosanoid pathway in the pathogenesis of colonic inflammation and CRC.


Subject(s)
Carcinogenesis , Colorectal Neoplasms , Cytochrome P-450 Enzyme System , Eicosanoids , Signal Transduction , Carcinogenesis/metabolism , Colorectal Neoplasms/physiopathology , Cytochrome P-450 Enzyme System/metabolism , Eicosanoids/metabolism , Epoxide Hydrolases/metabolism , Humans
18.
Nat Commun ; 10(1): 3665, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31413323

ABSTRACT

Anaerobic bacteria represent an overlooked rich source of biological and chemical diversity. Due to the challenge of cultivation and genetic intractability, assessing the capability of their biosynthetic gene clusters (BGCs) for secondary metabolite production requires an efficient heterologous expression system. However, this kind of host system is still unavailable. Here, we use the facultative anaerobe Streptococcus mutans UA159 as a heterologous host for the expression of BGCs from anaerobic bacteria. A natural competence based large DNA fragment cloning (NabLC) technique was developed, which can move DNA fragments up to 40-kb directly and integrate a 73.7-kb BGC to the genome of S. mutans UA159 via three rounds of NabLC cloning. Using this system, we identify an anti-infiltration compound, mutanocyclin, from undefined BGCs from human oral bacteria. We anticipate this host system will be useful for heterologous expression of BGCs from anaerobic bacteria.


Subject(s)
Bacteria, Anaerobic/genetics , Biosynthetic Pathways/genetics , Cloning, Molecular/methods , Multigene Family/genetics , Streptococcus mutans/genetics , Humans , Microbiota/genetics , Mouth , Peptides , Polyketides , Terpenes
19.
J Nutr Biochem ; 68: 1-6, 2019 06.
Article in English | MEDLINE | ID: mdl-31005846

ABSTRACT

Curcumin, a bis-α, ß-unsaturated ß-diketon dietary compound from turmeric, is among the most promising dietary compounds for preventing chronic diseases. Previous research has shown that curcumin is highly reactive toward protein thiols to form curcumin-protein adducts, however, the interactions of curcumin with proteins are under-studied. Here we report the design and synthesis of "click" chemistry probes of curcumin, mono-propargyl curcumin (mono-Cur) and di-propargyl curcumin (di-Cur), and use the click probes to study curcumin-proteins interactions in vitro and in vivo. We find that compared with di-Cur, the mono-Cur probe has more potent biological effects and enhanced effects to label proteins in cultured cells, suggesting that mono-Cur is a better click probe to study the biological actions of curcumin. Furthermore, using the mono-Cur probe, we find that oral administration of this probe in mice leads to formation of curcumin-protein adducts in colon and liver tissues of C57BL/6 mice, suggesting that curcumin could covalently modify cellular proteins in vivo. Together, these results could help us to better understand protein-curcumin interactions. These results could in part explain the poor pharmacokinetics of curcumin; in addition, formation of these protein adducts could contribute to the health-promoting effects of curcumin.


Subject(s)
Curcumin/pharmacokinetics , Molecular Probes/chemical synthesis , Proteins/metabolism , Administration, Oral , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Click Chemistry , Colon/drug effects , Colon/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Curcumin/chemistry , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Molecular Probes/administration & dosage
20.
J Agric Food Chem ; 67(33): 9168-9177, 2019 Aug 21.
Article in English | MEDLINE | ID: mdl-30810035

ABSTRACT

Strawberry (Fragaria chiloensis) is a major edible berry with various potential health benefits. This study determined the protective effects of whole strawberry (WS) against dextran-sulfate-sodium-induced colitis in mice. In colitic mice, dietary WS reduced the disease activity index, prevented the colon shortening and spleen enlargement, and alleviated the colonic tissue damages. The abundance of proinflammatory immune cells was reduced by dietary WS in the colonic mucosa, which was accompanied by the suppression of overproduction of proinflammatory cytokines. Western blotting and immunohistochemical analysis revealed that dietary WS decreased the expression of proinflammatory proteins in the colonic mucosa. Moreover, dietary WS partially reversed the alteration of gut microbiota in the colitic mice by increasing the abundance of potential beneficial bacteria, e.g., Bifidobacterium and Lactobacillus, and decreasing the abundance of potential harmful bacteria, e.g., Dorea and Bilophila. Dietary WS also restored the decreased production of short-chain fatty acids in the cecum of the colitic mice. The results revealed the anti-inflammatory effects and mechanisms of dietary WS in the colon, which is critical for the rational utilization of strawberry for the prevention of inflammation-driven diseases.


Subject(s)
Colitis/diet therapy , Colon/immunology , Dysbiosis/diet therapy , Fragaria/metabolism , Gastrointestinal Microbiome , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Colon/microbiology , Cytokines/immunology , Dextran Sulfate/adverse effects , Disease Models, Animal , Dysbiosis/immunology , Dysbiosis/metabolism , Dysbiosis/microbiology , Homeostasis , Humans , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...