Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Environ Sci Technol ; 58(21): 9272-9282, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38749055

ABSTRACT

Triclocarban (TCC), as a widely used antimicrobial agent, is accumulated in waste activated sludge at a high level and inhibits the subsequent anaerobic digestion of sludge. This study, for the first time, investigated the effectiveness of microbial electrolysis cell-assisted anaerobic digestion (MEC-AD) in mitigating the inhibition of TCC to methane production. Experimental results showed that 20 mg/L TCC inhibited sludge disintegration, hydrolysis, acidogenesis, and methanogenesis processes and finally reduced methane production from traditional sludge anaerobic digestion by 19.1%. Molecular docking revealed the potential inactivation of binding of TCC to key enzymes in these processes. However, MEC-AD with 0.6 and 0.8 V external voltages achieved much higher methane production and controlled the TCC inhibition to less than 5.8%. TCC in the MEC-AD systems was adsorbed by humic substances and degraded to dichlorocarbanilide, leading to a certain detoxification effect. Methanogenic activities were increased in MEC-AD systems, accompanied by complete VFA consumption. Moreover, the applied voltage promoted cell apoptosis and sludge disintegration to release biodegradable organics. Metagenomic analysis revealed that the applied voltage increased the resistance of electrode biofilms to TCC by enriching functional microorganisms (syntrophic VFA-oxidizing and electroactive bacteria and hydrogenotrophic methanogens), acidification and methanogenesis pathways, multidrug efflux pumps, and SOS response.


Subject(s)
Electrolysis , Anaerobiosis , Sewage/microbiology , Methane/metabolism , Carbanilides/pharmacology
2.
Microbiol Spectr ; 12(6): e0007124, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38700321

ABSTRACT

Novel antimicrobial agents are needed to combat antimicrobial resistance. This study tested novel pentafluorosulfanyl-containing triclocarban analogs for their potential antibacterial efficacy. Standard procedures were used to produce pentafluorosulfanyl-containing triclocarban analogs. Twenty new compounds were tested against seven Gram-positive and Gram-negative indicator strains as well as 10 clinical isolates for their antibacterial and antibiofilm activity. Mechanistic investigations focused on damage to cell membrane, oxidizing reduced thiols, iron-sulfur clusters, and oxidative stress to explain the compounds' activity. Safety profiles were assessed using cytotoxicity experiments in eukaryotic cell lines. Following screening, selected components had significantly better antibacterial and antibiofilm activity against Gram-positive bacteria in lower concentrations in comparison to ciprofloxacin and gentamycin. For instance, one compound had a minimum inhibitory concentration of <0.0003 mM, but ciprofloxacin had 0.08 mM. Mechanistic studies show that these novel compounds do not affect reduced thiol content, iron-sulfur clusters, or hydrogen peroxide pathways. Their impact comes from Gram-positive bacterial cell membrane damage. Tests on cell culture toxicity and host component safety showed promise. Novel diarylurea compounds show promise as Gram-positive antimicrobials. These compounds offer prospects for study and optimization. IMPORTANCE: The rise of antibiotic resistance among bacterial pathogens poses a significant threat to global health, underscoring the urgent need for novel antimicrobial agents. This study presents research on a promising class of novel compounds with potent antibacterial properties against Gram-positive bacteria, notably Staphylococcus aureus and MRSA. What sets these novel analogs apart is their superior efficacy at substantially lower concentrations compared with commonly used antibiotics like ciprofloxacin and gentamycin. Importantly, these compounds act by disrupting the bacterial cell membrane, offering a unique mechanism that could potentially circumvent existing resistance mechanisms. Preliminary safety assessments also highlight their potential for therapeutic use. This study not only opens new avenues for combating antibiotic-resistant infections but also underscores the importance of innovative chemical approaches in addressing the global antimicrobial resistance crisis.


Subject(s)
Anti-Bacterial Agents , Carbanilides , Gram-Positive Bacteria , Microbial Sensitivity Tests , Carbanilides/pharmacology , Carbanilides/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Gram-Positive Bacteria/drug effects , Humans , Biofilms/drug effects , Gram-Negative Bacteria/drug effects , Ciprofloxacin/pharmacology
3.
Appl Biochem Biotechnol ; 194(10): 4930-4945, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35674922

ABSTRACT

The most prevalent malignancy among women is breast cancer. Phytochemicals and their derivatives are rapidly being recognized as possible cancer complementary therapies because they can modify signaling pathways that lead to cell cycle control or directly alter cell cycle regulatory molecules. The phytochemicals' poor bioavailability and short half-life make them unsuitable as anticancer drugs. Applying PLGA-PEG NPs improves their solubility and tolerance while also reducing drug adverse effects. According to the findings, combining anti-tumor phytochemicals can be more effective in regulating several signaling pathways linked to tumor cell development. The point of the study was to compare the anti-proliferative impacts of combined artemisinin and metformin on cell cycle arrest and expression of cyclin D1 and apoptotic genes (bcl-2, Bax, survivin, caspase-7, and caspase-3), and also hTERT genes in breast cancer cells. T-47D breast cancer cells were treated with different concentrations of metformin (MET) and artemisinin (ART) co-loaded in PLGA-PEG NPs and free form. The MTT test was applied to assess drug cytotoxicity in T47D cells. The cell cycle distribution was investigated using flow cytometry and the expression levels of cyclin D1, hTERT, Bax, bcl-2, caspase-3, and caspase-7, and survivin genes were then determined using real-time PCR. The findings of the MTT test and flow cytometry revealed that each state was cytotoxic to T47D cells in a time and dose-dependent pattern. Compared to various state of drugs (free and nano state, pure and combination state) Met-Art-PLGA/PEG NPs demonstrated the strongest anti-proliferative impact and considerably inhibited the development of T-47D cells; also, treatment with nano-formulated forms of Met-Art combination resulted in substantial downregulation of hTERT, Bcl-2, cyclin D1, survivin, and upregulation of caspase-3, caspase-7, and Bax, in the cells, as compared to the free forms, as indicated by real-time PCR findings. The findings suggested that combining an ART/MET-loaded PLGA-PEG NP-based therapy for breast cancer could significantly improve treatment effectiveness.


Subject(s)
Alkylmercury Compounds , Antineoplastic Agents , Artemisinins , Breast Neoplasms , Carbanilides , Ethylmercury Compounds , Heterocyclic Compounds , Metformin , Nanoparticles , Trimethyltin Compounds , Antineoplastic Agents/chemistry , Apoptosis , Artemisinins/pharmacology , Artemisinins/therapeutic use , Benzalkonium Compounds/pharmacology , Benzalkonium Compounds/therapeutic use , Benzoflavones/pharmacology , Benzoflavones/therapeutic use , Breast Neoplasms/metabolism , Carbanilides/pharmacology , Carbanilides/therapeutic use , Caspase 3/genetics , Caspase 7 , Cell Line, Tumor , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin D1/pharmacology , Ethylmercury Compounds/pharmacology , Ethylmercury Compounds/therapeutic use , Female , Heterocyclic Compounds/pharmacology , Humans , Metformin/pharmacology , Metformin/therapeutic use , Methacholine Compounds , Nanoparticles/chemistry , Oximes/pharmacology , Oximes/therapeutic use , Plasmalogens/pharmacology , Plasmalogens/therapeutic use , Sulfonylurea Compounds/pharmacology , Sulfonylurea Compounds/therapeutic use , Survivin/pharmacology , Survivin/therapeutic use , Trimethyltin Compounds/pharmacology , bcl-2-Associated X Protein
4.
Acta Histochem ; 123(6): 151772, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34428603

ABSTRACT

Triclocarban (TCC), an antimicrobial compound commonly added to a wide range of household and personal hygiene care products, is one of the most prevalent endocrine-disrupting substances (EDS). This study was conducted to elucidate whether in utero and lactational exposure to TCC could adversely affect folliculogenesis and the onset of puberty in female rat offspring. Twenty pregnant Sprague Dawley rats were equally divided into Control and TCC dam groups (supplemented daily with drinking water enriched with 0.5 mg/L of TCC) from gestational day5 to postnatal day21 (PND21). Female offspring, 20 from control and 20 from TCC dams, were subdivided into 4 subgroups (PND21, PND28, PND35 & PND42). The day of vaginal opening and first estrous cycle were determined. Ovarian sections of the offspring were processed for H&E staining and for immunohistochemical expression of Ki67, Caspase-3 and androgen receptors (AR) on the granulosa cells of ovarian follicles. Follicular count and atretic index were assessed besides, serum estradiol, progesterone, FSH and LH, C-reactive protein (CRP), malondialdehyde (MDA) and total antioxidant capacity (TAC) were measured. TCC offspring exhibited a significant delay in the onset of puberty and impedance of normal transition of the primordial follicles to more developed ones with altered cyctoarchitecture. Also, TCC decreased follicular count, proliferation and gonado-somatic index while it increased atretic index, apoptosis and AR of the granulosa cells along with disturbance of the feminine hormonal profile and oxidant/antioxidant balance. This study highlighted the potential long-term consequences of in utero and lactational exposure to TCC on the postnatal development of the ovary in rat offspring.


Subject(s)
Carbanilides/adverse effects , Lactation/drug effects , Ovary/growth & development , Prenatal Exposure Delayed Effects/metabolism , Animals , Carbanilides/pharmacology , Female , Lactation/metabolism , Male , Ovary/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Sprague-Dawley
5.
Molecules ; 26(9)2021 May 10.
Article in English | MEDLINE | ID: mdl-34068616

ABSTRACT

In the late 1930s and early 1940s, it was discovered that the substitution on aromatic rings of hydrogen atoms with chlorine yielded a novel chemistry of antimicrobials. However, within a few years, many of these compounds and formulations showed adverse effects, including human toxicity, ecotoxicity, and unwanted environmental persistence and bioaccumulation, quickly leading to regulatory bans and phase-outs. Among these, the triclocarban, a polychlorinated aromatic antimicrobial agent, was employed as a major ingredient of toys, clothing, food packaging materials, food industry floors, medical supplies, and especially of personal care products, such as soaps, toothpaste, and shampoo. Triclocarban has been widely used for over 50 years, but only recently some concerns were raised about its endocrine disruptive properties. In September 2016, the U.S. Food and Drug Administration banned its use in over-the-counter hand and body washes because of its toxicity. The withdrawal of triclocarban has prompted the efforts to search for new antimicrobial compounds and several analogues of triclocarban have also been studied. In this review, an examination of different facets of triclocarban and its analogues will be analyzed.


Subject(s)
Carbanilides/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Biotransformation/drug effects , Carbanilides/chemistry , Carbanilides/toxicity , Ecotoxicology , Humans , Triclosan/chemistry , Triclosan/toxicity
6.
Toxicol In Vitro ; 69: 105000, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32942006

ABSTRACT

Targeted anti-tumor small molecules are considered to be promising candidates for cancer treatment. The novel diphenyl urea derivative (DUD) was synthesized by the molecular docking based on the structure optimization of Taspine (a natural product). In this study, we explored the anti-metastatic potential of DUD for NSCLC in vitro. DUD significantly suppressed A549 cell migration by reversing EMT. The inhibition was reflected on upregulation of E-cadherin and downregulation of N-cadherin, vimentin, Snail and HIF-1α. Meanwhile, DUD inhibited the ß-catenin nuclear translocation by upregulating Axin and downregulating the expression of APC, CK1 and phosphorylation of GSK3ß, and simultaneously decreasing MMP9 and MMP13 expression. Moreover, it was associated with the downregulation of the PI3K/Akt/mTOR signaling. Furthermore, we used XAV939, an ß-catenin inhibitor, to verify the mechanism of DUD. These results suggested that DUD inhibited A549 cells migration by reversing EMT via Wnt/ß-catenin and PI3K/Akt signaling. DUD might be a potential therapeutic drug candidate for NSCLC treatment.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Carbanilides/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Wnt Signaling Pathway/drug effects , Wound Healing/drug effects
7.
J Environ Sci Health B ; 55(11): 990-1001, 2020.
Article in English | MEDLINE | ID: mdl-32877275

ABSTRACT

Application of municipal biosolids in agriculture present a concern with potential uptake and bioaccumulation of pharmaceutical compounds from biosolids into agronomic plants. We evaluated the efficacy of biochar as a soil amendment to minimize uptake of antimicrobial agents (ciprofloxacin, triclocarban, and triclosan) in lettuce (Lactuca sativa) and carrot (Daucus carota) plants. Biochar reduced the concentration of ciprofloxacin and triclocarban in lettuce leaves and resulted in a 67% reduction of triclosan in carrot roots. There was no substantial difference in pharmaceutical concentrations in carrot and lettuce plant matter at low (2.0 g kg-1 soil) and high (20.4 g kg-1 soil) rates of applied biochar. The co-amendment of biochar and biosolids increased soil pH and nutrient content which were positively correlated with an increase in lettuce shoot biomass. Our results demonstrate the potential efficacy of using walnut shell biochar as a sorbent for pharmaceutical contaminants in soil without negatively affecting plant growth.


Subject(s)
Carbanilides/pharmacology , Charcoal , Ciprofloxacin/pharmacology , Daucus carota/drug effects , Lactuca/drug effects , Triclosan/pharmacokinetics , Agriculture/methods , Anti-Infective Agents/pharmacokinetics , Biomass , Biosolids , Daucus carota/growth & development , Daucus carota/metabolism , Lactuca/growth & development , Lactuca/metabolism , Plant Leaves/chemistry , Plant Leaves/drug effects , Plant Leaves/metabolism , Plant Roots/chemistry , Plant Roots/drug effects , Plant Roots/metabolism , Plant Shoots/drug effects , Soil/chemistry , Soil Pollutants/analysis , Soil Pollutants/pharmacokinetics
8.
Appl Environ Microbiol ; 86(16)2020 08 03.
Article in English | MEDLINE | ID: mdl-32503913

ABSTRACT

Triclocarban (TCC), a formerly used disinfectant, kills bacteria via an unknown mechanism of action. A structural hallmark is its N,N'-diaryl urea motif, which is also present in other antibiotics, including the recently reported small molecule PK150. We show here that, like PK150, TCC exhibits an inhibitory effect on Staphylococcus aureus menaquinone metabolism via inhibition of the biosynthesis protein demethylmenaquinone methyltransferase (MenG). However, the activity spectrum (MIC90) of TCC across a broad range of multidrug-resistant staphylococcus and enterococcus strains was much narrower than that of PK150. Accordingly, TCC did not cause an overactivation of signal peptidase SpsB, a hallmark of the PK150 mode of action. Furthermore, we were able to rule out inhibition of FabI, a confirmed target of the diaryl ether antibiotic triclosan (TCS). Differences in the target profiles of TCC and TCS were further investigated by proteomic analysis, showing complex but rather distinct changes in the protein expression profile of S. aureus Downregulation of the arginine deiminase pathway provided additional evidence for an effect on bacterial energy metabolism by TCC.IMPORTANCE TCC's widespread use as an antimicrobial agent has made it a ubiquitous environmental pollutant despite its withdrawal due to ecological and toxicological concerns. With its antibacterial mechanism of action still being unknown, we undertook a comparative target analysis between TCC, PK150 (a recently discovered antibacterial compound with structural resemblance to TCC), and TCS (another widely employed chlorinated biphenyl antimicrobial) in the bacterium Staphylococcus aureus We show that there are distinct differences in each compound's mode of action, but also identify a shared target between TCC and PK150, the interference with menaquinone metabolism by inhibition of MenG. The prevailing differences, however, which also manifest in a remarkably better broad-spectrum activity of PK150, suggest that even high levels of TCC or TCS resistance observed by continuous environmental exposure may not affect the potential of PK150 or related N,N'-diaryl urea compounds as new antibiotic drug candidates against multidrug-resistant infections.


Subject(s)
Bacterial Proteins/genetics , Carbanilides/pharmacology , Disinfectants/pharmacology , Enterococcus/drug effects , Methyltransferases/genetics , Staphylococcus aureus/drug effects , Bacterial Proteins/metabolism , Enterococcus/genetics , Enterococcus/metabolism , Methyltransferases/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
9.
J Cell Mol Med ; 24(1): 984-995, 2020 01.
Article in English | MEDLINE | ID: mdl-31742861

ABSTRACT

IL-2R pathway is a key regulator in the development of immune cells and has emerged as a promising drug target in cancer treatment, but there is a scarcity of related inhibitors. TPD7 is a novel biphenyl urea taspine derivate, which has been shown anti-cancer effect. Here, we demonstrated the anti-cancer activity of TPD7 in cutaneous T cell lymphoma and investigated the underlying mechanism of TPD7 through IL-2R signalling. The inhibitory effect of TPD7 on cell viability exhibited a strong correlation with the expression level of IL-2R, and cutaneous T cell lymphoma H9 and HUT78 cells were most sensitive to TPD7. TPD7 was nicely bound to IL-2R and down-regulated the mRNA and protein levels of IL-2R. Furthermore, TPD7 suppressed the downstream cascades of IL-2R including JAK/STAT, PI3K/AKT/mTOR and PLCγ/Raf/MAPK signalling, resulting in Bcl-2 mitochondrial apoptosis pathway and cell cycle proteins CDK/Cyclins regulation. And, these were verified by flow cytometry analysis that TPD7 facilitated cell apoptosis in H9 cells via mitochondrial pathway and impeded cell cycle progression at G2/M phase. TPD7 is a novel anti-cancer agent and may be a potential candidate for cutaneous T cell lymphoma treatment by regulating IL-2R signalling pathway.


Subject(s)
Biomarkers, Tumor/metabolism , Carbanilides/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hydroxylamines/pharmacology , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphoma, T-Cell, Cutaneous/drug therapy , Skin Neoplasms/drug therapy , Apoptosis , Biomarkers, Tumor/genetics , Cell Cycle , Cell Movement , Cell Proliferation , Gene Expression Profiling , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Lymphoma, T-Cell, Cutaneous/genetics , Lymphoma, T-Cell, Cutaneous/metabolism , Lymphoma, T-Cell, Cutaneous/pathology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Cells, Cultured
10.
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
11.
J Biochem Mol Toxicol ; 33(5): e22289, 2019 May.
Article in English | MEDLINE | ID: mdl-30657620

ABSTRACT

Triclocarban (TCC), which is used as an antimicrobial agent in personal care products, has been widely detected in aquatic ecosystems. However, the consequence of TCC exposure on embryo development is still elusive. Here, by using zebrafish embryos, we aimed to understand the developmental defects caused by TCC exposure. After exposure to 0.3, 30, and 300 µg/L TCC from 4-hour postfertilization (hpf) to 120 hpf, we observed that TCC exposure significantly increased the mortality and malformation, delayed hatching, and reduced body length. Exposure to TCC also affected the heart rate and expressions of cardiac development-related genes in zebrafish embryos. In addition, TCC exposure altered the expressions of the genes involved in hormonal pathways, indicating its endocrine disrupting effects. In sum, our data highlight the impact of TCC on embryo development and its interference with the hormone system of zebrafish.


Subject(s)
Anti-Infective Agents/adverse effects , Carbanilides/adverse effects , Embryo, Nonmammalian/embryology , Embryonic Development/drug effects , Endocrine Disruptors/adverse effects , Water Pollutants, Chemical/adverse effects , Zebrafish/embryology , Animals , Anti-Infective Agents/pharmacology , Carbanilides/pharmacology , Embryo, Nonmammalian/pathology , Endocrine Disruptors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Water Pollutants, Chemical/pharmacology
12.
J Mol Recognit ; 32(1): e2755, 2019 01.
Article in English | MEDLINE | ID: mdl-30033524

ABSTRACT

The variable VHH domains of camelid single chain antibodies have been useful in numerous biotechnology applications due to their simplicity, biophysical properties, and abilities to bind to their cognate antigens with high affinities and specificity. Their interactions with proteins have been well-studied, but considerably less work has been done to characterize their ability to bind haptens. A high-resolution structural study of three nanobodies (T4, T9, and T10) which have been shown to bind triclocarban (TCC, 3-(4-chlorophenyl)-1-(3,4-dichlorophenyl)urea) with near-nanomolar affinity shows that binding occurs in a tunnel largely formed by CDR1 rather than a surface or lateral binding mode seen in other nanobody-hapten interactions. Additional significant interactions are formed with a non-hypervariable loop, sometimes dubbed "CDR4". A comparison of apo and holo forms of T9 and T10 shows that the binding site undergoes little conformational change upon binding of TCC. Structures of three nanobody-TCC complexes demonstrated there was not a standard binding mode. T4 and T9 have a high degree of sequence identity and bind the hapten in a nearly identical manner, while the more divergent T10 binds TCC in a slightly displaced orientation with the urea moiety rotated approximately 180° along the long axis of the molecule. In addition to methotrexate, this is the second report of haptens binding in a tunnel formed by CDR1, suggesting that compounds with similar hydrophobicity and shape could be recognized by nanobodies in analogous fashion. Structure-guided mutations failed to improve binding affinity for T4 and T9 underscoring the high degree of natural optimization.


Subject(s)
Carbanilides/pharmacology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Animals , Antibody Specificity , Binding Sites , Camelus , Carbanilides/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Conformation , Protein Domains , Single-Domain Antibodies/genetics
13.
Molecules ; 23(11)2018 Nov 02.
Article in English | MEDLINE | ID: mdl-30400165

ABSTRACT

Concerns have been raised about the long-term accumulating effects of triclocarban, a polychlorinated diarylurea widely used as an antibacterial soap additive, in the environment and in human beings. Indeed, the Food and Drug Administration has recently banned it from personal care products. Herein, we report the synthesis, antibacterial activity and cytotoxicity of novel N,N'-diarylureas as triclocarban analogs, designed by reducing one or more chlorine atoms of the former and/or replacing them by the novel pentafluorosulfanyl group, a new bioisostere of the trifluoromethyl group, with growing importance in drug discovery. Interestingly, some of these pentafluorosulfanyl-bearing ureas exhibited high potency, broad spectrum of antimicrobial activity against Gram-positive bacterial pathogens, and high selectivity index, while displaying a lower spontaneous mutation frequency than triclocarban. Some lines of evidence suggest a bactericidal mode of action for this family of compounds.


Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Carbanilides/chemistry , Carbanilides/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Catheters/microbiology , Humans , Microbial Sensitivity Tests , Molecular Structure , Mutation Rate , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Structure-Activity Relationship
14.
Chemosphere ; 210: 93-101, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29986228

ABSTRACT

Triclocarban (TCC) is used in many household and personal hygiene products. TCC has been widely detected in wastewater around the world. The present study reveals that TCC can activate oxidative stress, induce total antioxidant capacity expression and lipid peroxidation, and increase the activities of superoxide dismutase and other antioxidant enzymes to resist oxidative damage. A significant induction of concentrations of proinflammatory mediator and nitric oxide (NO), accompanied by an upregulated expression of inducible NO synthase gene, was detected in zebrafish embryos exposed to TCC. The transcription of immune-response-related genes, including tnf-α, il-1ß, il-4, il-8, and cxcl-clc, was significantly upregulated on exposure to TCC. Furthermore, we found that the exposure of zebrafish embryos to TCC decreased immune cell recruiting in the head. Expressions of nf-κb, trif, myd88, irak4, and traf6 were altered on exposure to TCC. These results demonstrated that exposure to TCC at environmental concentrations significantly affects the expression of immune-response-related genes in zebrafish embryos following oxidative stress and the release of proinflammatory mediators through Toll-like receptor signaling pathway. Thus, we assumed that the ecological risk of TCC on aquatic organisms could not be ignored.


Subject(s)
Carbanilides/pharmacology , Immunity, Innate/drug effects , Oxidative Stress/drug effects , Zebrafish/metabolism , Animals , Antioxidants/metabolism , Aquatic Organisms/drug effects , Aquatic Organisms/metabolism , Carbanilides/toxicity , Immunity, Innate/genetics , Toll-Like Receptors/metabolism , Water Pollutants, Chemical/pharmacology , Zebrafish/embryology , Zebrafish/immunology
15.
Eur J Med Chem ; 155: 364-380, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29902722

ABSTRACT

Tuberculosis is the second leading cause of deaths worldwide. The inadequacy of existing drugs to treat TB due to developed resistance and TB-HIV synergism urges for new anti-TB drugs. Seventy-two benzo[d]thiazole-2-carbanilides have been synthesized through CDI-mediated direct coupling of benzo[d]thiazole-2-carboxylic acids with aromatic amines using a three step methodology which includes a green protocol for synthesis of ethyl benzo[d]thiazole-2-carboxylates, precursor of the desired carboxylic acids. The compounds were evaluated in vitro for anti-tubercular activity against M. tuberculosis H37Rv (ATCC27294 strain). Thirty-two compounds exhibiting MIC values in the range of 0.78-6.25 µg/mL (1.9-23 µM) were subjected to cell viability test against RAW 264.7 cell lines and thirty compounds were found to be non-toxic (<50% inhibition). The most active compounds with MIC of 0.78 µg/mL (e.g., 4i, 4n, 4s, 4w, 6f, 6h, 6u, 7e, 7h, 7p, 7r and 7w) exhibit therapeutic index of 64. The structure activity relationship of the N-arylbenzo[d]thiazole-2-carboxamides has been established for anti-mycobacterial activity. Molecular docking suggests that the compounds 7w, 4i and 4n bind to the catalytic site of the enzyme ATP Phosphoribosyltransferase (HisG) and might be attributed to their anti-TB potential. These can serve as a new starting point for the development of anti-TB agents with therapeutic potential.


Subject(s)
Antitubercular Agents/pharmacology , Carbanilides/pharmacology , Drug Design , Mycobacterium tuberculosis/drug effects , Thiazoles/pharmacology , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Carbanilides/chemical synthesis , Carbanilides/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Mice , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , RAW 264.7 Cells , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
16.
Oncol Rep ; 40(2): 1064-1072, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29901176

ABSTRACT

We recently reported that TPD7 suppressed tumor cell proliferation, and inhibited invasion, through the suppression of C-X-C chemokine receptor type 4 (CXCR4). In the present study, we investigated the anticancer effect of TPD7 on apoptosis and invasion of cervical cancer HeLa cells. Cell cycle analysis revealed that TPD7 decreased cyclin-dependent kinase (CDK)1 and cyclin D1 expression, and increased cyclin A expression, following S phase blockade. TPD7 induced chromatin condensation and significantly elevated the number of apoptotic cells, suggesting that its inhibitory effect on HeLa cells was due to the induction of cell cycle blockade and apoptosis. Mechanistically, TPD7 altered the extrinsic apoptosis pathway by upregulating Fas expression, and the intrinsic pathway by modulating Bcl-2 family proteins, p53, and NF-κB p65, leading to enhanced apoptosis. TPD7 inhibited HeLa cell invasion by downregulating the expression of matrix metalloproteinase (MMP)-9 and CXCR4 proteins. In vivo experiments revealed that TPD7 inhibited tumor growth in HeLa cell xenografted mice. These findings indicated that TPD7 may be a potential chemoprevention agent for the management of cervical carcinoma.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Carbanilides/pharmacology , Hydroxylamines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Urea/pharmacology , Uterine Cervical Neoplasms/drug therapy , fas Receptor/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Down-Regulation/drug effects , Female , HeLa Cells , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Receptors, CXCR4/metabolism , S Phase/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects , Uterine Cervical Neoplasms/metabolism
17.
Malar J ; 17(1): 121, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29558913

ABSTRACT

BACKGROUND: The increased resistance of the human malaria parasite Plasmodium falciparum to currently employed drugs creates an urgent call for novel anti-malarial drugs. Particularly, efforts should be devoted to developing fast-acting anti-malarial compounds in case clinical resistance increases to the first-line artemisinin-based combination therapy. SC83288, an amicarbalide derivative, is a clinical development candidate for the treatment of severe malaria. SC83288 is fast-acting and able to clear P. falciparum parasites at low nanomolar concentrations in vitro, as well as in a humanized SCID mouse model system in vivo. In this study, the antiplasmodial activity of SC83288 against artemisinins was profiled in order to assess its potential to replace, or be combined with, artemisinin derivatives. RESULTS: Based on growth inhibition and ring survival assays, no cross-resistance was observed between artemisinins and SC83288, using parasite lines that were resistant to either one of these drugs. In addition, no synergistic or antagonistic interaction was observed between the two drugs. This study further confirmed that SC83288 is a fast acting drug in several independent assays. Combinations of SC83288 and artesunate maintained the rapid parasite killing activities of both components. CONCLUSION: The results obtained in this study are consistent with artemisinins and SC83288 having distinct modes of action and different mechanisms of resistance. This study further supports efforts to continue the clinical development of SC83288 against severe malaria as an alternative to artemisinins in areas critically affected by artemisinin-resistance. Considering its fast antiplasmodial activity, SC83288 could be combined with a slow-acting anti-malarial drug.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Carbanilides/pharmacology , Plasmodium falciparum/drug effects , Animals , Antimalarials/administration & dosage , Antimalarials/pharmacokinetics , Artemisinins/administration & dosage , Artemisinins/pharmacokinetics , Carbanilides/administration & dosage , Carbanilides/pharmacokinetics , Drug Interactions , Drug Resistance , Molecular Structure
18.
Water Res ; 127: 150-161, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29045805

ABSTRACT

Triclocarban (TCC), one typical antibacterial agent being widely used in various applications, was found to be present in waste activated sludge at significant levels. To date, however, its effect on anaerobic fermentation of sludge has not been investigated. This work therefore aims to fill this knowledge gap. Experimental results showed that when TCC content in sludge increased from 26.7 ± 5.3 to 520.5 ± 12.6 mg per kilogram total suspended solids, the maximum concentration of short-chain fatty acids (SCFA) increased from 32.6 ± 2.5 to 228.2 ± 3.6 (without pH control) and from 211.7 ± 2.4 to 378.3 ± 3.2 mg COD/g VSS (initial pH 10), respectively. The large promotion of acetic acid was found to be the major reason for the enhancement of total SCFA production. Although a significant level of TCC was degraded in the fermentation process, SCFA was neither produced from TCC nor affected by its major intermediates at the relevant levels. It was found that TCC facilitated solubilization, acidogenesis, acetogenesis, and homoacetogenesis processes but inhibited methanogenesis process. Microbial analysis revealed that the increase of TCC increased the microbial community diversity, the abundances of SCFA (especially acetic acid) producers, and the activities of key enzymes relevant to acetic acid production.


Subject(s)
Carbanilides/pharmacology , Fatty Acids, Volatile/biosynthesis , Microbiota/drug effects , Sewage/microbiology , Acetic Acid/metabolism , Anaerobiosis , Biotechnology/methods , Carbanilides/metabolism , Fermentation , Hydrogen-Ion Concentration , Solubility , Waste Disposal, Fluid/methods
19.
EMBO Mol Med ; 9(12): 1732-1741, 2017 12.
Article in English | MEDLINE | ID: mdl-29030459

ABSTRACT

In 2016, the US Food and Drug Administration banned the use of specific microbicides in some household and personal wash products due to concerns that these chemicals might induce antibiotic resistance or disrupt human microbial communities. Triclosan and triclocarban (referred to as TCs) are the most common antimicrobials in household and personal care products, but the extent to which TC exposure perturbs microbial communities in humans, particularly during infant development, was unknown. We conducted a randomized intervention of TC-containing household and personal care products during the first year following birth to characterize whether TC exposure from wash products perturbs microbial communities in mothers and their infants. Longitudinal survey of the gut microbiota using 16S ribosomal RNA amplicon sequencing showed that TC exposure from wash products did not induce global reconstruction or loss of microbial diversity of either infant or maternal gut microbiotas. Broadly antibiotic-resistant species from the phylum Proteobacteria, however, were enriched in stool samples from mothers in TC households after the introduction of triclosan-containing toothpaste. When compared by urinary triclosan level, agnostic to treatment arm, infants with higher triclosan levels also showed an enrichment of Proteobacteria species. Despite the minimal effects of TC exposure from wash products on the gut microbial community of infants and adults, detected taxonomic differences highlight the need for consumer safety testing of antimicrobial self-care products on the human microbiome and on antibiotic resistance.


Subject(s)
Anti-Infective Agents/pharmacology , Carbanilides/pharmacology , Gastrointestinal Microbiome/drug effects , Triclosan/pharmacology , Adult , Bacteria/genetics , Bacteria/isolation & purification , Biodiversity , Chromatography, High Pressure Liquid , DNA, Bacterial/chemistry , DNA, Bacterial/isolation & purification , DNA, Bacterial/metabolism , Disinfectants/chemistry , Drug Resistance, Bacterial , Female , Humans , Infant , Liquid-Liquid Extraction , Longitudinal Studies , Proteobacteria/genetics , Proteobacteria/growth & development , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/metabolism , Sequence Analysis, DNA , Toothpastes/chemistry , Triclosan/isolation & purification , Triclosan/urine
20.
PLoS One ; 12(8): e0182821, 2017.
Article in English | MEDLINE | ID: mdl-28797064

ABSTRACT

Bacterial resistance to antibiotics remains an imposing global public health challenge. Of the most serious pathogens, methicillin-resistant Staphylococcus aureus (MRSA) is problematic given strains have emerged that exhibit resistance to several antibiotic classes including ß-lactams and agents of last resort such as vancomycin. New antibacterial agents composed of unique chemical scaffolds are needed to counter this public health challenge. The present study examines two synthetic diphenylurea compounds 1 and 2 that inhibit growth of clinically-relevant isolates of MRSA at concentrations as low as 4 µg/mL and are non-toxic to human colorectal cells at concentrations up to 128 µg/mL. Both compounds exhibit rapid bactericidal activity, completely eliminating a high inoculum of MRSA within four hours. MRSA mutants exhibiting resistance to 1 and 2 could not be isolated, indicating a low likelihood of rapid resistance emerging to these compounds. Bacterial cytological profiling revealed the diphenylureas exert their antibacterial activity by targeting bacterial cell wall synthesis. Both compounds demonstrate the ability to resensitize vancomycin-resistant Staphylococcus aureus to the effect of vancomycin. The present study lays the foundation for further investigation and development of diphenylurea compounds as a new class of antibacterial agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbanilides/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Anti-Bacterial Agents/therapeutic use , Carbanilides/therapeutic use , Humans , Methicillin/pharmacology , Methicillin/therapeutic use , Methicillin Resistance/drug effects , Microbial Sensitivity Tests , Vancomycin/pharmacology , Vancomycin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL