201
|
Saquib Q, Ahmed S, Ahmad MS, Al-Rehaily AJ, Siddiqui MA, Faisal M, Ahmad J, Alsaleh AN, Alatar AA, Al-Khedhairy AA. Anticancer efficacies of persicogenin and homoeriodictyol isolated from Rhus retinorrhoea. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
202
|
Karg CA, Wang P, Kluibenschedl F, Müller T, Allmendinger L, Vollmar AM, Moser S. Phylloxanthobilins are Abundant Linear Tetrapyrroles from Chlorophyll Breakdown with Activities Against Cancer Cells. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000692] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Cornelia A. Karg
- Pharmaceutical Biology Pharmacy Department Ludwig‐Maximilians University of Munich Butenandtstraße 5‐13 81377 Munich Germany
| | - Pengyu Wang
- Pharmaceutical Biology Pharmacy Department Ludwig‐Maximilians University of Munich Butenandtstraße 5‐13 81377 Munich Germany
| | - Florian Kluibenschedl
- Institute of Organic Chemistry University of Innsbruck Innrain 80‐82 6020 Innsbruck Austria
| | - Thomas Müller
- Institute of Organic Chemistry University of Innsbruck Innrain 80‐82 6020 Innsbruck Austria
| | - Lars Allmendinger
- Pharmaceutical Chemistry Pharmacy Department Ludwig‐Maximilians University of Munich Butenandtstraße 5‐13 81377 Munich Germany
| | - Angelika M. Vollmar
- Pharmaceutical Biology Pharmacy Department Ludwig‐Maximilians University of Munich Butenandtstraße 5‐13 81377 Munich Germany
| | - Simone Moser
- Pharmaceutical Biology Pharmacy Department Ludwig‐Maximilians University of Munich Butenandtstraße 5‐13 81377 Munich Germany
| |
Collapse
|
203
|
Meßner M, Schmitt S, Ardelt MA, Fröhlich T, Müller M, Pein H, Huber-Cantonati P, Ortler C, Koenig LM, Zobel L, Koeberle A, Arnold GJ, Rothenfußer S, Kiemer AK, Gerbes AL, Zischka H, Vollmar AM, Pachmayr J. Metabolic implication of tigecycline as an efficacious second-line treatment for sorafenib-resistant hepatocellular carcinoma. FASEB J 2020; 34:11860-11882. [PMID: 32652772 DOI: 10.1096/fj.202001128r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
Abstract
Sorafenib represents the current standard of care for patients with advanced-stage hepatocellular carcinoma (HCC). However, acquired drug resistance occurs frequently during therapy and is accompanied by rapid tumor regrowth after sorafenib therapy termination. To identify the mechanism of this therapy-limiting growth resumption, we established robust sorafenib resistance HCC cell models that exhibited mitochondrial dysfunction and chemotherapeutic crossresistance. We found a rapid relapse of tumor cell proliferation after sorafenib withdrawal, which was caused by renewal of mitochondrial structures alongside a metabolic switch toward high electron transport system (ETS) activity. The translation-inhibiting antibiotic tigecycline impaired the biogenesis of mitochondrial DNA-encoded ETS subunits and limited the electron acceptor turnover required for glutamine oxidation. Thereby, tigecycline prevented the tumor relapse in vitro and in murine xenografts in vivo. These results offer a promising second-line therapeutic approach for advanced-stage HCC patients with progressive disease undergoing sorafenib therapy or treatment interruption due to severe adverse events.
Collapse
Affiliation(s)
- Martina Meßner
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.,Institute of Pharmacy, Center for Public Health, Paracelsus Medical University, Salzburg, Austria
| | - Sabine Schmitt
- School of Medicine, Institute of Toxicology and Environmental Hygiene, Technical University Munich, Munich, Germany
| | - Maximilian A Ardelt
- Institute of Pharmacy, Center for Public Health, Paracelsus Medical University, Salzburg, Austria
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Centre, LMU Munich, Munich, Germany
| | - Martin Müller
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Helmut Pein
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Petra Huber-Cantonati
- Institute of Pharmacy, Center for Public Health, Paracelsus Medical University, Salzburg, Austria
| | - Carina Ortler
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Lars M Koenig
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany
| | - Lena Zobel
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Andreas Koeberle
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany.,Michael Popp Research Institute, University of Innsbruck, Innsbruck, Austria
| | - Georg J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Centre, LMU Munich, Munich, Germany
| | - Simon Rothenfußer
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Alexander L Gerbes
- Department of Medicine 2, Liver Center Munich, University Hospital, LMU Munich, Munich, Germany
| | - Hans Zischka
- School of Medicine, Institute of Toxicology and Environmental Hygiene, Technical University Munich, Munich, Germany.,Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Johanna Pachmayr
- Institute of Pharmacy, Center for Public Health, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
204
|
Synthesis, cytotoxicity of some pyrazoles and pyrazolo[1,5-a]pyrimidines bearing benzothiazole moiety and investigation of their mechanism of action. Bioorg Chem 2020; 102:104053. [PMID: 32673889 DOI: 10.1016/j.bioorg.2020.104053] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 06/01/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023]
Abstract
A novel series of pyrazoles and pyrazolo[1,5-a]pyrimidines bearing benzothiazole moiety were designed and synthesized. Chemical structures were confirmed by spectral data and elemental analyses. Nine compounds were selected and screened for their cytotoxic activity at the National Cancer Institute (NCI), USA against 60 cancer cell lines in a single dose assay. Compounds 4 and 5 exerted the most potent growth inhibitory activity against most cancer cell lines with growth inhibition (GI%) ranges from 44.86% to 84.59% and 31.20% to 52.36%, respectively. Consequently, they were further investigated through IC50 determination using five dose MTT colorimetric assay against three sensitive cell lines, leukemia CCRF-CEM, non-small cell lung cancer HOP-92 and liver cancer Hep-G2. Compound 4 exhibited potent cytotoxic activity against the three tested cell lines with IC50 16.34, 3.45 and 7.79 μM, respectively representing half potency, 3.5 folds potency and nearly equipotent to roscovitine. To investigate its mechanism of action, cell cycle analysis of compound 4 was conducted and showed that it induced cell cycle arrest at G2/M phase and apoptosis in HOP-92 cells. In correlation with the previous results, caspase-3 activation was tested and illustrated elevation in its concentration by nearly 14 folds than control. Besides, enzyme inhibition assay of compound 4 was evaluated towards two common antitumor targets namely KDM1 and CDK1 showing significant inhibitory activity with IC50 0.096 and 0.078 μM, respectively.
Collapse
|
205
|
Lima EJSPD, Fontes SS, Nogueira ML, Silva VR, Santos LDS, D'Elia GMA, Dias RB, Sales CBS, Rocha CAG, Vannier-Santos MA, Soares MBP, Costa EV, Silva FMAD, Koolen HHF, Bezerra DP. Essential oil from leaves of Conobea scoparioides (Cham. & Schltdl.) Benth. (Plantaginaceae) causes cell death in HepG2 cells and inhibits tumor development in a xenograft model. Biomed Pharmacother 2020; 129:110402. [PMID: 32574969 DOI: 10.1016/j.biopha.2020.110402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/30/2020] [Accepted: 06/13/2020] [Indexed: 12/24/2022] Open
Abstract
Conobea scoparioides (Cham. & Schltdl.) Benth. (syn. Sphaerotheca scoparioides Cham. & Schldtl.) (Plantaginaceae), popularly known as "pataqueira", "vassourinha-do-brejo" and/or "hierba-de-sapo", is a popular medicinal plant used to treat leishmaniasis, pain and beriberi. In addition, inhibition of cell adhesion, antioxidant, cytotoxic and leishmanicidal activities of compounds or fractions of C. scoparioides have been reported. In the present work, chemical constituents and in vitro and in vivo anti-liver cancer potential of essential oil (EO) from leaves of C. scoparioides were investigated using human hepatocellular carcinoma HepG2 cells as a cell model. EO was obtained by hydrodistillation using a Clevenger-type apparatus and characterized by GC-MS and GC-FID. The in vitro cytotoxic effect was evaluated on three human cancer cell lines (MCF-7, HepG2 and HCT116) and one human non-cancerous cell line (MRC-5) using the Alamar blue assay. Phosphatidylserine externalization and cell cycle distribution were quantified in HepG2 cells by flow cytometry after 48 h incubation. The effectiveness of EO in anti-liver cancer model was studied with HepG2 cells grafted on C.B. 17 SCID mice. The main constituents of EO were thymol methyl ether (62 %), thymol (16 %) and α-phellandrene (14 %). EO displayed an in vitro cytotoxic effect against all human cancer cell lines and caused externalization of phosphatidylserine and DNA fragmentation in HepG2 cells, suggesting induction of apoptotic-like cell death. In vivo tumor mass inhibition of 36.7 and 55.8 % was observed for treatment with EO at doses of 40 and 80 mg/kg, respectively. These results indicate in vitro and in vivo anti-liver cancer potential of EO from leaves of C. scoparioides.
Collapse
Affiliation(s)
- Emilly J S P de Lima
- Metabolomics and Mass Spectrometry Research Group, Amazonas State University (UEA), Manaus, Amazonas, 690065-130, Brazil
| | - Sheila S Fontes
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Mateus L Nogueira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Valdenizia R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Luciano de S Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Gigliola M A D'Elia
- Metabolomics and Mass Spectrometry Research Group, Amazonas State University (UEA), Manaus, Amazonas, 690065-130, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; Department of Clinical Propaedeutics and Integrated Clinical, Faculty of Dentistry, Federal University of Bahia, Salvador, Bahia, 40301-155, Brazil
| | - Caroline B S Sales
- Department of Biomorphology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-902, Brazil
| | - Clarissa A Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; Department of Clinical Propaedeutics and Integrated Clinical, Faculty of Dentistry, Federal University of Bahia, Salvador, Bahia, 40301-155, Brazil
| | | | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Emmanoel V Costa
- Department of Chemistry, Federal University of Amazonas (UFAM), Manaus, Amazonas, 69080-900, Brazil
| | - Felipe M A da Silva
- Department of Chemistry, Federal University of Amazonas (UFAM), Manaus, Amazonas, 69080-900, Brazil
| | - Hector H F Koolen
- Metabolomics and Mass Spectrometry Research Group, Amazonas State University (UEA), Manaus, Amazonas, 690065-130, Brazil.
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil.
| |
Collapse
|
206
|
Belyaeva EA, Sokolova TV. Mitigating effect of paxilline against injury produced by Cd 2+ in rat pheochromocytoma PC12 and ascites hepatoma AS-30D cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 196:110519. [PMID: 32244116 DOI: 10.1016/j.ecoenv.2020.110519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/15/2020] [Accepted: 03/17/2020] [Indexed: 02/08/2023]
Abstract
On two rat cell lines, pheochromocytoma PC12 and ascites hepatoma AS-30D, and on rat liver mitochondria we studied action of paxilline (lipophilic mycotoxin from fungus Penicillium paxilli which is blocker of large-conductance potassium channels) against harmful effects of Cd(II) - one of the most dangerous toxic metals and environmental pollutants. We investigated an influence of paxilline on cell viability and mitochondrial function in the presence and in the absence of Cd2+. As found, paxilline protected partially from the Cd2+-induced cytotoxicity, namely taken in concentration of 1 μM it decreased the Cd2+-induced cell necrosis in average by 10-14 or 13-23% for AS-30D and PC12 cells, respectively. Nevertheless, paxilline did not affect the Cd2+-induced apoptosis of AS-30D cells. The alleviating concentration of paxilline reduced an intracellular production of reactive oxygen species (ROS) in PC12 cells intoxicated by Cd2+ and enhanced the ROS production in control AS-30D cells; however, it weakly affected mitochondrial membrane potential of the cells in the absence and in the presence of Cd2+. The ameliorative concentration of paxilline decreased the maximal respiration rates of control cells of both types after short-term (3-5 h) treatment with it while the rates reached their control levels after long-term (24-48 h) incubation with the drug. Paxilline was not protective against the Cd2+-induced membrane permeability and respiration rate changes in isolated rat liver mitochondria. As result, the mitochondrial electron transport chain was concluded to contribute in the mitigating effect of paxilline against the Cd2+-produced cell injury.
Collapse
Affiliation(s)
- Elena A Belyaeva
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Thorez Pr. 44, 194223, St.-Petersburg, Russia.
| | - Tatyana V Sokolova
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Thorez Pr. 44, 194223, St.-Petersburg, Russia
| |
Collapse
|
207
|
Cyperus articulatus L. (Cyperaceae) Rhizome Essential Oil Causes Cell Cycle Arrest in the G 2/M Phase and Cell Death in HepG2 Cells and Inhibits the Development of Tumors in a Xenograft Model. Molecules 2020; 25:molecules25112687. [PMID: 32527068 PMCID: PMC7321242 DOI: 10.3390/molecules25112687] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/01/2020] [Accepted: 06/04/2020] [Indexed: 12/21/2022] Open
Abstract
Cyperus articulatus L. (Cyperaceae), popularly known in Brazil as “priprioca” or “piriprioca”, is a tropical and subtropical plant used in popular medical practices to treat many diseases, including cancer. In this study, C. articulatus rhizome essential oil (EO), collected from the Brazilian Amazon rainforest, was addressed in relation to its chemical composition, induction of cell death in vitro and inhibition of tumor development in vivo, using human hepatocellular carcinoma HepG2 cells as a cell model. EO was obtained by hydrodistillation using a Clevenger-type apparatus and characterized qualitatively and quantitatively by gas chromatography coupled to mass spectrometry (GC-MS) and gas chromatography with flame ionization detection (GC-FID), respectively. The cytotoxic activity of EO was examined against five cancer cell lines (HepG2, HCT116, MCF-7, HL-60 and B16-F10) and one non-cancerous one (MRC-5) using the Alamar blue assay. Cell cycle distribution and cell death were investigated using flow cytometry in HepG2 cells treated with EO after 24, 48 and 72 h of incubation. The cells were also stained with May–Grunwald–Giemsa to analyze the morphological changes. The anti-liver-cancer activity of EO in vivo was evaluated in C.B-17 severe combined immunodeficient (SCID) mice with HepG2 cell xenografts. The main representative substances of this EO sample were muskatone (11.6%), cyclocolorenone (10.3%), α-pinene (8.26%), pogostol (6.36%), α-copaene (4.83%) and caryophyllene oxide (4.82%). EO showed IC50 values for cancer cell lines ranging from 28.5 µg/mL for HepG2 to >50 µg/mL for HCT116, and an IC50 value for non-cancerous of 46.0 µg/mL (MRC-5), showing selectivity indices below 2-fold for all cancer cells tested. HepG2 cells treated with EO showed cell cycle arrest at G2/M along with internucleosomal DNA fragmentation. The morphological alterations included cell shrinkage and chromatin condensation. Treatment with EO also increased the percentage of apoptotic-like cells. The in vivo tumor mass inhibition rates of EO were 46.5–50.0%. The results obtained indicate the anti-liver-cancer potential of C. articulatus rhizome EO.
Collapse
|
208
|
Attia SM, Al-Khalifa MK, Al-Hamamah MA, Alotaibi MR, Attia MSM, Ahmad SF, Ansari MA, Nadeem A, Bakheet SA. Vorinostat is genotoxic and epigenotoxic in the mouse bone marrow cells at the human equivalent doses. Toxicology 2020; 441:152507. [PMID: 32512035 DOI: 10.1016/j.tox.2020.152507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/10/2020] [Accepted: 06/02/2020] [Indexed: 12/16/2022]
Abstract
Vorinostat was approved as the first histone deacetylase inhibitor for the management of cutaneous T cell lymphoma. However, it's in vivo genetic and epigenetic effects on non-cancerous cells remain poorly understood. As genetic and epigenetic changes play a critical role in the pathogenesis of carcinogenesis, we investigated whether vorinostat induces genetic and epigenetic alterations in mouse bone marrow cells. Bone marrow cells were isolated 24 h following the last oral administration of vorinostat at the doses of 25, 50, or 100 mg/kg/day for five days (approximately equal to the recommended human doses). The cells were then used to assess clastogenicity and aneugenicity by the micronucleus test complemented by fluorescence in situ hybridization assay; DNA strand breaks, oxidative DNA strand breaks, and DNA methylation by the modified comet assay; apoptosis by annexin V/PI staining analysis and the occurrence of the hypodiploid DNA content; and DNA damage/repair gene expression by polymerase chain reaction (PCR) Array. The expression of the mRNA transcripts were also confirmed by real-time PCR and western blot analysis. Vorinostat caused structural chromosomal damage, numerical chromosomal abnormalities, DNA strand breaks, oxidative DNA strand breaks, DNA hypomethylation, and programed cell death in a dose-dependent manner. Furthermore, the expression of numerous genes implicated in DNA damage/repair were altered after vorinostat treatment. Accordingly, the genetic/epigenetic mechanism(s) of action of vorinostat may play a role in its carcinogenicity and support the continued study and development of new compounds with lower toxicity.
Collapse
Affiliation(s)
- Sabry M Attia
- College of Pharmacy, Pharmacology and Toxicology Department, King Saud University, Riyadh, Saudi Arabia.
| | - Mohamed K Al-Khalifa
- College of Pharmacy, Pharmacology and Toxicology Department, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed A Al-Hamamah
- College of Pharmacy, Pharmacology and Toxicology Department, King Saud University, Riyadh, Saudi Arabia
| | - Moureq R Alotaibi
- College of Pharmacy, Pharmacology and Toxicology Department, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed S M Attia
- College of Pharmacy, Pharmacology and Toxicology Department, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh F Ahmad
- College of Pharmacy, Pharmacology and Toxicology Department, King Saud University, Riyadh, Saudi Arabia
| | - Mushtaq A Ansari
- College of Pharmacy, Pharmacology and Toxicology Department, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- College of Pharmacy, Pharmacology and Toxicology Department, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- College of Pharmacy, Pharmacology and Toxicology Department, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
209
|
Naleskina LA, Lukianova NY, Lozovska YV, Todor IM, Andrusishyna IM, Kunska LM, Chekhun VF. Changes of Morphological Characteristics and Metabolic Profile of Walker-256 Carcinosarcoma under the Impact of Exogenous Lactoferrin. CYTOL GENET+ 2020. [DOI: 10.3103/s0095452720030093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
210
|
Dittmann J, Haydn T, Metzger P, Ward GA, Boerries M, Vogler M, Fulda S. Next-generation hypomethylating agent SGI-110 primes acute myeloid leukemia cells to IAP antagonist by activating extrinsic and intrinsic apoptosis pathways. Cell Death Differ 2020; 27:1878-1895. [PMID: 31831875 PMCID: PMC7244748 DOI: 10.1038/s41418-019-0465-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022] Open
Abstract
Therapeutic efficacy of first-generation hypomethylating agents (HMAs) is limited in elderly acute myeloid leukemia (AML) patients. Therefore, combination strategies with targeted therapies are urgently needed. Here, we discover that priming with SGI-110 (guadecitabine), a next-generation HMA, sensitizes AML cells to ASTX660, a novel antagonist of cellular inhibitor of apoptosis protein 1 and 2 (cIAP1/2) and X-linked IAP (XIAP). Importantly, SGI-110 and ASTX660 synergistically induced cell death in a panel of AML cell lines as well as in primary AML samples while largely sparing normal CD34+ human progenitor cells, underlining the translational relevance of this combination. Unbiased transcriptome analysis revealed that SGI-110 alone or in combination with ASTX660 upregulated the expression of key regulators of both extrinsic and intrinsic apoptosis signaling pathways such as TNFRSF10B (DR5), FAS, and BAX. Individual knockdown of the death receptors TNFR1, DR5, and FAS significantly reduced SGI-110/ASTX660-mediated cell death, whereas blocking antibodies for tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) or FAS ligand (FASLG) failed to provide protection. Also, TNFα-blocking antibody Enbrel had little protective effect on SGI-110/ASTX660-induced cell death. Further, SGI-110 and ASTX660 acted in concert to promote cleavage of caspase-8 and BID, thereby providing a link between extrinsic and intrinsic apoptotic pathways. Consistently, sequential treatment with SGI-110 and ASTX660-triggered loss of mitochondrial membrane potential (MMP) and BAX activation which contributes to cell death, as BAX silencing significantly protected from SGI-110/ASTX660-mediated apoptosis. Together, these events culminated in the activation of caspases-3/-7, nuclear fragmentation, and cell death. In conclusion, SGI-110 and ASTX660 cooperatively induced apoptosis in AML cells by engaging extrinsic and intrinsic apoptosis pathways, highlighting the therapeutic potential of this combination for AML.
Collapse
Affiliation(s)
- Jessica Dittmann
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Tinka Haydn
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Patrick Metzger
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University Freiburg, Freiburg im Breisgau, Germany
- Institute of Molecular Medicine and Cell Research (IMMZ), Albert Ludwigs-University Freiburg, Freiburg im Breisgau, Germany
- Faculty of Biology, University Freiburg, Freiburg im Breisgau, Germany
| | | | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University Freiburg, Freiburg im Breisgau, Germany
- Institute of Molecular Medicine and Cell Research (IMMZ), Albert Ludwigs-University Freiburg, Freiburg im Breisgau, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg im Breisgau, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Meike Vogler
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
211
|
Liu Y, Jia Y, Yang K, Li R, Xiao X, Zhu K, Wang Z. Metformin Restores Tetracyclines Susceptibility against Multidrug Resistant Bacteria. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902227. [PMID: 32596101 PMCID: PMC7312304 DOI: 10.1002/advs.201902227] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 03/29/2020] [Indexed: 05/22/2023]
Abstract
Highly persistent incidence of multidrug resistant (MDR) bacterial pathogens constitutes a global burden for public health. An alternative strategy to alleviate such a crisis is to identify promising compounds to restore antibiotics activity against MDR bacteria. It is reported that the antidiabetic drug metformin exhibits the potentiation effect on tetracycline antibiotics, particularly doxycycline and minocycline, against MDR S. aureus, E. faecalis, E. coli, and S. enteritidis. Mechanistic studies demonstrate that metformin promotes intracellular accumulation of doxycycline in tetracycline-resistant E. coli. In addition, metformin boosts the immune response and alleviates the inflammatory responses in vitro. Last, metformin fully restores the activity of doxycycline in three animal infection models. Collectively, these results reveal the potential of metformin as a novel tetracyclines adjuvant to circumvent MDR bacterial pathogens and to improve the treatment outcome of recalcitrant infections.
Collapse
Affiliation(s)
- Yuan Liu
- Institute of Comparative MedicineCollege of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouJiangsu225009China
| | - Yuqian Jia
- Institute of Comparative MedicineCollege of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
| | - Kangni Yang
- Institute of Comparative MedicineCollege of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
| | - Ruichao Li
- Institute of Comparative MedicineCollege of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouJiangsu225009China
| | - Xia Xiao
- Institute of Comparative MedicineCollege of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouJiangsu225009China
| | - Kui Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human HealthCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Zhiqiang Wang
- Institute of Comparative MedicineCollege of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouJiangsu225009China
| |
Collapse
|
212
|
Simchi L, Panov J, Morsy O, Feuermann Y, Kaphzan H. Novel Insights into the Role of UBE3A in Regulating Apoptosis and Proliferation. J Clin Med 2020; 9:jcm9051573. [PMID: 32455880 PMCID: PMC7290732 DOI: 10.3390/jcm9051573] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/05/2020] [Accepted: 05/17/2020] [Indexed: 12/13/2022] Open
Abstract
The UBE3A gene codes for a protein with two known functions, a ubiquitin E3-ligase which catalyzes ubiquitin binding to substrate proteins and a steroid hormone receptor coactivator. UBE3A is most famous for its critical role in neuronal functioning. Lack of UBE3A protein expression leads to Angelman syndrome (AS), while its overexpression is associated with autism. In spite of extensive research, our understanding of UBE3A roles is still limited. We investigated the cellular and molecular effects of Ube3a deletion in mouse embryonic fibroblasts (MEFs) and Angelman syndrome (AS) mouse model hippocampi. Cell cultures of MEFs exhibited enhanced proliferation together with reduced apoptosis when Ube3a was deleted. These findings were supported by transcriptome and proteome analyses. Furthermore, transcriptome analyses revealed alterations in mitochondria-related genes. Moreover, an analysis of adult AS model mice hippocampi also found alterations in the expression of apoptosis- and proliferation-associated genes. Our findings emphasize the role UBE3A plays in regulating proliferation and apoptosis and sheds light into the possible effects UBE3A has on mitochondrial involvement in governing this balance.
Collapse
|
213
|
Wang W, Liu Y, Niu J, Lin W. Discrimination of live and dead cells with two different sets of signals and unique application in vivo imaging. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 231:118115. [PMID: 32007905 DOI: 10.1016/j.saa.2020.118115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/15/2020] [Accepted: 01/24/2020] [Indexed: 06/10/2023]
Abstract
Discriminating living and dead cells is of great significance for the study of apoptosis. In this work, we have developed a unique fluorescent probe (RPIC) for discriminating live and dead cells with duel-channel fluorescence imaging under double excitation and double emission mode. Dead cells treated with RPIC shows weak fluorescence signals in red channel, however, strong fluorescence signals are appeared in red channel in live cells. Weak and strong green fluorescence signals present at live cells and dead cells, respectively. Moreover, RPIC can detect successfully apoptosis of cancer cells. For in-vivo imaging, RPIC can discriminate successfully live and dead zebrafish with the same method. More interestingly, it is found that RPIC possesses the ability of discriminating normal mice and tumor mice.
Collapse
Affiliation(s)
- Weishan Wang
- Institute of Fluorescent Probes for Biological Imaging, School of Materials Science and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Shandong 250022, PR China
| | - Yong Liu
- Institute of Fluorescent Probes for Biological Imaging, School of Materials Science and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Shandong 250022, PR China
| | - Jie Niu
- Institute of Fluorescent Probes for Biological Imaging, School of Materials Science and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Shandong 250022, PR China
| | - Weiying Lin
- Institute of Fluorescent Probes for Biological Imaging, School of Materials Science and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Shandong 250022, PR China.
| |
Collapse
|
214
|
Ewes WA, Elmorsy MA, El-Messery SM, Nasr MN. Synthesis, biological evaluation and molecular modeling study of [1,2,4]-Triazolo[4,3-c]quinazolines: New class of EGFR-TK inhibitors. Bioorg Med Chem 2020; 28:115373. [DOI: 10.1016/j.bmc.2020.115373] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/21/2022]
|
215
|
Zickri MB, Sadek EM, Fares AE, Heteba NG, Reda AM. Effect of Stem Cells, Ascorbic Acid and SERCA1a Gene Transfected Stem Cells in Experimentally Induced Type I Diabetic Myopathy. Int J Stem Cells 2020; 13:163-175. [PMID: 32114738 PMCID: PMC7119208 DOI: 10.15283/ijsc18066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/06/2018] [Accepted: 01/15/2019] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibition was proved in streptozotocin (STZ)-diabetic rats. The present study aimed at investigating and comparing the therapeutic effect of bone marrow mesenchymal stem cells (BMMSCs), BMMSCs combined with ascorbic acid (AA) and SERCA1a gene transfected BMMSCs in induced type I diabetic myopathy of male albino rat. Methods and Results 54 rats were divided into donor group of 6 rats for isolation, propagation and characterization of BMMSCs and SERCA1a transfected BMMSCs, groups I∼V 48 rats. Group I of 8 control rats, group II (Diabetic) of 10 rats given STZ 50 mg/kg intraperitoneal, group III (BMMSCs) of 10 rats given STZ and BMMSCs intravenous (IV), group IV (BMMSCs and AA) of 10 rats given STZ, BMMSCs IV and AA 500 mg/kg and group V (SERCA 1a transfected BMMSCs) of 10 rats given STZ and SERCA1a transfected BMMSCs IV. The rats were sacrificed after 8 weeks. Gastrocnemius specimens were subjected to biochemical, histological, morphometric and statistical studies. Diabetic rats revealed inflammatory and degenerative muscle changes, a significant increase in blood glucose level, mean DNA fragmentation and mean MDA values and a significant decrease in mean GSH and catalase values, area of pale nuclei, area% of CD105 and CD34 +ve cells, SERCA1a protein and gene values. The morphological changes regressed by therapy. In group III significant decrease in DNA fragmentation and MDA, significant increase in GSH and catalase, significant increase in the mean area of pale nuclei, area % of CD105 and CD34 +ve cells versus diabetic group. In group IV, same findings as group III versus diabetic and BMMSCs groups. In group V, same findings as group IV versus diabetic and treated groups. Western blot and PCR proved a mean value of SERCA1a protein and gene comparable to the control group. Mean calcium concentration values revealed a significant increase in the diabetic group, in BMMSCs and AA group versus control and SERCA1a group. Conclusions SERCA1a transfected BMMSCs proved a definite therapeutic effect, more remarkable than BMMSCs combined with AA. This effect was evidenced histologically and confirmed by significant changes in the biochemical tests indicating oxidative stress, muscle calcium concentration, morphometric parameters and PCR values of SERCA1a.
Collapse
Affiliation(s)
- Maha B Zickri
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Cairo University, Giza, Egypt.,Faculty of Oral and Dental Medicine, Future University in Egypt (FUE), New Cairo City, Egypt
| | - Eman M Sadek
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Amal E Fares
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Cairo University, Giza, Egypt
| | | | - Ahmed M Reda
- Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt.,Faculty of Pharmacy, Near East University, North Cyprus, Cyprus
| |
Collapse
|
216
|
Zbakh H, Zubía E, De Los Reyes C, Calderón-Montaño JM, Motilva V. Anticancer Activities of Meroterpenoids Isolated from the Brown Alga Cystoseira usneoides against the Human Colon Cancer Cells HT-29. Foods 2020; 9:foods9030300. [PMID: 32155797 PMCID: PMC7143549 DOI: 10.3390/foods9030300] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 12/03/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common types of cancers and a leading cause of cancer death worldwide. The current treatment for CRC mainly involves surgery, radiotherapy, and chemotherapy. However, due to the side effects and the emergence of drug resistance, the search for new anticancer agents, pharmacologically safe and effective, is needed. In the present study, we have investigated the anticancer effects of eight algal meroterpenoids (AMTs, 1-8) isolated from the brown seaweed Cystoseira usneoides and their underlying mechanisms of action using HT-29, a highly metastatic human colon cancer cell line. All the tested meroterpenoids inhibited the growth of HT-29 malignant cells and were less toxic towards non-cancer colon cells, with the AMTs 1 and 5 exhibiting selectivity indexes of 5.26 and 5.23, respectively. Treatment of HT-29 cells with the AMTs 1, 2, 3, 4, 5, and 7 induced cell cycle arrest in G2/M phase and, in some instances, apoptosis (compounds 2, 3, and 5). Compounds 1-8 also exhibited significant inhibitory effects on the migration and/or invasion of colon cancer cells. Mechanistic analysis demonstrated that the AMTs 1, 2, 5, 6, 7, and 8 reduced phosphorylation levels of extracellular signal-regulated kinase (ERK) and the AMTs 2, 3, 4, 5, 7, and 8 decreased phosphorylation of c-JUN N-terminal kinase (JNK). Moreover, the AMTs 1, 2, 3, 4, 7, and 8 inhibited phosphorylation levels of protein kinase B (AKT) in colon carcinoma cells. These results provide new insights into the mechanisms and functions of the meroterpenoids of C. usneoides, which exhibit an anticancer effect on HT-29 colon cancer cells by inducing cell cycle arrest and apoptosis via the downregulation of ERK/JNK/AKT signaling pathways.
Collapse
Affiliation(s)
- Hanaa Zbakh
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (H.Z.); (J.M.C.-M.)
- Department of Biology, Faculty of Sciences, University of Abdelmalek Essaâdi, Tetouan 93000, Morocco
| | - Eva Zubía
- Department of Organic Chemistry, Faculty of Marine and Environmental Sciences, University of Cadiz, 11510 Puerto Real (Cádiz), Spain; (E.Z.); (C.D.L.R.)
| | - Carolina De Los Reyes
- Department of Organic Chemistry, Faculty of Marine and Environmental Sciences, University of Cadiz, 11510 Puerto Real (Cádiz), Spain; (E.Z.); (C.D.L.R.)
| | - José M. Calderón-Montaño
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (H.Z.); (J.M.C.-M.)
| | - Virginia Motilva
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (H.Z.); (J.M.C.-M.)
- Correspondence:
| |
Collapse
|
217
|
Mattei JC, Bouvier-Labit C, Barets D, Macagno N, Chocry M, Chibon F, Morando P, Rochwerger RA, Duffaud F, Olschwang S, Salas S, Jiguet-Jiglaire C. Pan Aurora Kinase Inhibitor: A Promising Targeted-Therapy in Dedifferentiated Liposarcomas With Differential Efficiency Depending on Sarcoma Molecular Profile. Cancers (Basel) 2020; 12:E583. [PMID: 32138169 PMCID: PMC7139289 DOI: 10.3390/cancers12030583] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/24/2020] [Accepted: 02/28/2020] [Indexed: 11/17/2022] Open
Abstract
Soft tissue sarcoma (STS) are rare and aggressive tumours. Their classification includes numerous histological subtypes of frequent poor prognosis. Liposarcomas (LPS) are the most frequent type among them, and the aggressiveness and deep localization of dedifferentiated LPS are linked to high levels of recurrence. Current treatments available today lead to five-year overall survival has remained stuck around 60%-70% for the past three decades. Here, we highlight a correlation between Aurora kinasa A (AURKA) and AURKB mRNA overexpression and a low metastasis - free survival. AURKA and AURKB expression analysis at genomic and protein level on a 9-STS cell lines panel highlighted STS heterogeneity, especially in LPS subtype. AURKA and AURKB inhibition by RNAi and drug targeting with AMG 900, a pan Aurora Kinase inhibitor, in four LPS cell lines reduces cell survival and clonogenic proliferation, inducing apoptosis and polyploidy. When combined with doxorubicin, the standard treatment in STS, aurora kinases inhibitor can be considered as an enhancer of standard treatment or as an independent drug. Kinome analysis suggested its effect was linked to the inhibition of the MAP-kinase pathway, with differential drug resistance profiles depending on molecular characteristics of the tumor. Aurora Kinase inhibition by AMG 900 could be a promising therapy in STS.
Collapse
Affiliation(s)
- Jean Camille Mattei
- Aix-Marseille University, Inserm, MMG, 13005 Marseille, France; (J.C.M.); (C.B.-L.); (R.A.R.); (F.D.); (S.O.); (S.S.)
- APHM, Hôpital Nord, Service d'Orthopédie et traumatologie, 13015 Marseille, France
| | - Corinne Bouvier-Labit
- Aix-Marseille University, Inserm, MMG, 13005 Marseille, France; (J.C.M.); (C.B.-L.); (R.A.R.); (F.D.); (S.O.); (S.S.)
- APHM, Hôpital de la Timone, Service d’Anatomie Pathologique et de Neuropathologie, 13005 Marseille, France; (D.B.); (N.M.)
| | - Doriane Barets
- APHM, Hôpital de la Timone, Service d’Anatomie Pathologique et de Neuropathologie, 13005 Marseille, France; (D.B.); (N.M.)
| | - Nicolas Macagno
- APHM, Hôpital de la Timone, Service d’Anatomie Pathologique et de Neuropathologie, 13005 Marseille, France; (D.B.); (N.M.)
| | - Mathieu Chocry
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France; (M.C.); (P.M.)
| | | | - Philippe Morando
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France; (M.C.); (P.M.)
| | - Richard Alexandre Rochwerger
- Aix-Marseille University, Inserm, MMG, 13005 Marseille, France; (J.C.M.); (C.B.-L.); (R.A.R.); (F.D.); (S.O.); (S.S.)
- APHM, Hôpital Nord, Service d'Orthopédie et traumatologie, 13015 Marseille, France
| | - Florence Duffaud
- Aix-Marseille University, Inserm, MMG, 13005 Marseille, France; (J.C.M.); (C.B.-L.); (R.A.R.); (F.D.); (S.O.); (S.S.)
- APHM, Hôpital de la Timone, Service d’Oncologie adulte, 13005 Marseille, France
| | - Sylviane Olschwang
- Aix-Marseille University, Inserm, MMG, 13005 Marseille, France; (J.C.M.); (C.B.-L.); (R.A.R.); (F.D.); (S.O.); (S.S.)
- APHM, Hôpital de la Timone, Département de Génétique Médicale, 13005 Marseille, France
- Ramsay Générale de Santé, Hôpital Clairval, Institut de Cancérologie, 13005 Marseille, France
| | - Sébastien Salas
- Aix-Marseille University, Inserm, MMG, 13005 Marseille, France; (J.C.M.); (C.B.-L.); (R.A.R.); (F.D.); (S.O.); (S.S.)
- APHM, Hôpital de la Timone, Service d’Oncologie adulte, 13005 Marseille, France
| | - Carine Jiguet-Jiglaire
- APHM, Hôpital de la Timone, Service d’Anatomie Pathologique et de Neuropathologie, 13005 Marseille, France; (D.B.); (N.M.)
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France; (M.C.); (P.M.)
- APHM, Centre de Ressources Biologiques, 13005 Marseille, France
| |
Collapse
|
218
|
Cytotoxicity and cell death induced by engineered nanostructures (quantum dots and nanoparticles) in human cell lines. J Biol Inorg Chem 2020; 25:325-338. [DOI: 10.1007/s00775-020-01764-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/12/2020] [Indexed: 10/24/2022]
|
219
|
Ragab FA, Nissan YM, Seif EM, Maher A, Arafa RK. Synthesis and in vitro investigation of novel cytotoxic pyrimidine and pyrazolopyrimidne derivatives showing apoptotic effect. Bioorg Chem 2020; 96:103621. [DOI: 10.1016/j.bioorg.2020.103621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/01/2020] [Accepted: 01/22/2020] [Indexed: 12/17/2022]
|
220
|
Mühlberger M, Unterweger H, Band J, Lehmann C, Heger L, Dudziak D, Alexiou C, Lee G, Janko C. Loading of Primary Human T Lymphocytes with Citrate-Coated Superparamagnetic Iron Oxide Nanoparticles Does Not Impair Their Activation after Polyclonal Stimulation. Cells 2020; 9:cells9020342. [PMID: 32024193 PMCID: PMC7072432 DOI: 10.3390/cells9020342] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/14/2020] [Accepted: 01/28/2020] [Indexed: 12/27/2022] Open
Abstract
For the conversion of immunologically cold tumors, characterized by a low T cell infiltration, into hot tumors, it is necessary to enrich T cells in the tumor area. One possibility is the use of magnetic fields to direct T cells into the tumor. For this purpose, primary T cells that were freshly isolated from human whole blood were loaded with citrate-coated superparamagnetic iron oxide nanoparticles (SPIONCitrate). Cell toxicity and particle uptake were investigated by flow cytometry and atomic emission spectroscopy. The optimum loading of the T cells without any major effect on their viability was achieved with a particle concentration of 75 µg Fe/mL and a loading period of 24 h. The cellular content of SPIONCitrate was sufficient to attract these T cells with a magnet which was monitored by live-cell imaging. The functionality of the T cells was only slightly influenced by SPIONCitrate, as demonstrated by in vitro stimulation assays. The proliferation rate as well as the expression of co-stimulatory and inhibitory surface molecules (programmed cell death 1 (PD-1), lymphocyte activation gene 3 (LAG-3), T cell immunoglobulin and mucin domain containing 3 (Tim-3), C-C motif chemokine receptor 7 (CCR7), CD25, CD45RO, CD69) was investigated and found to be unchanged. Our results presented here demonstrate the feasibility of loading primary human T lymphocytes with superparamagnetic iron oxide nanoparticles without influencing their viability and functionality while achieving sufficient magnetizability for magnetically controlled targeting. Thus, the results provide a strong fundament for the transfer to tumor models and ultimately for new immunotherapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Marina Mühlberger
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.M.)
- Department of Chemistry and Pharmacy, Division of Pharmaceutics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Harald Unterweger
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.M.)
| | - Julia Band
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.M.)
| | - Christian Lehmann
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Universitätsklinikum Erlangen, 91052 Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Lukas Heger
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Universitätsklinikum Erlangen, 91052 Erlangen, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Universitätsklinikum Erlangen, 91052 Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.M.)
| | - Geoffrey Lee
- Department of Chemistry and Pharmacy, Division of Pharmaceutics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.M.)
- Correspondence: ; Tel.: +49-9131-85-33142
| |
Collapse
|
221
|
Xu H, Lv S, Jiang S, Lu J, Lin L. Radical scavenging activities of peptide from Asian clam (Corbicula fluminea) and its protective effects on oxidative damage induced by hydrogen peroxide in HepG2 cells. J Food Biochem 2020; 44:e13146. [PMID: 31944325 DOI: 10.1111/jfbc.13146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 12/25/2019] [Accepted: 12/27/2019] [Indexed: 11/29/2022]
Abstract
The soft part of Asian clam (Corbicula fluminea) was hydrolyzed using trypsin and the hydrolysates were separated by ultrafiltration using the membrane with molecular weight cutoff of 10 and 5 kDa. Three fractions (F10000, F10000-5000, and F5000) were obtained. The F5000 showed the strongest scavenging abilities to DPPH (2,2-diphenyl-1-picrylhydrazyl), superoxide anion, and hydroxyl radicals, and was further fractionated into four fractions (component I, II, III, and IV) by gel chromatography. The protective effects of these components on oxidative damage induced by hydrogen peroxide (H2 O2 ) in HepG2 cells were evaluated. Meanwhile, component I separated from F5000 had more positive effects on increasing the activity of catalase, decreasing lipid peroxidation, inhibiting H2 O2 -induced apoptosis, and higher yield. The component I was further separated using anion exchange chromatography and reversed phase high-performance liquid chromatography, and the peptide sequence of Lys-Gly-Pro-Ala- Pro-Phe-Tyr-Pro-Leu was identified by mass with molecular weight of 988.3 Da. PRACTICAL APPLICATIONS: Asian clam (C. fluminea) is used for liver protection in traditional Chinese medicine. The present study investigated the radical scavenging activity of the hydrolysates from C. fluminea and the protective effects of the hydrolysate fractions on oxidative damage induced by H2 O2 in HepG2 cells. In addition, a peptide was purified from the hydrolysate and the amino acid sequence of it was identified. Although chemical antioxidant has some side effects on health, the peptide with antioxidant activity obtained from C. fluminea would have more extensive application in food and nutraceutical.
Collapse
Affiliation(s)
- Hao Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Shun Lv
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.,Key Laboratory for Agricultural Products Processing of Anhui Province, Hefei, China
| | - Shaotong Jiang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.,Key Laboratory for Agricultural Products Processing of Anhui Province, Hefei, China
| | - Jianfeng Lu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.,Key Laboratory for Agricultural Products Processing of Anhui Province, Hefei, China.,Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei, China
| | - Lin Lin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.,Key Laboratory for Agricultural Products Processing of Anhui Province, Hefei, China
| |
Collapse
|
222
|
Carvalho DEL, Oliveira KM, Bomfim LM, Soares MBP, Bezerra DP, Batista AA, Correa RS. Nucleobase Derivatives as Building Blocks to Form Ru(II)-Based Complexes with High Cytotoxicity. ACS OMEGA 2020; 5:122-130. [PMID: 31956759 PMCID: PMC6963899 DOI: 10.1021/acsomega.9b01921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 12/17/2019] [Indexed: 05/04/2023]
Abstract
Two new Ru(II)-based complexes containing 2-thiouracil derivatives, known as 2-thiouracil (2TU) and 6-methyl-2-thiouracil (6m2TU), were synthesized using cis,trans-[RuCl2(PPh3)2(bipy)] as a precursor. The obtained compounds with a general formula trans-[Ru(2TU)(PPh3)2(bipy)]PF6 (1) and trans-[Ru(6m2TU)(PPh3)2(bipy)]PF6 (2) were characterized by analytical techniques such as NMR, UV-vis, and IR spectroscopies, elementary analysis, mass spectrometry, and single-crystal X-ray diffraction. Moreover, the investigation of the complexes-DNA interaction were carried out using spectrophotometric titrations and showed that the complexes present a weak interaction with this biomolecule. The compounds were evaluated against HL-60, K-562, HepG2, and B16-F10 cancer cells and against noncancer cells (PBMCs). The results of the biological assay revealed that complex 2 is more promising than complex 1. Finally, the present study suggests that complexes 1 and 2 causes cell death by apoptosis, significantly increasing the percentage of apoptotic HL-60 cells, in which the compounds altered the cell cycle, reducing the cells in G1/G0, G2/M, and S phases.
Collapse
Affiliation(s)
- Diogo E L Carvalho
- Departamento de Química, Instituto de Ciências Exatas e Biológicas-Campus Morro do Cruzeiro, Universidade Federal de Ouro Preto-UFOP, CEP 35400-000 Ouro Preto, MG, Brazil
| | - Katia M Oliveira
- Departamento de Química, Instituto de Ciências Exatas e Biológicas-Campus Morro do Cruzeiro, Universidade Federal de Ouro Preto-UFOP, CEP 35400-000 Ouro Preto, MG, Brazil
| | - Larissa M Bomfim
- Fundação Oswaldo Cruz, Gonçalo Moniz, Rua Waldemar Falcão, 121, Candeal, CEP 40296-710 Salvador, BA, Brazil
| | - Milena B P Soares
- Fundação Oswaldo Cruz, Gonçalo Moniz, Rua Waldemar Falcão, 121, Candeal, CEP 40296-710 Salvador, BA, Brazil
| | - Daniel P Bezerra
- Fundação Oswaldo Cruz, Gonçalo Moniz, Rua Waldemar Falcão, 121, Candeal, CEP 40296-710 Salvador, BA, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos-UFSCar, Rod. Washington Luís, km 235 - SP-310, CEP 13565-905 São Carlos, SP, Brazil
| | - Rodrigo S Correa
- Departamento de Química, Instituto de Ciências Exatas e Biológicas-Campus Morro do Cruzeiro, Universidade Federal de Ouro Preto-UFOP, CEP 35400-000 Ouro Preto, MG, Brazil
| |
Collapse
|
223
|
Niemelä E, Desai D, Niemi R, Doroszko M, Özliseli E, Kemppainen K, Rahman NA, Sahlgren C, Törnquist K, Eriksson JE, Rosenholm JM. Nanoparticles carrying fingolimod and methotrexate enables targeted induction of apoptosis and immobilization of invasive thyroid cancer. Eur J Pharm Biopharm 2020; 148:1-9. [PMID: 31917332 DOI: 10.1016/j.ejpb.2019.12.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/29/2019] [Accepted: 12/30/2019] [Indexed: 02/08/2023]
Abstract
Metastatic tumors are the main cause of cancer-related death, as the invading cancer cells disrupt normal functions of distant organs and are nearly impossible to eradicate by traditional cancer therapeutics. This is of special concern when the cancer has created multiple metastases and extensive surgery would be too dangerous to execute. Therefore, combination chemotherapy is often the selected treatment form. However, drug cocktails often have severe adverse effects on healthy cells, whereby the development of targeted drug delivery could minimize side-effects of drugs and increase the efficacy of the combination therapy. In this study, we utilized the folate antagonist methotrexate (MTX) as targeting ligand conjugated onto mesoporous silica nanoparticles (MSNs) for selective eradication of folate receptor-expressing invasive thyroid cancer cells. The MSNs was subsequently loaded with the drug fingolimod (FTY720), which has previously been shown to efficiently inhibit proliferation and invasion of aggressive thyroid cancer cells. To assess the efficiency of our carrier system, comprehensive in vitro methods were employed; including flow cytometry, confocal microscopy, viability assays, invasion assay, and label-free imaging techniques. The in vitro results show that MTX-conjugated and FTY720-loaded MSNs potently attenuated both the proliferation and invasion of the cancerous thyroid cells while keeping the off-target effects in normal thyroid cells reasonably low. For a more physiologically relevant in vivo approach we utilized the chick chorioallantoic membrane (CAM) assay, showing decreased invasive behavior of the thyroid derived xenografts and an increased necrotic phenotype compared to tumors that received the free drug cocktail. Thus, the developed multidrug-loaded MSNs effectively induced apoptosis and immobilization of invasive thyroid cancer cells, and could potentially be used as a carrier system for targeted drug delivery for the treatment of diverse forms of aggressive cancers that expresses folate receptors.
Collapse
Affiliation(s)
- E Niemelä
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - D Desai
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - R Niemi
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - M Doroszko
- Institute of Biomedicine, University of Turku, Finland; Department of Immunology, Genetics and Pathology, Section for Neuro-oncology, Uppsala University, Sweden
| | - E Özliseli
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - K Kemppainen
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - N A Rahman
- Institute of Biomedicine, University of Turku, Finland; Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Poland
| | - C Sahlgren
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - K Törnquist
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; Minerva Foundation Institute for Medical Research, Biomedicum, Helsinki, Finland
| | - J E Eriksson
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
| | - J M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
| |
Collapse
|
224
|
Discovery of steroidal lactam conjugates of POPAM-NH2 with potent anticancer activity. Future Med Chem 2020; 12:19-35. [DOI: 10.4155/fmc-2019-0255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim: Steroidal prodrugs of nitrogen mustards such as estramustine and prednimustine have proven effective anticancer agents in clinical use since the 1970s. In this work, we aimed to develop steroidal prodrugs of the novel nitrogen mustard POPAM-NH2. POPAM-NH2 is a melphalan analogue that was coupled with three different steroidal lactams. Methodology: The new conjugates were preclinically tested for anticancer activity against nine human and one rodent cancer experimental models, in vitro and in vivo. Results & conclusion: All the steroidal alkylators showed high antitumor activity, in vitro and in vivo, in the experimental systems tested. Moreover, these hybrid compounds showed by far superior anticancer activity compared with the alkylating agents, melphalan and POPAM-NH2.
Collapse
|
225
|
In vitro and in vivo growth inhibition of human acute promyelocytic leukemia HL-60 cells by Guatteria megalophylla Diels (Annonaceae) leaf essential oil. Biomed Pharmacother 2019; 122:109713. [PMID: 31918282 DOI: 10.1016/j.biopha.2019.109713] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 01/18/2023] Open
Abstract
Guatteria megalophylla Diels (Annonaceae) is an 8-10 m tall tree that grows near streams and is widely spread throughout Colombian, Ecuadorian, Peruvian, Brazilian and Guianese Amazon rainforest. Herein, we investigated for the first time the chemical composition and in vitro and in vivo anti-leukemia potential of G. megalophylla leaf essential oil (EO) using human promyelocytic leukemia HL-60 cells as model. EO was obtained by a hydrodistillation clevenger-type apparatus and characterized quali- and quantitatively by GC-MS and GC-FID, respectively. In vitro cytotoxic potential of EO was evaluated in human cancer cell lines (HL-60, MCF-7 CAL27, HSC-3, HepG2 and HCT116) and in human non-cancer cell line (MRC-5) by Alamar blue method. Annexin V/propidium iodide staining, cell cycle distribution and reactive oxygen species (ROS) were assessed by flow cytometry for HL-60 cells treated with EO. In vivo efficacy of EO (50 and 100 mg/kg) was evaluated in C.B-17 SCID mice with HL-60 cell xenografts. Chemical composition analyses showed spathulenol, γ-muurolene, bicyclogermacrene, β-elemene and δ-elemene as main constituents of assayed sample. EO displayed in vitro cytotoxicity, including anti-leukemia effect with IC50 value of 12.51 μg/mL for HL-60 cells. EO treatment caused augment of phosphatidylserine externalization and DNA fragmentation without increasing of ROS in HL-60 cells. In vivo tumor mass inhibition rates of EO was 16.6-48.8 %. These data indicate anti-leukemia potential of G. megalophylla leaf EO.
Collapse
|
226
|
Kanamori Y, Via LD, Macone A, Canettieri G, Greco A, Toninello A, Agostinelli E. Aged garlic extract and its constituent, S-allyl-L-cysteine, induce the apoptosis of neuroblastoma cancer cells due to mitochondrial membrane depolarization. Exp Ther Med 2019; 19:1511-1521. [PMID: 32010332 PMCID: PMC6966145 DOI: 10.3892/etm.2019.8383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/19/2019] [Indexed: 12/13/2022] Open
Abstract
Aged garlic extract (AGE) has been demonstrated to have therapeutic properties in tumors; however its mechanisms of action have not yet been fully elucidated. A previous study revealed that AGE exerts an anti-proliferative effect on a panel of both sensitive [wild-type (WT)] and multidrug-resistant (MDR) human cancer cells. Following treatment of the cells with AGE, cytofluorimetric analysis revealed the occurrence of dose-dependent mitochondrial membrane depolarization (MMD). In this study, in order to further clarify the mechanisms of action of AGE, the effects of AGE on mitochondria isolated from rat liver mitochondria (RLM) were also examined. AGE induced an effect on the components of the electrochemical gradient (ΔµH+), mitochondrial membrane potential (ΔΨm) and mitochondrial electrochemical gradient (ΔpHm). The mitochondrial membrane dysfunctions of RLM induced by AGE, namely the decrease in both membrane potential and chemical gradient were associated with a higher oxidation of both the endogenous glutathione and pyridine nucleotide content. To confirm the anti-proliferative effects of AGE, experiments were performed on the human neuroblastoma (NB) cancer cells, SJ-N-KP and the MYCN-amplified IMR5 cells, using its derivative S-allyl-L-cysteine (SAC), with the aim of providing evidence of the anticancer activity of this compound and its possible molecular mechanism as regards the induction of cytotoxicity. Following treatment of the cells with SAC at 20 mM, cell viability was determined by MTT assay and apoptosis was detected by flow cytometry, using Annexin V-FITC labeling. The percentages of cells undergoing apoptosis was found to be 48.0% in the SJ-N-KP and 50.1% in the IMR5 cells. By cytofluorimetric analysis, it was suggested that the target of SAC are the mitochondria. Mitochondrial activity was examined by labeling the cells with the probe, 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylimidacarbocyanine iodide (JC-1). Following treatment with SAC at 50 mM, both NB cell lines exhibited a marked increase in MMD. On the whole, the findings of this study indicate that both natural products, AGE and SAC, cause cytotoxicity to tumor cells via the induction of mitochondrial permeability transition (MPT).
Collapse
Affiliation(s)
- Yuta Kanamori
- Department of Biochemical Sciences 'A. Rossi Fanelli', Sapienza University of Rome, I-00185 Rome, Italy
| | - Lisa Dalla Via
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, I-35131 Padua, Italy
| | - Alberto Macone
- Department of Biochemical Sciences 'A. Rossi Fanelli', Sapienza University of Rome, I-00185 Rome, Italy
| | - Gianluca Canettieri
- Pasteur Laboratory, Department of Molecular Medicine, Sapienza University of Rome, I-00161 Rome, Italy.,International Polyamines Foundation-ONLUS, I-00159 Rome, Italy
| | - Antonio Greco
- Department of Sensory Organs, Sapienza University of Rome, I-00161 Rome, Italy
| | - Antonio Toninello
- International Polyamines Foundation-ONLUS, I-00159 Rome, Italy.,Department of Biomedical Sciences, University of Padua, I-35131 Padua, Italy
| | - Enzo Agostinelli
- Department of Biochemical Sciences 'A. Rossi Fanelli', Sapienza University of Rome, I-00185 Rome, Italy.,International Polyamines Foundation-ONLUS, I-00159 Rome, Italy
| |
Collapse
|
227
|
Wawrzyńska M, Duda M, Hołowacz I, Kaczorowska A, Ulatowska-Jarża A, Buzalewicz I, Kałas W, Wysokińska E, Biały D, Podbielska H, Kopaczyńska M. Photoactive Pore Matrix for In Situ Delivery of a Photosensitizer in Vascular Smooth Muscle Cells Selective PDT. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E4110. [PMID: 31818025 PMCID: PMC6947284 DOI: 10.3390/ma12244110] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022]
Abstract
In this study we present the porous silica-based material that can be used for in situ drug delivery, offering effective supply of active compounds regardless its water solubility. To demonstrate usability of this new material, three silica-based materials with different pore size distribution as a matrix for doping with Photolon (Ph) and Protoporphyrin IX (PPIX) photosensitizers, were prepared. These matrices can be used for coating cardiovascular stents used for treatment of the coronary artery disease and enable intravascular photodynamic therapy (PDT), which can modulate the vascular response to injury caused by stent implantation-procedure that should be thought as an alternative for drug eluting stent. The FTIR spectroscopic analysis confirmed that all studied matrices have been successfully functionalized with the target photosensitizers. Atomic force microscopy revealed that resulting photoactive matrices were very smooth, which can limit the implantation damage and reduce the risk of restenosis. No viability loss of human peripheral blood lymphocytes and no erythrocyte hemolysis upon prolonged incubations on matrices indicated good biocompatibility of designed materials. The suitability of photoactive surfaces for PDT was tested in two cell lines relevant to stent implantation: vascular endothelial cells (HUVECs) and vascular smooth muscle cells (VSMC). It was demonstrated that 2 h incubation on the silica matrices was sufficient for uptake of the encapsulated photosensitizers. Moreover, the amount of the absorbed photosensitizer was sufficient for induction of a phototoxic reaction as shown by a rise of the reactive oxygen species in photosensitized VSMC. On the other hand, limited reactive oxygen species (ROS) induction in HUVECs in our experimental set up suggests that the proposed method of PDT may be less harmful for the endothelial cells and may decrease a risk of the restenosis. Presented data clearly demonstrate that porous silica-based matrices are capable of in situ delivery of photosensitizer for PDT of VSMC.
Collapse
Affiliation(s)
- Magdalena Wawrzyńska
- Department of Emergency Medical Service, Wroclaw Medical University, Parkowa 34, 51-616 Wroclaw, Poland;
| | - Maciej Duda
- Department of Biomedical Engineering, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland; (M.D.); (I.H.); (A.K.); (A.U.-J.); (I.B.); (H.P.)
| | - Iwona Hołowacz
- Department of Biomedical Engineering, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland; (M.D.); (I.H.); (A.K.); (A.U.-J.); (I.B.); (H.P.)
| | - Aleksandra Kaczorowska
- Department of Biomedical Engineering, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland; (M.D.); (I.H.); (A.K.); (A.U.-J.); (I.B.); (H.P.)
| | - Agnieszka Ulatowska-Jarża
- Department of Biomedical Engineering, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland; (M.D.); (I.H.); (A.K.); (A.U.-J.); (I.B.); (H.P.)
| | - Igor Buzalewicz
- Department of Biomedical Engineering, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland; (M.D.); (I.H.); (A.K.); (A.U.-J.); (I.B.); (H.P.)
| | - Wojciech Kałas
- Department of Experimental Oncology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Rudolfa Weigla 12, 53-114 Wroclaw, Poland; (W.K.)
| | - Edyta Wysokińska
- Department of Experimental Oncology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Rudolfa Weigla 12, 53-114 Wroclaw, Poland; (W.K.)
| | - Dariusz Biały
- Department and Clinic of Cardiology, Wroclaw Medical University, Borowska 213, 50-556 Wrocław, Poland;
| | - Halina Podbielska
- Department of Biomedical Engineering, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland; (M.D.); (I.H.); (A.K.); (A.U.-J.); (I.B.); (H.P.)
| | - Marta Kopaczyńska
- Department of Biomedical Engineering, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland; (M.D.); (I.H.); (A.K.); (A.U.-J.); (I.B.); (H.P.)
| |
Collapse
|
228
|
Styrylcoumarin 7-SC2 induces apoptosis in SW480 human colon adenocarcinoma cells and inhibits azoxymethane-induced aberrant crypt foci formation in BALB/c mice. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02487-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
229
|
Alupei MC, Maity P, Esser PR, Krikki I, Tuorto F, Parlato R, Penzo M, Schelling A, Laugel V, Montanaro L, Scharffetter-Kochanek K, Iben S. Loss of Proteostasis Is a Pathomechanism in Cockayne Syndrome. Cell Rep 2019; 23:1612-1619. [PMID: 29742419 DOI: 10.1016/j.celrep.2018.04.041] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 03/02/2018] [Accepted: 04/09/2018] [Indexed: 01/04/2023] Open
Abstract
Retarded growth and neurodegeneration are hallmarks of the premature aging disease Cockayne syndrome (CS). Cockayne syndrome proteins take part in the key step of ribosomal biogenesis, transcription of RNA polymerase I. Here, we identify a mechanism originating from a disturbed RNA polymerase I transcription that impacts translational fidelity of the ribosomes and consequently produces misfolded proteins. In cells from CS patients, the misfolded proteins are oxidized by the elevated reactive oxygen species (ROS) and provoke an unfolded protein response that represses RNA polymerase I transcription. This pathomechanism can be disrupted by the addition of pharmacological chaperones, suggesting a treatment strategy for CS. Additionally, this loss of proteostasis was not observed in mouse models of CS.
Collapse
Affiliation(s)
- Marius Costel Alupei
- Clinic of Dermatology and Allergic Diseases, University Medical Center, Albert-Einstein Allee 23, 89081 Ulm, Germany
| | - Pallab Maity
- Clinic of Dermatology and Allergic Diseases, University Medical Center, Albert-Einstein Allee 23, 89081 Ulm, Germany
| | - Philipp Ralf Esser
- Allergy Research Group, Department of Dermatology, University Medical Center Freiburg, Faculty of Medicine, 79104 Freiburg, Germany
| | - Ioanna Krikki
- Clinic of Dermatology and Allergic Diseases, University Medical Center, Albert-Einstein Allee 23, 89081 Ulm, Germany
| | - Francesca Tuorto
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Rosanna Parlato
- Institute of Applied Physiology, Ulm University, 89081 Ulm, Germany; Institute of Anatomy and Medical Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Marianna Penzo
- Laboratorio di Patologia Clinica, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Adrian Schelling
- Clinic of Dermatology and Allergic Diseases, University Medical Center, Albert-Einstein Allee 23, 89081 Ulm, Germany
| | - Vincent Laugel
- Laboratoire de Génétique Médicale - INSERM U1112, Institut de Génétique Médicale d'Alsace (IGMA), Faculté de médecine de Strasbourg, 11 rue Humann, 67000 Strasbourg, France
| | - Lorenzo Montanaro
- Laboratorio di Patologia Clinica, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Karin Scharffetter-Kochanek
- Clinic of Dermatology and Allergic Diseases, University Medical Center, Albert-Einstein Allee 23, 89081 Ulm, Germany
| | - Sebastian Iben
- Clinic of Dermatology and Allergic Diseases, University Medical Center, Albert-Einstein Allee 23, 89081 Ulm, Germany.
| |
Collapse
|
230
|
Zimmermann S, Klinger-Strobel M, Bohnert JA, Wendler S, Rödel J, Pletz MW, Löffler B, Tuchscherr L. Clinically Approved Drugs Inhibit the Staphylococcus aureus Multidrug NorA Efflux Pump and Reduce Biofilm Formation. Front Microbiol 2019; 10:2762. [PMID: 31849901 PMCID: PMC6901667 DOI: 10.3389/fmicb.2019.02762] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/12/2019] [Indexed: 11/26/2022] Open
Abstract
Staphylococcus aureus has acquired resistance to antibiotics since their first use. The S. aureus protein NorA, an efflux pump belonging to the major facilitator superfamily (MFS), contributes to resistance to fluoroquinolones (e.g., ciprofloxacin), biocides, dyes, quaternary ammonium compounds, and antiseptics. Different compounds have been identified as potential efflux pump inhibitors (EPIs) of NorA that result in increased intracellular concentration of antibiotics, restoring their antibacterial activity and cell susceptibility. However, none of the currently known EPIs have been approved for clinical use, probably due to their toxicity profiles. In the present study, we screened approved drugs for possible efflux pump inhibition. By screening a compound library of approximately 1200 different drugs, we identified nilotinib, a tyrosine kinase inhibitor, as showing the best efflux pump inhibitory activity, with a fractional inhibitory concentration index of 0.1875, indicating synergism with ciprofloxacin, and a minimum effective concentration as low as 0.195 μM. Moreover, at 0.39 μM, nilotinib, in combination with 8 μg/mL of ciprofloxacin, led to a significant reduction in biofilm formation and preformed mature biofilms. This is the first description of an approved drug that can be used as an efflux pump inhibitor and to reduce biofilms formation at clinically achievable concentrations.
Collapse
Affiliation(s)
- Saskia Zimmermann
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Mareike Klinger-Strobel
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Jürgen A Bohnert
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany.,Institute of Medical Microbiology, Greifswald University Hospital, Greifswald, Germany
| | - Sindy Wendler
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Jürgen Rödel
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Mathias W Pletz
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Lorena Tuchscherr
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| |
Collapse
|
231
|
Albiero LR, de Andrade MF, Marchi LF, Landi-Librandi AP, de Figueiredo-Rinhel ASG, Carvalho CA, Kabeya LM, de Oliveira RDR, Azzolini AECS, Pupo MT, da Silva Emery F, Lucisano-Valim YM. Immunomodulating action of the 3-phenylcoumarin derivative 6,7-dihydroxy-3-[3',4'-methylenedioxyphenyl]-coumarin in neutrophils from patients with rheumatoid arthritis and in rats with acute joint inflammation. Inflamm Res 2019; 69:115-130. [PMID: 31786615 DOI: 10.1007/s00011-019-01298-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 09/25/2019] [Accepted: 11/06/2019] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE To examine whether free (3-PD-5free) and/or liposomal (3-PD-5lipo) 6,7-dihydroxy-3-[3',4'-methylenedioxyphenyl]-coumarin (3-PD-5) (1) modulate the effector functions of neutrophils from patients with rheumatoid arthritis under remission (i-RA) and with active disease (a-RA), in vitro; and (2) exert anti-inflammatory effect in a rat model of zymosan-induced acute joint inflammation. METHODS AND RESULTS Incorporation of 3-PD-5 into unilamellar liposomes of soya phosphatidylcholine and cholesterol was efficient (57.5 ± 7.9%) and yielded vesicles with low diameter (133.7 ± 18.4 nm), polydispersity index (0.39 ± 0.06), and zeta potential (- 1.22 ± 0.34 mV). 3-PD-5free (1 µM) and 3-PD-5lipo (3 µM) equally suppressed elastase release and reactive oxygen species generation in neutrophils from healthy subjects and i-RA and a-RA patients, stimulated with immune complexes. 3-PD-5free (20 µM) suppressed the release of neutrophil extracellular traps and chemotaxis in vitro, without clear signs of cytotoxicity. 3-PD-5lipo (1.5 mg/kg, i.p.) diminished joint edema and synovial infiltration of total leukocytes and neutrophils, without changing the synovial levels of TNF-α, IL-1β, and IL-6. CONCLUSION Altogether, the results reported herein indicate that 3-PD-5 is a promising modulator of the early stages of acute joint inflammation that can help to diminish not only excessive neutrophil infiltration in the synovia but also neutrophil activation and its outcomes in RA patients.
Collapse
Affiliation(s)
- Lucinéia Reuse Albiero
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brazil. .,Federal University of Mato Grosso, Sinop, MT, Brazil.
| | - Micássio Fernandes de Andrade
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brazil. .,School of Health Sciences, The State University of Rio Grande do Norte, Mossoró, RN, Brazil.
| | - Larissa Fávaro Marchi
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Ana Paula Landi-Librandi
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Andréa Silva Garcia de Figueiredo-Rinhel
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Camila Andressa Carvalho
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Luciana Mariko Kabeya
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Renê Donizeti Ribeiro de Oliveira
- Division of Rheumatology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brazil
| | - Ana Elisa Caleiro Seixas Azzolini
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Mônica Tallarico Pupo
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Flávio da Silva Emery
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Yara Maria Lucisano-Valim
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
232
|
Maackia amurensis agglutinin induces apoptosis in cultured drug resistant human non-small cell lung cancer cells. Glycoconj J 2019; 36:473-485. [DOI: 10.1007/s10719-019-09891-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/15/2019] [Accepted: 09/09/2019] [Indexed: 12/21/2022]
|
233
|
Dance Training Improves Cytokine Secretion and Viability of Neutrophils in Diabetic Patients. Mediators Inflamm 2019; 2019:2924818. [PMID: 31827375 PMCID: PMC6886327 DOI: 10.1155/2019/2924818] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/14/2019] [Accepted: 10/24/2019] [Indexed: 12/13/2022] Open
Abstract
Background Evidence suggests that exercise improves neutrophil function. The decreased functional longevity of neutrophils and their increased clearance from infectious sites contribute to the increased susceptibility to infection and severity of infection observed in patients with diabetes. Objective Herein, we investigated the effects of a dance program on neutrophil number, function, and death in type 2 diabetes mellitus (T2DM) patients and healthy volunteers. Methods Ten patients with T2DM and twelve healthy individuals participated in a moderate-intensity dance training program for 4 months. The plasma levels of leptin, free fatty acids (FFAs), tumour necrosis factor-α (TNF-α), C-reactive protein (CRP), interleukin-1β (IL-1β), and interleukin-1 receptor antagonist (IL-1ra); neutrophil counts; extent of DNA fragmentation; cell membrane integrity; and production of TNF-α, interleukin-8 (IL-8), interleukin-6 (IL-6), and IL-1β in neutrophils were measured before and after training. Results Training reduced plasma levels of TNF-α (1.9-fold in controls and 2.2-fold in patients with T2DM) and CRP (1.4-fold in controls and 3.4-fold in patients with T2DM). IL-1ra levels were higher in the control group (2.2-fold) after training. After training, neutrophil DNA fragmentation was decreased in patients with T2DM (90%), while the number of neutrophils increased (70% in controls and 1.1-fold in patients with T2DM). Conclusion Dance training is a nonpharmacological strategy to reduce inflammation and improve neutrophil clearance in patients with T2DM.
Collapse
|
234
|
Development of a Stromal Microenvironment Experimental Model Containing Proto-Myofibroblast Like Cells and Analysis of Its Crosstalk with Melanoma Cells: A New Tool to Potentiate and Stabilize Tumor Suppressor Phenotype of Dermal Myofibroblasts. Cells 2019; 8:cells8111435. [PMID: 31739477 PMCID: PMC6912587 DOI: 10.3390/cells8111435] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022] Open
Abstract
Melanoma is one of the most aggressive solid tumors and includes a stromal microenvironment that regulates cancer growth and progression. The components of stromal microenvironment such as fibroblasts, fibroblast aggregates and cancer-associated fibroblasts (CAFs) can differently influence the melanoma growth during its distinct stages. In this work, we have developed and studied a stromal microenvironment model, represented by fibroblasts, proto-myofibroblasts, myofibroblasts and aggregates of inactivated myofibroblasts, such as spheroids. In particular, we have generated proto-myofibroblasts from primary cutaneous myofibroblasts. The phenotype of proto-myofibroblasts is characterized by a dramatic reduction of α-smooth muscle actin (α-SMA) and cyclooxygenase-2 (COX-2) protein levels, as well as an enhancement of cell viability and migratory capability compared with myofibroblasts. Furthermore, proto-myofibroblasts display the mesenchymal marker vimentin and less developed stress fibers, with respect to myofibroblasts. The analysis of crosstalk between the stromal microenvironment and A375 or A2058 melanoma cells has shown that the conditioned medium of proto-myofibroblasts is cytotoxic, mainly for A2058 cells, and dramatically reduces the migratory capability of both cell lines compared with the melanoma-control conditioned medium. An array analysis of proto-myofibroblast and melanoma cell-conditioned media suggests that lower levels of some cytokines and growth factors in the conditioned medium of proto-myofibroblasts could be associated with their anti-tumor activity. Conversely, the conditioned media of melanoma cells do not influence the cell viability, outgrowth, and migration of proto-myofibroblasts from spheroids. Interestingly, the conditioned medium of proto-myofibroblasts does not alter the cell viability of both BJ-5ta fibroblast cells and myofibroblasts. Hence, proto-myofibroblasts could be useful in the study of new therapeutic strategies targeting melanoma.
Collapse
|
235
|
Frattaruolo L, Fiorillo M, Brindisi M, Curcio R, Dolce V, Lacret R, Truman AW, Sotgia F, Lisanti MP, Cappello AR. Thioalbamide, A Thioamidated Peptide from Amycolatopsis alba, Affects Tumor Growth and Stemness by Inducing Metabolic Dysfunction and Oxidative Stress. Cells 2019; 8:cells8111408. [PMID: 31717378 PMCID: PMC6912574 DOI: 10.3390/cells8111408] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022] Open
Abstract
Thioalbamide, a thioamidated peptide biosynthesized by Amycolatopsis alba, is a thioviridamide-like molecule, and is part of a family of natural products representing a focus of biotechnological and pharmaceutical research in recent years due to their potent anti-proliferative and cytotoxic activities on malignant cells. Despite the high antitumor potential observed at nanomolar concentrations, the mechanisms underlying thioalbamide activity are still not known. In this work, the cellular effects induced by thioalbamide treatment on breast cancer cell lines were evaluated for the first time, highlighting the ability of this microbial natural peptide to induce mitochondrial dysfunction, oxidative stress, and apoptotic cell death. Furthermore, we demonstrate that thioalbamide can inhibit the propagation of cancer stem-like cells, which are strongly dependent on mitochondrial function and are responsible for chemotherapy resistance, metastasis, and tumor recurrence.
Collapse
Affiliation(s)
- Luca Frattaruolo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy; (L.F.); (M.F.); (M.B.); (R.C.); (V.D.)
| | - Marco Fiorillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy; (L.F.); (M.F.); (M.B.); (R.C.); (V.D.)
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC), University of Salford, Greater Manchester M5 4WT, UK
| | - Matteo Brindisi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy; (L.F.); (M.F.); (M.B.); (R.C.); (V.D.)
| | - Rosita Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy; (L.F.); (M.F.); (M.B.); (R.C.); (V.D.)
| | - Vincenza Dolce
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy; (L.F.); (M.F.); (M.B.); (R.C.); (V.D.)
| | - Rodney Lacret
- Department of Molecular Microbiology, John Innes Centre, Colney Lane, Norwich NR4 7UH, UK; (R.L.); (A.W.T.)
| | - Andrew W. Truman
- Department of Molecular Microbiology, John Innes Centre, Colney Lane, Norwich NR4 7UH, UK; (R.L.); (A.W.T.)
| | - Federica Sotgia
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC), University of Salford, Greater Manchester M5 4WT, UK
- Correspondence: (F.S.); (M.P.L.); (A.R.C.)
| | - Michael P. Lisanti
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC), University of Salford, Greater Manchester M5 4WT, UK
- Correspondence: (F.S.); (M.P.L.); (A.R.C.)
| | - Anna Rita Cappello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy; (L.F.); (M.F.); (M.B.); (R.C.); (V.D.)
- Correspondence: (F.S.); (M.P.L.); (A.R.C.)
| |
Collapse
|
236
|
Yudhistiara B, Weber KJ, Huber PE, Ruehle A, Brons S, Haering P, Debus J, Hauswald H. Carbon ion and proton beam irradiation of a normal human TK6 lymphoblastoid cell line within a magnetic field of 1.0 tesla. Cancer Manag Res 2019; 11:8327-8335. [PMID: 31686914 PMCID: PMC6751770 DOI: 10.2147/cmar.s212310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/11/2019] [Indexed: 01/04/2023] Open
Abstract
Background Considering the increasing simultaneous application of magnetic resonance imaging (MRI) for more precise photon radiotherapy, it will be likely for particle radiotherapy to adopt MRI for future image guiding. It will then be imperative to evaluate the potential biological effects of a magnetic field (MF) on particle irradiation. This study explores such effects on the highly radiosensitive TK6 lymphoblastoid human cell line. Methods The following three parameters were measured after irradiation with either carbon ion or proton beams using spread out Bragg peaks and applying different doses within a perpendicular 1.0 T MF: (1) cell survival fraction (14 days postirradiation), (2) treatment-specific apoptosis, which was determined through the measurement of population in the sub-G1 phase, and (3) cell cycle progression by means of flow cytometry. These were compared to the same parameters measured without an MF. Results The clonogenic assay in both treatment groups showed almost identical survival curves with overlapping error bars. The calculated α values with and without an MF were 2.18 (σ=0.245) and 2.17 (σ=0.234) for carbon ions and 1.08 (σ=0.138) and 1.13 (σ=0.0679) for protons, respectively. Similarly, the treatment-specific apoptosis and cell cycle progression showed almost identical curves with overlapping error bars. A two-sample, unpooled t-test analysis was implemented for comparison of all mean values and showed p-values >0.05. Conclusion No statistically significant difference in biological response of the TK6 cells was observed when they were irradiated using spreadout Bragg peaks within a perpendicular 1.0 T MF as compared to those, which received the same dose without the MF. This should serve as another supporting piece of evidence toward the implementation of MRI in particle radiotherapy, though further research is necessary.
Collapse
Affiliation(s)
- B Yudhistiara
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg 69120, Germany.,National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| | - K J Weber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg 69120, Germany.,National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| | - P E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg 69120, Germany.,Clinical Cooperation Unit Molecular Radiation Oncology E055, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A Ruehle
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg 69120, Germany.,Clinical Cooperation Unit Molecular Radiation Oncology E055, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - S Brons
- National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg 69120, Germany
| | - P Haering
- Department of Radiation Physics E040, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - J Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg 69120, Germany.,National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg 69120, Germany.,Clinical Cooperation Unit E050, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - H Hauswald
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg 69120, Germany.,National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg 69120, Germany.,Clinical Cooperation Unit E050, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
237
|
In vitro assessment of the cytotoxicity of Gallium(III) complexes with Isoniazid-Derived Hydrazones: Effects on clonogenic survival of HCT-116 cells. Inorganica Chim Acta 2019. [DOI: 10.1016/j.ica.2019.119079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
238
|
De Stefanis D, Scimè S, Accomazzo S, Catti A, Occhipinti A, Bertea CM, Costelli P. Anti-Proliferative Effects of an Extra-Virgin Olive Oil Extract Enriched in Ligstroside Aglycone and Oleocanthal on Human Liver Cancer Cell Lines. Cancers (Basel) 2019; 11:cancers11111640. [PMID: 31653043 PMCID: PMC6896128 DOI: 10.3390/cancers11111640] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/27/2022] Open
Abstract
Oleocanthal and ligstroside aglycone are olive oil-derived polyphenols. The former interferes with tumor growth with minor or no cytotoxicity on non-tumorigenic primary cell lines. The information about the bioactivity of ligstroside aglycone are scanty, with the exception of a known antioxidant power. Hepatocellular carcinoma is a malignant tumor with high mortality rates. Systemic chemotherapy is only marginally effective and is frequently complicated by toxicity. Previous observations have shown that hepatocellular carcinoma cell lines become more sensitive to taxol when it is combined with Tumor Necrosis Factor α (TNFα). The present work aimed to assess the effects of a polyphenolic extract containing both oleocanthal and ligstroside aglycone on proliferation and/or death in three liver cancer cell lines (HepG2, Huh7 and Hep3B). The possibility to enhance such effect by the addition of TNFα was also investigated. Both cell proliferation and death were enhanced by the exposure to the polyphenolic extract. Such effect was associated with induction of autophagy and could be potentiated by TNFα. The presence of ligstroside aglycone in the extract lowered the oleocanthal concentration required for cytotoxicity. These results show for the first time that the effects of a polyphenol extract can be potentiated by TNFα and that modulation of autophagy likely account for these effects.
Collapse
Affiliation(s)
- Daniela De Stefanis
- Department of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, University of Turin, 10125 Torino, Italy.
| | - Salvatore Scimè
- Department of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, University of Turin, 10125 Torino, Italy.
| | - Simone Accomazzo
- Department of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, University of Turin, 10125 Torino, Italy.
| | - Andrea Catti
- Department of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, University of Turin, 10125 Torino, Italy.
| | - Andrea Occhipinti
- Department of Life Sciences and Systems Biology, University of Turin, 10125 Torino, Italy.
| | | | - Paola Costelli
- Department of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, University of Turin, 10125 Torino, Italy.
| |
Collapse
|
239
|
Chen W, Li S, Kulkarni AS, Huang L, Cao J, Qian K, Wan J. Single Cell Omics: From Assay Design to Biomedical Application. Biotechnol J 2019; 15:e1900262. [DOI: 10.1002/biot.201900262] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/11/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Wenyi Chen
- School of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200241 P. R. China
| | - Shumin Li
- School of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200241 P. R. China
| | - Anuja Shreeram Kulkarni
- School of Biomedical EngineeringMed‐X Research InstituteShanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Lin Huang
- School of Biomedical EngineeringMed‐X Research InstituteShanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Jing Cao
- School of Biomedical EngineeringMed‐X Research InstituteShanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Kun Qian
- School of Biomedical EngineeringMed‐X Research InstituteShanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Jingjing Wan
- School of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200241 P. R. China
| |
Collapse
|
240
|
Bartel K, Müller R, von Schwarzenberg K. Differential regulation of AMP-activated protein kinase in healthy and cancer cells explains why V-ATPase inhibition selectively kills cancer cells. J Biol Chem 2019; 294:17239-17248. [PMID: 31604821 DOI: 10.1074/jbc.ra119.010243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/16/2019] [Indexed: 01/02/2023] Open
Abstract
The cellular energy sensor AMP-activated protein kinase (AMPK) is a metabolic hub regulating various pathways involved in tumor metabolism. Here we report that vacuolar H+-ATPase (V-ATPase) inhibition differentially affects regulation of AMPK in tumor and nontumor cells and that this differential regulation contributes to the selectivity of V-ATPase inhibitors for tumor cells. In nonmalignant cells, the V-ATPase inhibitor archazolid increased phosphorylation and lysosomal localization of AMPK. We noted that AMPK localization has a prosurvival role, as AMPK silencing decreased cellular growth rates. In contrast, in cancer cells, we found that AMPK is constitutively active and that archazolid does not affect its phosphorylation and localization. Moreover, V-ATPase-independent AMPK induction in tumor cells protected them from archazolid-induced cytotoxicity, further underlining the role of AMPK as a prosurvival mediator. These observations indicate that AMPK regulation is uncoupled from V-ATPase activity in cancer cells and that this makes them more susceptible to cell death induction by V-ATPase inhibitors. In both tumor and healthy cells, V-ATPase inhibition induced a distinct metabolic regulatory cascade downstream of AMPK, affecting ATP and NADPH levels, glucose uptake, and reactive oxygen species production. We could attribute the prosurvival effects to AMPK's ability to maintain redox homeostasis by inhibiting reactive oxygen species production and maintaining NADPH levels. In summary, the results of our work indicate that V-ATPase inhibition has differential effects on AMPK-mediated metabolic regulation in cancer and healthy cells and explain the tumor-specific cytotoxicity of V-ATPase inhibition.
Collapse
Affiliation(s)
- Karin Bartel
- Department of Pharmacy, Pharmaceutical Biology, Ludwig Maximilians University, 81377 Munich, Germany
| | - Rolf Müller
- Helmholtz Center for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, and Department of Pharmacy at Saarland University, Saarland University Campus, 66123 Saarbrücken, Germany
| | - Karin von Schwarzenberg
- Department of Pharmacy, Pharmaceutical Biology, Ludwig Maximilians University, 81377 Munich, Germany
| |
Collapse
|
241
|
Kinase-Based Screening of Marine Natural Extracts Leads to the Identification of a Cytotoxic High Molecular Weight Metabolite from the Mediterranean Sponge Crambe tailliezi. Mar Drugs 2019; 17:md17100569. [PMID: 31600933 PMCID: PMC6836018 DOI: 10.3390/md17100569] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/18/2022] Open
Abstract
Regulated cell death (RCD) results from the activation of one or more signal transduction modules both in physiological or pathological conditions. It is now established that RCD is involved in numerous human diseases, including cancer. As regulated cell death processes can be modulated by pharmacological tools, the research reported here aims to characterize new marine compounds acting as RCD modulators. Protein kinases (PKs) are key signaling actors in various RCDs notably through the control of either mitosis (e.g., the PKs Aurora A and B) or necroptosis (e.g., RIPK1 and RIPK3). From the primary screening of 27 various extracts of marine organisms collected in the Mediterranean Sea, an extract and subsequently a purified high molecular weight compound dubbed P3, were isolated from the marine sponge Crambe tailliezi and characterized as a selective inhibitor of PKs Aurora A and B. Furthermore, P3 was shown to induce apoptosis and to decrease proliferation and mitotic index of human osteosarcoma U-2 OS cells.
Collapse
|
242
|
Mlejnek P, Dolezel P, Maier V, Kikalova K, Skoupa N. N-acetylcysteine dual and antagonistic effect on cadmium cytotoxicity in human leukemia cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 71:103213. [PMID: 31288199 DOI: 10.1016/j.etap.2019.103213] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/15/2019] [Accepted: 06/18/2019] [Indexed: 06/09/2023]
Abstract
Although cadmium (Cd2+) is unable to form reactive oxygen species (ROS) directly, many of its adverse effects are connected to increased ROS generation resulting in cell death. In support of this supposition, a large number of studies have shown protective effects of antioxidants such as N-acetylcysteine (NAC) against cadmium induced cytotoxicity. Here, we describe the cytotoxic effects of Cd2+ on human leukemia U937 and K562 cells that were not mediated by oxidative stress. Surprisingly, we observed that addition of low concentrations of NAC can drastically potentiate cadmium cytotoxicity solely via ROS production. However, all adverse effects of the metal were prevented by NAC at high concentrations. Detailed analysis indicated that the protective effect of NAC was mediated by its ability to form stable complex with cadmium [Cd(NAC)2]. In conclusion, NAC exhibits dual and antagonistic effects on Cd2+ cytotoxicity in human leukemia cells.
Collapse
Affiliation(s)
- P Mlejnek
- Department of Anatomy, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, Olomouc 77515, Czech Republic.
| | - P Dolezel
- Department of Anatomy, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, Olomouc 77515, Czech Republic
| | - V Maier
- Department of Anatomy, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, Olomouc 77515, Czech Republic
| | - K Kikalova
- Department of Anatomy, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, Olomouc 77515, Czech Republic
| | - N Skoupa
- Department of Anatomy, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, Olomouc 77515, Czech Republic
| |
Collapse
|
243
|
Othman DI, Selim KB, El-Sayed MAA, Tantawy AS, Amen Y, Shimizu K, Okauchi T, Kitamura M. Design, Synthesis and Anticancer Evaluation of New Substituted Thiophene-Quinoline Derivatives. Bioorg Med Chem 2019; 27:115026. [DOI: 10.1016/j.bmc.2019.07.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 10/26/2022]
|
244
|
Leu YS, Chen YJ, Chen CC, Huang HL. Induction of Autophagic Death of Human Hepatocellular Carcinoma Cells by Armillaridin from Armillaria mellea. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:1365-1380. [DOI: 10.1142/s0192415x19500708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The honey mushroom, Armillaria mellea, is known to have medicinal qualities and has been used in recent years as a health food and dietary supplement worldwide. In Asia, it is commonly consumed as an herbal medicine, being a key component of the Chinese preparation “Tien-ma”. Here, we examined the antitumor effects of armillaridin, a bioactive compound isolated from A. mellea, on human hepatocellular carcinoma (HCC) cells. Armillaridin inhibited the growth of human Huh7, HepG2, and HA22T HCC cells, and its cytotoxicity was confirmed by observations of its induction of mitochondrial transmembrane potential collapse. However, armillaridin treatment did not result in large numbers of cells with fragmented chromosomal DNA, suggesting that apoptosis was not responsible for these effects. We therefore tested for signs of autophagic cell death following armillaridin administration. Armillaridin induced LC3 aggregation in green fluorescent protein-LC3-overexpressing cells. Moreover, flow cytometry and immunoblotting revealed that it increased the number of acridine orange-positive cells and upregulated autophagy-related proteins, respectively. Furthermore, armillaridin cytotoxicity was suppressed by the autophagy inhibitor 3-methyladenine. In summary, our results indicated that armillaridin induces HCC cell death by autophagy, and demonstrated the potential of armillaridin as an antihepatoma agent.
Collapse
Affiliation(s)
- Yi-Shing Leu
- Department of Otolaryngology, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Yu-Jen Chen
- Department of Medical Research and Department of Radiation Oncology, Mackay Memorial Hospital, Taipei, Taiwan, ROC
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Institute of Pharmacology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Chien-Chih Chen
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Huey-Lan Huang
- Department of Bioscience Technology, College of Health Science, Chang Jung Christian University, Tainan, Taiwan, ROC
| |
Collapse
|
245
|
Gentile EA, Castronuovo CC, Cuestas ML, Gómez N, Davio C, Oubiña JR, Mathet VL. F127 poloxamer effect on cytotoxicity induction of tumour cell cultures treated with doxorubicin. ACTA ACUST UNITED AC 2019; 71:1655-1662. [PMID: 31456253 DOI: 10.1111/jphp.13158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/28/2019] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Hepatocellular carcinoma is the most common liver malignancy and the third leading cause of cancer death worldwide. One crucial limitation in the pharmacotherapy for this tumour is its chemotherapy-resistant nature produced by the overexpression of several members of the ATP-binding cassette protein family that efflux drugs out of cells, as observed with the breast cancer resistant protein (BCRP). OBJECTIVES This study aimed to assess the ability of Pluronic® F127 to reverse the multidrug resistance phenotype in two human hepatocellular cell lines. METHODS PLC/PRF/5 and SKHep1 cells were exposed to Pluronic® F127 at several concentrations. The effect of F127 on BCRP expression (mRNA and protein), mitochondrial transmembrane potential and cell hypodiploidy was assessed. Finally, the effect of this copolymer on cytotoxicity of doxorubicin in both hepatoma cell lines was investigated, as expressed by its reverse resistance index. KEY FINDINGS It was demonstrated that F127 in both cell lines contributes to chemosensitization, as shown by BCRP down-regulation, an altered mitochondrial transmembrane potential and hypodiploidy and reverse resistance index values. A remarkable dependence of these effects significantly correlated with the copolymer concentration. CONCLUSIONS These findings further uncover the potential usefulness of this copolymer as multidrug resistance reversal agent, increasing the efficacy of cancer therapies.
Collapse
Affiliation(s)
- Emiliano Alberto Gentile
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPAM), Ciudad Autónoma de Buenos Aires, Argentina
| | - Cynthia Cecilia Castronuovo
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPAM), Ciudad Autónoma de Buenos Aires, Argentina
| | - María Luján Cuestas
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPAM), Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia Gómez
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Carlos Davio
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina
| | - José Raúl Oubiña
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPAM), Ciudad Autónoma de Buenos Aires, Argentina
| | - Verónica Lidia Mathet
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPAM), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
246
|
Sagasser J, Ma BN, Baecker D, Salcher S, Hermann M, Lamprecht J, Angerer S, Obexer P, Kircher B, Gust R. A New Approach in Cancer Treatment: Discovery of Chlorido[ N, N'-disalicylidene-1,2-phenylenediamine]iron(III) Complexes as Ferroptosis Inducers. J Med Chem 2019; 62:8053-8061. [PMID: 31369259 DOI: 10.1021/acs.jmedchem.9b00814] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chlorido[N,N'-disalicylidene-1,2-phenylenediamine]iron(III) complexes generate lipid-based ROS and induce ferroptosis in leukemia and neuroblastoma cell lines. The extent of ferroptosis on the mode of action is regulated by simple modifications of the substituents at the 1,2-phenylenediamine moiety. In HL-60 cells, the unsubstituted lead exclusively caused ferroptosis. For instance, a 4-F substituent shifted the mode of action toward both ferroptosis and necroptosis, while the analogously chlorinated derivative exerted only necroptosis. Remarkably, cell-death in NB1 neuroblastoma cells was solely induced by ferroptosis, independent of the used substituents. The effects were higher than that of the therapeutically applied drug cisplatin. These data clearly demonstrate for the first time that not only iron ions but also iron salophene complexes are potent ferroptosis inducers, which can be optimized as antitumor agents.
Collapse
Affiliation(s)
- Jessica Sagasser
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck , University of Innsbruck, CCB-Center for Chemistry and Biomedicine , Innrain 80-82 , 6020 Innsbruck , Austria
| | - Benjamin N Ma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck , University of Innsbruck, CCB-Center for Chemistry and Biomedicine , Innrain 80-82 , 6020 Innsbruck , Austria
| | - Daniel Baecker
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck , University of Innsbruck, CCB-Center for Chemistry and Biomedicine , Innrain 80-82 , 6020 Innsbruck , Austria
| | - Stefan Salcher
- Tyrolean Cancer Research Institute , Innrain 66 , 6020 Innsbruck , Austria
| | - Martin Hermann
- Department of Anesthesiology and Critical Care Medicine , Medical University Innsbruck , Anichstraße 35 , 6020 Innsbruck , Austria
| | - Julia Lamprecht
- Tyrolean Cancer Research Institute , Innrain 66 , 6020 Innsbruck , Austria
| | - Stefanie Angerer
- Tyrolean Cancer Research Institute , Innrain 66 , 6020 Innsbruck , Austria.,Immunobiology and Stem Cell Laboratory, Department of Internal Medicine V (Hematology and Oncology) , Medical University Innsbruck , Anichstraße 35 , 6020 Innsbruck , Austria
| | - Petra Obexer
- Tyrolean Cancer Research Institute , Innrain 66 , 6020 Innsbruck , Austria.,Department of Pediatrics II , Medical University Innsbruck , Innrain 66 , 6020 Innsbruck , Austria
| | - Brigitte Kircher
- Tyrolean Cancer Research Institute , Innrain 66 , 6020 Innsbruck , Austria.,Immunobiology and Stem Cell Laboratory, Department of Internal Medicine V (Hematology and Oncology) , Medical University Innsbruck , Anichstraße 35 , 6020 Innsbruck , Austria
| | - Ronald Gust
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck , University of Innsbruck, CCB-Center for Chemistry and Biomedicine , Innrain 80-82 , 6020 Innsbruck , Austria
| |
Collapse
|
247
|
Tayarani-Najaran Z, Akaberi M, Hassanzadeh B, Shirazi N, Asili J, Al-Najjar H, Sahebkar A, Emami SA. Analysis of the Essential Oils of Five Artemisia Species and Evaluation of their Cytotoxic and Proapoptotic Effects. Mini Rev Med Chem 2019; 19:902-912. [PMID: 30864526 DOI: 10.2174/1389557519666190311155021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 04/24/2018] [Accepted: 11/11/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND In this study, the essential oil composition and cytotoxic activities of five Artemisia species were determined. METHODS The collected plants were water-distilled separately to obtain oils which were then subjected to gas chromatography (GC) and gas chromatography/mass spectrometry GC/MS analyses to identify their compositions. Cancer cells were exposed to different concentrations of samples and cell viability was measured using AlamarBlue® assay. Apoptotic cells were analyzed by propidium iodide (PI) staining and flow cytometry. RESULTS & CONCLUSION To study the amount of pro-apoptotic proteins and the apoptosis mechanism, Western blot method was used. Although all samples were cytotoxic at the highest concentration, the oil of A. kulbadica showed the strongest activity among other plants. Carvacrol (IC50 21.11 μg/ml) had the most cytotoxic effects among other components. Carvacrol, 1,8-cineole and 4-terpineole caused an increase in the amount of Bax protein and cleaved peroxisome proliferator-activated receptors (PPAR) and caspase proteins in DU 145 cells.
Collapse
Affiliation(s)
- Zahra Tayarani-Najaran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Akaberi
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahareh Hassanzadeh
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nazila Shirazi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Asili
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hadi Al-Najjar
- Department of Pharmacy, College of Health Science, Public Authority for Applied Education and Training (PAAET), Kuwait Health Science Pharmacy, Jabriya, Kuwait
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Ahmad Emami
- Department of Traditional Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
248
|
Oellerich T, Schneider C, Thomas D, Knecht KM, Buzovetsky O, Kaderali L, Schliemann C, Bohnenberger H, Angenendt L, Hartmann W, Wardelmann E, Rothenburger T, Mohr S, Scheich S, Comoglio F, Wilke A, Ströbel P, Serve H, Michaelis M, Ferreirós N, Geisslinger G, Xiong Y, Keppler OT, Cinatl J. Selective inactivation of hypomethylating agents by SAMHD1 provides a rationale for therapeutic stratification in AML. Nat Commun 2019; 10:3475. [PMID: 31375673 PMCID: PMC6677770 DOI: 10.1038/s41467-019-11413-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 07/08/2019] [Indexed: 02/08/2023] Open
Abstract
Hypomethylating agents decitabine and azacytidine are regarded as interchangeable in the treatment of acute myeloid leukemia (AML). However, their mechanisms of action remain incompletely understood, and predictive biomarkers for HMA efficacy are lacking. Here, we show that the bioactive metabolite decitabine triphosphate, but not azacytidine triphosphate, functions as activator and substrate of the triphosphohydrolase SAMHD1 and is subject to SAMHD1-mediated inactivation. Retrospective immunohistochemical analysis of bone marrow specimens from AML patients at diagnosis revealed that SAMHD1 expression in leukemic cells inversely correlates with clinical response to decitabine, but not to azacytidine. SAMHD1 ablation increases the antileukemic activity of decitabine in AML cell lines, primary leukemic blasts, and xenograft models. AML cells acquire resistance to decitabine partly by SAMHD1 up-regulation. Together, our data suggest that SAMHD1 is a biomarker for the stratified use of hypomethylating agents in AML patients and a potential target for the treatment of decitabine-resistant leukemia. In acute myeloid leukemia, hypomethylating agents decitabine and azacytidine are used interchangeably. Here, the authors show that the major metabolite of decitabine, but not azacytidine, is subject to SAMHD1 inactivation, highlighting SAMHD1 as a potential biomarker and therapeutic target
Collapse
Affiliation(s)
- Thomas Oellerich
- Department of Medicine II, Hematology/Oncology, Goethe University of Frankfurt, Frankfurt, 60590, Germany.,German Cancer Consortium/German Cancer Research Center, Heidelberg, 69120, Germany.,Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, 60596, Germany
| | - Constanze Schneider
- Department of Medicine II, Hematology/Oncology, Goethe University of Frankfurt, Frankfurt, 60590, Germany.,Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, 60596, Germany.,Institute of Medical Virology, University of Frankfurt, Frankfurt, 60590, Germany
| | - Dominique Thomas
- pharmazentrum frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University of Frankfurt, Frankfurt, 60590, Germany
| | - Kirsten M Knecht
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Olga Buzovetsky
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Lars Kaderali
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, 17475, Germany
| | | | | | - Linus Angenendt
- Department of Medicine A, University Hospital Münster, Münster, 48149, Germany
| | - Wolfgang Hartmann
- Gerhard Domagk Institute for Pathology, University Hospital Münster, Münster, 48149, Germany
| | - Eva Wardelmann
- Gerhard Domagk Institute for Pathology, University Hospital Münster, Münster, 48149, Germany
| | - Tamara Rothenburger
- Institute of Medical Virology, University of Frankfurt, Frankfurt, 60590, Germany
| | - Sebastian Mohr
- Department of Medicine II, Hematology/Oncology, Goethe University of Frankfurt, Frankfurt, 60590, Germany
| | - Sebastian Scheich
- Department of Medicine II, Hematology/Oncology, Goethe University of Frankfurt, Frankfurt, 60590, Germany
| | - Federico Comoglio
- Department of Haematology, Cambridge Institute of Medical Research, Cambridge University, Cambridge, CB2 0XY, UK
| | - Anne Wilke
- Department of Medicine II, Hematology/Oncology, Goethe University of Frankfurt, Frankfurt, 60590, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center, Göttingen, 37075, Germany
| | - Hubert Serve
- Department of Medicine II, Hematology/Oncology, Goethe University of Frankfurt, Frankfurt, 60590, Germany.,German Cancer Consortium/German Cancer Research Center, Heidelberg, 69120, Germany.,Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, 60596, Germany
| | - Martin Michaelis
- Industrial Biotechnology Centre and School of Biosciences, University of Kent, Canterbury, CT2 7NJ, UK
| | - Nerea Ferreirós
- pharmazentrum frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University of Frankfurt, Frankfurt, 60590, Germany
| | - Gerd Geisslinger
- pharmazentrum frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University of Frankfurt, Frankfurt, 60590, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Project group Translational Medicine and Pharmacology (TMP), Frankfurt, 60596, Germany
| | - Yong Xiong
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Oliver T Keppler
- Institute of Medical Virology, University of Frankfurt, Frankfurt, 60590, Germany. .,Max von Pettenkofer Institute, Virology, Faculty of Medicine, LMU München, Munich, 80336, Germany.
| | - Jindrich Cinatl
- Institute of Medical Virology, University of Frankfurt, Frankfurt, 60590, Germany.
| |
Collapse
|
249
|
Amiri Tehranizadeh Z, Sankian M, Fazly Bazzaz BS, Chamani J, Mehri S, Baratian A, Saberi MR. The immunotoxin activity of exotoxin A is sensitive to domain modifications. Int J Biol Macromol 2019; 134:1120-1131. [PMID: 31129209 DOI: 10.1016/j.ijbiomac.2019.05.137] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/30/2019] [Accepted: 05/21/2019] [Indexed: 11/29/2022]
Abstract
Immunotoxins are a class of recombinant proteins which consist of an antibody and a part of a bacterial or herbal toxin. Immunotoxins containing Pseudomonas aeruginosa exotoxin A (PEA) have been found to be very applicable in clinical trials. Many obstacles such as solubility and absorbency reduce their usability in solid tumors. The current study aims to overcome the mentioned barriers by addition and removal of functional and non-functional domains with a structural approach. In the experimental section, we took advantage of molecular dynamics simulations to predict the functionality of candidate immunotoxins which target human HER2 receptors and confirmed our findings with in vitro experiments. We found out when no changes were made to domain II of PEA, addition of solubilizing domains to immunotoxins would not reduce their targeting and anti-tumor activity, while increasing the yield of expression and stability. On the other side, when we replaced domain II with eleven amino acids of furin cleavage site (FCS), the activity of the immunotoxin was mainly affected by the FCS neighboring domains and linkers. A combination of seven beneficial point mutations in domain III was also assessed and reconfirmed that the toxicity of the immunotoxin would be reduced dramatically. The obtained results indicate that the addition or removal of domains cannot depict the activity of immunotoxins and the matter should be assessed structurally in advance.
Collapse
Affiliation(s)
- Zeinab Amiri Tehranizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mojtaba Sankian
- Immunobiochemistry Lab, Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Bibi Sedigheh Fazly Bazzaz
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Jamshidkhan Chamani
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Soghra Mehri
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ali Baratian
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Saberi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Bioinformatics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
250
|
Bezerra PHA, Ferreira IM, Franceschi BT, Bianchini F, Ambrósio L, Cintra ACO, Sampaio SV, de Castro FA, Torqueti MR. BthTX-I from Bothrops jararacussu induces apoptosis in human breast cancer cell lines and decreases cancer stem cell subpopulation. J Venom Anim Toxins Incl Trop Dis 2019; 25:e20190010. [PMID: 31384244 PMCID: PMC6665320 DOI: 10.1590/1678-9199-jvatitd-2019-0010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 06/04/2019] [Indexed: 12/25/2022] Open
Abstract
Background: Breast cancer is the neoplasm with both the highest incidence and mortality
rate among women worldwide. Given the known snake venom cytotoxicity towards
several tumor types, we evaluated the effects of BthTX-I from
Bothrops jararacussu on MCF7, SKBR3, and MDAMB231
breast cancer cell lines. Methods: BthTX-I cytotoxicity was determined via MTT
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazoliumbromide assay. Cell
death was measured by a hypotonic fluorescent solution method,
annexin-V-FITC/propidium iodide staining and by apoptotic/autophagic protein
expression. Cancer stem cells (CSCs) were quantified by flow cytometry using
anti-CD24-FITC and anti-CD44-APC antibodies and propidium iodide. Results: BthTX-I at 102 µg/mL induced cell death in all cell lines. The toxin induced
apoptosis in MCF7, SKBR3, and MDAMB231 in a dose-dependent manner, as
confirmed by the increasing number of hypodiploid nuclei. Expression of
pro-caspase 3, pro-caspase 8 and Beclin-1 proteins were increased, while the
level of the antiapoptotic protein Bcl-2 was diminished in MCF7 cells.
BthTX-I changed the staining pattern of CSCs in MDAMB231 cells by increasing
expression of CD24 receptors, which mediated cell death. Conclusions: BthTX-I induces apoptosis and autophagy in all breast cancer cell lines
tested and also reduces CSCs subpopulation, which makes it a promising
therapeutic alternative for breast cancer.
Collapse
Affiliation(s)
- Patrícia Heloise Alves Bezerra
- Laboratory of Clinical Cytology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Isadora Marques Ferreira
- Laboratory of Clinical Cytology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Beatriz Tinoco Franceschi
- Laboratory of Clinical Cytology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Francine Bianchini
- Laboratory of Clinical Cytology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Luciana Ambrósio
- Laboratory of Hematology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Adélia Cristina O Cintra
- Laboratory of Toxinology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- Laboratory of Toxinology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Fabíola Attié de Castro
- Laboratory of Hematology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Maria Regina Torqueti
- Laboratory of Clinical Cytology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| |
Collapse
|