Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Gut Microbes ; 16(1): 2337312, 2024.
Article En | MEDLINE | ID: mdl-38591915

Clostridioides difficile causes a range of debilitating intestinal symptoms that may be fatal. It is particularly problematic as a hospital-acquired infection, causing significant costs to the health care system. Antibiotics, such as vancomycin and fidaxomicin, are still the drugs of choice for C. difficile infections, but their effectiveness is limited, and microbial interventions are emerging as a new treatment option. This paper focuses on alternative treatment approaches, which are currently in various stages of development and can be divided into four therapeutic strategies. Direct killing of C. difficile (i) includes beside established antibiotics, less studied bacteriophages, and their derivatives, such as endolysins and tailocins. Restoration of microbiota composition and function (ii) is achieved with fecal microbiota transplantation, which has recently been approved, with standardized defined microbial mixtures, and with probiotics, which have been administered with moderate success. Prevention of deleterious effects of antibiotics on microbiota is achieved with agents for the neutralization of antibiotics that act in the gut and are nearing regulatory approval. Neutralization of C. difficile toxins (iii) which are crucial virulence factors is achieved with antibodies/antibody fragments or alternative binding proteins. Of these, the monoclonal antibody bezlotoxumab is already in clinical use. Immunomodulation (iv) can help eliminate or prevent C. difficile infection by interfering with cytokine signaling. Small-molecule agents without bacteriolytic activity are usually selected by drug repurposing and can act via a variety of mechanisms. The multiple treatment options described in this article provide optimism for the future treatment of C. difficile infection.


Clostridioides difficile , Clostridium Infections , Gastrointestinal Microbiome , Humans , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Fecal Microbiota Transplantation , Vancomycin/pharmacology , Clostridium Infections/drug therapy , Clostridium Infections/prevention & control
2.
Cell Death Dis ; 13(10): 860, 2022 10 08.
Article En | MEDLINE | ID: mdl-36209148

Chronic lymphocytic leukemia (CLL) is a hematological neoplasm of CD19-positive mature-appearing B lymphocytes. Despite the clinical success of targeted therapies in CLL, the development of resistance diminishes their therapeutic activity. This is also true for the Bcl-2 antagonist venetoclax. We investigated the molecular mechanisms that drive venetoclax resistance in CLL, with a clear focus to provide new strategies to successfully combat it. Activation of CLL cells with IFNγ, PMA/ionomycin, and sCD40L diminished the cytotoxicity of venetoclax. We demonstrated that the metabolic activity of cells treated with 1 nM venetoclax alone was 48% of untreated cells, and was higher for cells co-treated with IFNγ (110%), PMA/ionomycin (78%), and sCD40L (62%). As of molecular mechanism, we showed that PMA/ionomycin and sCD40L triggered translocation of NFκB in primary CLL cells, while IFNγ activated p38 MAPK, suppressed spontaneous and venetoclax-induced apoptosis and induced formation of the immunoproteasome. Inhibition of immunoproteasome with ONX-0914 suppressed activity of immunoproteasome and synergized with venetoclax against primary CLL cells. On the other hand, inhibition of p38 MAPK abolished cytoprotective effects of IFNγ. We demonstrated that venetoclax-resistant (MEC-1 VER) cells overexpressed p38 MAPK and p-Bcl-2 (Ser70), and underexpressed Mcl-1, Bax, and Bak. Inhibition of p38 MAPK or immunoproteasome triggered apoptosis in CLL cells and overcame the resistance to venetoclax of MEC-1 VER cells and venetoclax-insensitive primary CLL cells. In conclusion, the p38 MAPK pathway and immunoproteasome represent novel targets to combat venetoclax resistance in CLL.


Antineoplastic Agents , Leukemia, Lymphocytic, Chronic, B-Cell , Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Drug Resistance, Neoplasm , Humans , Ionomycin/pharmacology , Ionomycin/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides , bcl-2-Associated X Protein , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Sci Rep ; 11(1): 21354, 2021 11 01.
Article En | MEDLINE | ID: mdl-34725457

Anchorage-independent growth of cancer cells in vitro is correlated to metastasis formation in vivo. Metformin use is associated with decreased breast cancer incidence and currently evaluated in cancer clinical trials. The combined treatment with metformin and 2-deoxy-D-glucose (2DG) in vitro induces detachment of viable MDA-MB-231 breast cancer cells that retain their proliferation capacity. This might be important for cell detachment from primary tumors, but the metabolic changes involved are unknown. We performed LC/MS metabolic profiling on separated attached and detached MDA-MB-231 cells treated with metformin and/or 2DG. High 2DG and metformin plus 2DG altered the metabolic profile similarly to metformin, inferring that metabolic changes are necessary but not sufficient while the specific effects of 2DG are crucial for detachment. Detached cells had higher NADPH levels and lower fatty acids and glutamine levels compared to attached cells, supporting the role of AMPK activation and reductive carboxylation in supporting anchorage-independent survival. Surprisingly, the metabolic profile of detached cells was closer to untreated control cells than attached treated cells, suggesting detachment might help cells adapt to energy stress. Metformin treated cells had higher fatty and amino acid levels with lower purine nucleotide levels, which is relevant for understanding the anticancer mechanisms of metformin.


Deoxyglucose/pharmacology , Hypoglycemic Agents/pharmacology , Metabolome/drug effects , Metformin/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Metabolomics , Triple Negative Breast Neoplasms/metabolism
4.
Int J Mol Sci ; 22(16)2021 Aug 10.
Article En | MEDLINE | ID: mdl-34445316

Inhibition of pyruvate dehydrogenase kinase (PDK) emerged as a potential strategy for treatment of cancer and metabolic disorders. Dichloroacetate (DCA), a prototypical PDK inhibitor, reduces the abundance of some PDK isoenzymes. However, the underlying mechanisms are not fully characterized and may differ across cell types. We determined that DCA reduced the abundance of PDK1 in breast (MDA-MB-231) and prostate (PC-3) cancer cells, while it suppressed both PDK1 and PDK2 in skeletal muscle cells (L6 myotubes). The DCA-induced PDK1 suppression was partially dependent on hypoxia-inducible factor-1α (HIF-1α), a transcriptional regulator of PDK1, in cancer cells but not in L6 myotubes. However, the DCA-induced alterations in the mRNA and the protein levels of PDK1 and/or PDK2 did not always occur in parallel, implicating a role for post-transcriptional mechanisms. DCA did not inhibit the mTOR signaling, while inhibitors of the proteasome or gene silencing of mitochondrial proteases CLPP and AFG3L2 did not prevent the DCA-induced reduction of the PDK1 protein levels. Collectively, our results suggest that DCA reduces the abundance of PDK in an isoform-dependent manner via transcriptional and post-transcriptional mechanisms. Differential response of PDK isoenzymes to DCA might be important for its pharmacological effects in different types of cells.


Dichloroacetic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Muscle Fibers, Skeletal/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , ATP-Dependent Proteases/antagonists & inhibitors , ATP-Dependent Proteases/metabolism , ATPases Associated with Diverse Cellular Activities/antagonists & inhibitors , ATPases Associated with Diverse Cellular Activities/metabolism , Animals , Cell Line, Tumor , Endopeptidase Clp/antagonists & inhibitors , Endopeptidase Clp/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , PC-3 Cells , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Rats
5.
Int J Mol Sci ; 21(20)2020 Oct 13.
Article En | MEDLINE | ID: mdl-33066271

Many studies evaluated the short-term in vitro toxicity of nanoparticles (NPs); however, long-term effects are still not adequately understood. Here, we investigated the potential toxic effects of biomedical (polyacrylic acid and polyethylenimine coated magnetic NPs) and two industrial (SiO2 and TiO2) NPs following different short-term and long-term exposure protocols on two physiologically different in vitro models that are able to differentiate: L6 rat skeletal muscle cell line and biomimetic normal porcine urothelial (NPU) cells. We show that L6 cells are more sensitive to NP exposure then NPU cells. Transmission electron microscopy revealed an uptake of NPs into L6 cells but not NPU cells. In L6 cells, we obtained a dose-dependent reduction in cell viability and increased reactive oxygen species (ROS) formation after 24 h. Following continuous exposure, more stable TiO2 and polyacrylic acid (PAA) NPs increased levels of nuclear factor Nrf2 mRNA, suggesting an oxidative damage-associated response. Furthermore, internalized magnetic PAA and TiO2 NPs hindered the differentiation of L6 cells. We propose the use of L6 skeletal muscle cells and NPU cells as a novel approach for assessment of the potential long-term toxicity of relevant NPs that are found in the blood and/or can be secreted into the urine.


Nanoparticles/toxicity , Toxicity Tests/methods , Animals , Cell Line , Cell Survival , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/physiology , Muscle Cells/metabolism , Muscle Cells/physiology , NF-E2-Related Factor 2/metabolism , Nanoparticles/chemistry , Polyesters/chemistry , Rats , Reactive Oxygen Species/metabolism , Swine , Titanium/chemistry , Urothelium/cytology
6.
Biochem Biophys Res Commun ; 515(2): 296-302, 2019 07 23.
Article En | MEDLINE | ID: mdl-31146913

Metabolic pathways of cancer cells depend on the concentrations of nutrients in their micro-environment as well as on the cell-to-cell interactions. Here we examined the effects of glucose, pyruvate and glutamine on the sensitivity of MDA-MB-231 cells to metabolic drug metformin using standard 2D culture, in which cells are grown in a monolayer, and 3D tumor spheroids, in which three-dimensional growth of cells better mimics a tumor. To examine effects of nutrients on metformin action, MDA-MB-231 cells were grown in commonly used media (DMEM, MEM and RPMI-1640) that differ mainly in the concentrations of amino acids. We used MTS assay and Hoechst and propidium iodide staining to determine cell number, viability and survival, respectively. We also determined the size of tumor spheroids and assessed effects of nutrients on metformin-stimulated AMP-activated protein kinase activation. Non-essential amino acids suppressed the effects of metformin on MDA-MB-231 cells in a 2D culture and in 3D tumor spheroids. Glutamine and pyruvate weakly diminished the effects of metformin in 2D culture. Furthermore, glucose protected tumor spheroids against metformin-induced disintegration. Our results show that nutrient availability must be considered when we evaluate the effects of metformin in 2D culture and in biologically more relevant 3D tumor spheroids.


Metformin/pharmacology , Triple Negative Breast Neoplasms/drug therapy , AMP-Activated Protein Kinases/metabolism , Cell Culture Techniques/methods , Cell Survival/drug effects , Enzyme Activation/drug effects , Female , Glucose/metabolism , Glucose/pharmacology , Glutamine/metabolism , Glutamine/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Nutrients/metabolism , Pyruvic Acid/metabolism , Pyruvic Acid/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/drug effects
7.
Sci Rep ; 7(1): 1761, 2017 05 11.
Article En | MEDLINE | ID: mdl-28496098

Triple naegative breast cancer has an increased rate of distant metastasis and consequently poor prognosis. To metastasize, breast cancer cells must detach from the main tumour mass and resist anoikis, a programmed cell death induced by lack of cell-extracellular matrix communication. Although cancer cells must detach to metastasize in vivo, the viability of floating cancer cells in vitro is rarely investigated. Here we show that co-treatment of anoikis-resistant MDA-MB-231 cells with metformin and 2-deoxy-D-glucose (2-DG) increased the percentage of floating cells, of which about 95% were viable. Floating cells resumed their proliferation once they were reseeded in the pharmacological compound-free medium. Similar effects on detachment were observed on anoikis-prone MCF-7 cells. Co-treatment of MDA-MB-231 cells with metformin and 2-DG induced a strong activation of AMP-activated protein kinase (AMPK), which was reduced by AMPK inhibitor compound C that prevented detachment of MDA-MB-231 cells. However, direct AMPK activators A-769662 and AICAR did not have any major effect on the percentage of floating MDA-MB-231 cells, indicating that AMPK activation is necessary but not sufficient for triggering detachment of cancer cells. Our results demonstrate that separate analysis of floating and attached cancer cells might be important for evaluation of anti-cancer agents.


Breast Neoplasms/pathology , Deoxyglucose/pharmacology , Metformin/pharmacology , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Biphenyl Compounds , Breast Neoplasms/enzymology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Female , Humans , Pyrones/pharmacology , Ribonucleotides/pharmacology , Thiophenes/pharmacology
8.
Toxicol Lett ; 270: 108-118, 2017 Mar 15.
Article En | MEDLINE | ID: mdl-28223195

Potential nanoparticle (NP) toxicity is one of crucial problems that limit the applicability of NPs. When designing NPs for biomedical and biotechnological applications it is thus important to understand the mechanisms of their toxicity. In this study, we analysed the stress responses of previously designed polyacrylic acid (PAA) and polyethylenimine (PEI) coated NPs on primary human myoblasts (MYO) and B16 mouse melanoma cell line. Negatively charged PAA did not induce cell toxicity, reactive oxygen species (ROS) or activate the transcription factor NF-κB in either cell line even at high concentrations (100µg/ml). On the other hand, positively charged PEI NPs induced a concentration dependent necrotic cell death and an increase in ROS following 24h incubation already at low concentrations (>4µg/ml). Moreover, PEI NPs induced NF-κB activation 15-30min after incubation in MYO cells, most probably through activation of TLR4 receptor. Interestingly, there was no NF-κB response to PEI NPs in B16 cells.


Cobalt/chemistry , Cobalt/toxicity , Ferric Compounds/chemistry , Ferric Compounds/toxicity , Myoblasts/drug effects , Nanoparticles/chemistry , Nanoparticles/toxicity , Acrylic Resins/chemistry , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Myoblasts/cytology , Myoblasts/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Polyethyleneimine/chemistry , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
...