Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Food Chem ; 341(Pt 1): 128237, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33091666

ABSTRACT

This study investigated the stability of dicarbonyl compounds (DCs), 3-deoxyglucosone (3-DG), glyoxal (GO) and methylglyoxal (MGO) during simulated gastrointestinal digestion processes and the impact these compounds have on the gut microbiota. DCs pass almost unaltered through the in-vitro gastrointestinal digestion phases (concentration loss: 11% for 3-DG, 24% for GO and MGO) and have an effect on the fermentative digestion process, reducing the total gut bacterial population up to 6 Log10 units. Previous studies have shown no antimicrobial activity for 3-DG, however, for the first time it has been shown that when incubated with faecal bacteria 3-DG strongly depressed this microbial community. The influence of dicarbonyl compounds on the anaerobic fermentation processes was confirmed by the reduced production of short-chain fatty acids. Considering the modern Western diet, characterised by high consumption of ultra-processed foods rich in dicarbonyl compounds, this could lead to a reduction of bacteria important for the microbiome.


Subject(s)
Aldehydes/chemistry , Aldehydes/pharmacology , Digestion , Fermentation , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/metabolism , Stomach/physiology , Aldehydes/metabolism , Humans , Intestinal Mucosa/microbiology , Stomach/microbiology
2.
PLoS One ; 10(6): e0131241, 2015.
Article in English | MEDLINE | ID: mdl-26107737

ABSTRACT

Blocking the enzyme Fatty Acid Synthase (FASN) leads to apoptosis of HER2-positive breast carcinoma cells. The hypothesis is that blocking FASN, in combination with anti-HER2 signaling agents, would be an effective antitumor strategy in preclinical HER2+ breast cancer models of trastuzumab and lapatinib resistance. We developed and molecularly characterized in vitro HER2+ models of resistance to trastuzumab (SKTR), lapatinib (SKLR) and both (SKLTR). The cellular interactions of combining anti-FASN polyphenolic compounds (EGCG and the synthetic G28UCM) with anti-HER2 signaling drugs (trastuzumab plus pertuzumab and temsirolimus) were analyzed. Tumor growth inhibition after treatment with EGCG, pertuzumab, temsirolimus or the combination was evaluated in two in vivo orthoxenopatients: one derived from a HER2+ patient and another from a patient who relapsed on trastuzumab and lapatinib-based therapy. SKTR, SKLR and SKLTR showed hyperactivation of EGFR and p-ERK1/2 and PI3KCA mutations. Dual-resistant cells (SKLTR) also showed hyperactivation of HER4 and recovered levels of p-AKT compared with mono-resistant cells. mTOR, p-mTOR and FASN expression remained stable in SKTR, SKLR and SKLTR. In vitro, anti-FASN compounds plus pertuzumab showed synergistic interactions in lapatinib- and dual- resistant cells and improved the results of pertuzumab plus trastuzumab co-treatment. FASN inhibitors combined with temsirolimus displayed the strongest synergistic interactions in resistant cells. In vivo, both orthoxenopatients showed strong response to the antitumor activity of the combination of EGCG with pertuzumab or temsirolimus, without signs of toxicity. We showed that the simultaneous blockade of FASN and HER2 pathways is effective in cells and in breast cancer models refractory to anti-HER2 therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Fatty Acid Synthase, Type I/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Adhesion , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Humans , Lapatinib , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Naphthalenes/pharmacology , Neoplasm Invasiveness , Neoplasm Transplantation , Quinazolines/pharmacology , Signal Transduction/drug effects , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Trastuzumab/pharmacology
3.
PLoS One ; 10(4): e0123122, 2015.
Article in English | MEDLINE | ID: mdl-25855977

ABSTRACT

Neuropathic pain is common in peripheral nerve injury and often fails to respond to ordinary medication. Here, we investigated whether the two novel epigallocatechin-3-gallate (EGCG) polyphenolic derivatives, compound 23 and 30, reduce the neuropathic pain in mice chronic constriction nerve injury (CCI). First, we performed a dose-response study to evaluate nociceptive sensation after administration of EGCG and its derivatives 23 and 30, using the Hargreaves test at 7 and 21 days after injury (dpi). We daily administered EGCG, 23 and 30 (10 to 100 mg/Kg; i.p.) during the first week post-CCI. None of the doses of compound 23 caused significant pain diminution, whereas 50mg/kg was optimal for both EGCG and 30 to delay the latency of paw withdrawal. With 50 mg/Kg, we showed that EGCC prevented the thermal hyperalgesia from 7 to 21 dpi and compound 30 from 14 to 56 dpi. To evaluate the molecular mechanisms underpinning why EGCG and compound 30 differentially prevented the thermal hyperalgesia, we studied several biochemical parameters in the dorsal horn of the spinal cord at 14 and 56 dpi. We showed that the effect observed with EGCG and compound 30 was related to the inhibition of fatty acid synthase (FASN), a known target of these polyphenolic compounds. Additionally, we observed that EGCG and compound 30 reduced the expression of CCI-mediated inflammatory proteins and the nuclear localization of nuclear factor-kappa B at 14 dpi, but not at 56 dpi. We also strongly detected a decrease of synaptic plasma membrane levels of N-methyl-D-asparte receptor 2B in CCI-mice treated with compound 30 at 56 dpi. Altogether, compound 30 reduced the chronic thermal hyperalgesia induced by CCI better than the natural compound EGCG. Thus, our findings provide a rationale for the preclinical development of compound 30 as an agent to treat neuropathic pain.


Subject(s)
Catechin/analogs & derivatives , Neuralgia/drug therapy , Pain/drug therapy , Spinal Cord Injuries/drug therapy , Animals , Catechin/administration & dosage , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytokines/biosynthesis , Cytokines/metabolism , Fatty Acid Synthase, Type I/biosynthesis , Fatty Acid Synthase, Type I/metabolism , Fatty Acids/metabolism , Gene Expression Regulation/drug effects , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Male , Mice , Neuralgia/metabolism , Neuralgia/physiopathology , Pain/metabolism , Pain/physiopathology , Pain Threshold , Peripheral Nerve Injuries , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology
4.
ACS Chem Biol ; 10(3): 834-43, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25486266

ABSTRACT

Essential cell division protein FtsZ is considered an attractive target in the search for antibacterials with novel mechanisms of action to overcome the resistance problem. FtsZ undergoes GTP-dependent assembly at midcell to form the Z-ring, a dynamic structure that evolves until final constriction of the cell. Therefore, molecules able to inhibit its activity will eventually disrupt bacterial viability. In this work, we report a new series of small molecules able to replace GTP and to specifically inhibit FtsZ, blocking the bacterial division process. These new synthesized inhibitors interact with the GTP-binding site of FtsZ (Kd = 0.4-0.8 µM), display antibacterial activity against Gram-positive pathogenic bacteria, and show selectivity against tubulin. Biphenyl derivative 28 stands out as a potent FtsZ inhibitor (Kd = 0.5 µM) with high antibacterial activity [MIC (MRSA) = 7 µM]. In-depth analysis of the mechanism of action of compounds 22, 28, 33, and 36 has revealed that they act as effective inhibitors of correct FtsZ assembly, blocking bacterial division and thus leading to filamentous undivided cells. These findings provide a compelling rationale for the development of compounds targeting the GTP-binding site as antibacterial agents and open the door to antibiotics with novel mechanisms of action.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Bacillus subtilis/drug effects , Bacterial Proteins/antagonists & inhibitors , Biphenyl Compounds/chemical synthesis , Cytoskeletal Proteins/antagonists & inhibitors , Guanosine Triphosphate/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Naphthalenes/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/chemistry , Bacillus subtilis/growth & development , Bacterial Proteins/chemistry , Binding Sites , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Cytoskeletal Proteins/chemistry , Kinetics , Methicillin-Resistant Staphylococcus aureus/chemistry , Methicillin-Resistant Staphylococcus aureus/growth & development , Microbial Sensitivity Tests , Models, Molecular , Naphthalenes/chemistry , Naphthalenes/pharmacology , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
5.
J Med Chem ; 55(11): 5013-23, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22559865

ABSTRACT

Fatty acid synthase (FASN) is a lipogenic enzyme that is highly expressed in different human cancers. Here we report the development of a new series of polyphenolic compounds 5-30 that have been evaluated for their cytotoxic capacity in SK-Br3 cells, a human breast cancer cell line with high FASN expression. The compounds with an IC(50) < 50 µM have been tested for their ability to inhibit FASN activity. Among them, derivative 30 blocks the 90% of FASN activity at low concentration (4 µM), is highly cytotoxic in a broad panel of tumor cells, induces apoptosis, and blocks the activation of HER2, AKT, and ERK pathways. Remarkably, 30 does not activate carnitine palmitoyltransferase-1 (CPT-1) nor induces in mice weight loss, which are the main drawbacks of other previously described FASN inhibitors. Thus, FASN inhibitor 30 may aid the validation of this enzyme as a therapeutic target for the treatment of cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Fatty Acid Synthases/antagonists & inhibitors , Polyphenols/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Cell Line, Tumor , Drug Screening Assays, Antitumor , Enzyme Activation , Humans , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Polyphenols/chemistry , Polyphenols/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Weight Loss/drug effects
6.
Breast Cancer Res ; 13(6): R131, 2011.
Article in English | MEDLINE | ID: mdl-22177475

ABSTRACT

INTRODUCTION: Inhibiting the enzyme Fatty Acid Synthase (FASN) leads to apoptosis of breast carcinoma cells, and this is linked to human epidermal growth factor receptor 2 (HER2) signaling pathways in models of simultaneous expression of FASN and HER2. METHODS: In a xenograft model of breast carcinoma cells that are FASN+ and HER2+, we have characterised the anticancer activity and the toxicity profile of G28UCM, the lead compound of a novel family of synthetic FASN inhibitors. In vitro, we analysed the cellular and molecular interactions of combining G28UCM with anti-HER drugs. Finally, we tested the cytotoxic ability of G28UCM on breast cancer cells resistant to trastuzumab or lapatinib, that we developed in our laboratory. RESULTS: In vivo, G28UCM reduced the size of 5 out of 14 established xenografts. In the responding tumours, we observed inhibition of FASN activity, cleavage of poly-ADPribose polymerase (PARP) and a decrease of p-HER2, p- protein kinase B (AKT) and p-ERK1/2, which were not observed in the nonresponding tumours. In the G28UCM-treated animals, no significant toxicities occurred, and weight loss was not observed. In vitro, G28UCM showed marked synergistic interactions with trastuzumab, lapatinib, erlotinib or gefitinib (but not with cetuximab), which correlated with increases in apoptosis and with decreases in the activation of HER2, extracellular signal-regulated kinase (ERK)1/2 and AKT. In trastuzumab-resistant and in lapatinib-resistant breast cancer cells, in which trastuzumab and lapatinib were not effective, G28UCM retained the anticancer activity observed in the parental cells. CONCLUSIONS: G28UCM inhibits fatty acid synthase (FASN) activity and the growth of breast carcinoma xenografts in vivo, and is active in cells with acquired resistance to anti-HER2 drugs, which make it a candidate for further pre-clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Naphthalenes/pharmacology , Receptor, ErbB-2/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Female , Gallic Acid/administration & dosage , Gallic Acid/pharmacology , Gallic Acid/toxicity , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Naphthalenes/administration & dosage , Naphthalenes/toxicity , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 15(24): 7608-7615, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20008854

ABSTRACT

PURPOSE: Fatty acid synthase (FASN) is overexpressed in human breast carcinoma. The natural polyphenol (-)-epigallocatechin-3-gallate blocks in vitro FASN activity and leads to apoptosis in breast cancer cells without any effects on carnitine palmitoyltransferase-1 (CPT-1) activity, and in vivo, does not decrease body weight. We synthesized a panel of new polyphenolic compounds and tested their effects on breast cancer models. EXPERIMENTAL DESIGN: We evaluated the in vitro effects of the compounds on breast cancer cell growth (SK-Br3, MCF-7, and MDA-MB-231), apoptosis [as assessed by cleavage of poly(ADP-ribose) polymerase], cell signaling (HER2, ERK1/2, and AKT), and fatty acid metabolism enzymes (FASN and CPT-1). In vivo, we have evaluated their antitumor activity and their effect on body weight in a mice model of BT474 breast cancer cells. RESULTS: Two compounds potently inhibited FASN activity and showed high cytotoxicity. Moreover, the compounds induced apoptosis and caused a marked decrease in the active forms of HER2, AKT, and ERK1/2 proteins. Interestingly, the compounds did not stimulate CPT-1 activity in vitro. We show evidence that one of the FASN inhibitors blocked the growth of BT474 breast cancer xenografts and did not induce weight loss in vivo. CONCLUSIONS: The synthesized polyphenolic compounds represent a novel class of FASN inhibitors, with in vitro and in vivo anticancer activity, that do not exhibit cross-activation of beta-oxidation and do not induce weight loss in animals. One of the compounds blocked the growth of breast cancer xenografts. These FASN inhibitors may represent new agents for breast cancer treatment. (Clin Cancer Res 2009;15(24):7608-15).

SELECTION OF CITATIONS
SEARCH DETAIL
...