101
|
Maher MA, Naha PC, Mukherjee SP, Byrne HJ. Numerical simulations of in vitro nanoparticle toxicity – The case of poly(amido amine) dendrimers. Toxicol In Vitro 2014; 28:1449-60. [DOI: 10.1016/j.tiv.2014.07.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/18/2014] [Accepted: 07/28/2014] [Indexed: 01/29/2023]
|
102
|
Blaudszun AR, Moldenhauer G, Schneider M, Philippi A. A photosensitizer delivered by bispecific antibody redirected T lymphocytes enhances cytotoxicity against EpCAM-expressing carcinoma cells upon light irradiation. J Control Release 2014; 197:58-68. [PMID: 25449805 DOI: 10.1016/j.jconrel.2014.10.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 09/05/2014] [Accepted: 10/28/2014] [Indexed: 01/19/2023]
Abstract
Recently conducted clinical trials have provided impressive evidence that chemotherapy resistant metastatic melanoma and several hematological malignancies can be cured using adoptive T cell therapy or T cell-recruiting bispecific antibodies. However, a significant fraction of patients did not benefit from these treatments. Here we have evaluated the feasibility of a novel combination therapy which aims to further enhance the killing potential of bispecific antibody-redirected T lymphocytes by using these cells as targeted delivery system for photosensitizing agents. For a first in vitro proof-of-concept study, ex vivo activated human donor T cells were loaded with a poly(styrene sulfonate) (PSS)-complex of the model photosensitizer 5,10,15,20-tetrakis(3-hydroxyphenyl)porphyrin (mTHPP). In the absence of light and when loading with the water-soluble PSS/mTHPP-complex occurred at a tolerable concentration, viability and cytotoxic function of loaded T lymphocytes were not impaired. When "drug-enhanced" T cells were co-cultivated with EpCAM-expressing human carcinoma cells, mTHPP was transferred to target cells. Notably, in the presence of a bispecific antibody, which cross-links effector and target cells thereby inducing the cytolytic activity of cytotoxic T lymphocytes, significantly more photosensitizer was transferred. Consequently, upon irradiation of co-cultures, redirected drug-loaded T cells were more effective in killing A549 lung and SKOV-3 ovarian carcinoma cells than retargeted unloaded T lymphocytes. Particularly, the additive approach using redirected unloaded T cells in combination with appropriate amounts of separately applied PSS/mTHPP was less efficient as well. Thus, by loading T lymphocytes with a stimulus-sensitive anti-cancer drug, we were able to enhance the cytotoxic capacity of carrier cells. Photosensitizer boosted T cells could open new perspectives for adoptive T cell therapy as well as targeted photodynamic therapy.
Collapse
Affiliation(s)
- André-René Blaudszun
- Environment and Bio Group, Korea Institute of Science and Technology (KIST) Europe Forschungsgesellschaft mbH, Saarland University, Campus E7 1, Saarbrücken D-66123, Germany.
| | - Gerhard Moldenhauer
- Department of Translational Immunology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg D-69120, Germany
| | - Marc Schneider
- Department of Pharmaceutics and Biopharmacy, Philipps-University, Ketzerbach 63, Marburg D-35037, Germany
| | - Anja Philippi
- Environment and Bio Group, Korea Institute of Science and Technology (KIST) Europe Forschungsgesellschaft mbH, Saarland University, Campus E7 1, Saarbrücken D-66123, Germany
| |
Collapse
|
103
|
Bonaterra GA, Wakenhut F, Röthlein D, Wolf M, Bistrian BR, Driscoll D, Kinscherf R. Cytoprotection by omega-3 fatty acids as a therapeutic drug vehicle when combined with nephrotoxic drugs in an intravenous emulsion: Effects on intraglomerular mesangial cells. Toxicol Rep 2014; 1:843-857. [PMID: 28962296 PMCID: PMC5598520 DOI: 10.1016/j.toxrep.2014.10.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 08/18/2014] [Accepted: 10/10/2014] [Indexed: 12/03/2022] Open
Abstract
Distinct concentrations of Ketorolac/Gentamicin are toxic to mesangial (MES 13) cells. Omega-3 fatty acids protect intraglomerular mesangial cells against nephrotoxic drugs. Omega-6 fatty acids do not protect MES 13 cells against nephrotoxic drugs. Cytoprotection by n-3 PUFA may be due to increased intracellular redox state.
During therapeutic interventions, blood concentrations of intravenously applied drugs are higher, and their onset of pharmacological action is faster than with other routes of drug administration. However, acute drug therapy often produces nephrotoxic side effects, as commonly seen after treatment with Ketorolac or Gentamicin leading to questions about their use, especially for patients at risk for acute renal failure. Omega-6(n-6) and omega-3(n-3) polyunsaturated fatty acids (PUFA) affect eicosanoid metabolism, which plays a role in the regulation of inflammation. Eicosanoids derived from n-6 FA have proinflammatory and immunoactive functions, whereas eicosanoids derived from n-3 PUFA have anti-inflammatory and cytoprotective properties. We hypothesized that providing such injectable drugs with nephrotoxic potential in combination with n3-PUFAs from the outset, might afford rapid cytoprotection of renal cells, given the recent evidence that intravenously administered n3-PUFAs are rapidly incorporated into cell membranes. We used intraglomerular mesangial cells (MES13) that are sensitive to treatment with Ketorolac or Gentamicin instead of proximal tubular cells which do not respond to Ketorolac. We found a significant inhibition of Ketorolac (0.25, 0.5, 1 mM) or Gentamicin (2.5, 5 mM) induced cytotoxicity after pretreatment of MES13 cells with 0.01% of 20%w/v LipOmega-3 Emulsion 9/1, containing 90:10 wt/wt mixture of fish oil derived triglycerides to medium chain triglycerides.
Collapse
Affiliation(s)
- Gabriel Alejandro Bonaterra
- Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, Robert-Koch-Str. 8, 35032 Marburg, Germany
| | - Florian Wakenhut
- B. Braun Melsungen AG, Hospital Care Division, Am Schwerzelshof, 34212 Melsungen, Germany
| | - Doris Röthlein
- B. Braun Melsungen AG, Hospital Care Division, Am Schwerzelshof, 34212 Melsungen, Germany
| | - Martin Wolf
- B. Braun Melsungen AG, Hospital Care Division, Am Schwerzelshof, 34212 Melsungen, Germany
| | - Bruce Ryan Bistrian
- Beth Israel Deaconess medical Center, Harvard Medical School, 1 Deaconess Road, Boston, MA 02215, USA
| | - David Driscoll
- Stable Solutions LLC, Easton Industrial Park, 19 Norfolk Avenue, South Easton, MA 02375, USA.,University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Ralf Kinscherf
- Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, Robert-Koch-Str. 8, 35032 Marburg, Germany
| |
Collapse
|
104
|
Reissis Y, García-Gareta E, Korda M, Blunn GW, Hua J. The effect of temperature on the viability of human mesenchymal stem cells. Stem Cell Res Ther 2014; 4:139. [PMID: 24238300 PMCID: PMC4055049 DOI: 10.1186/scrt350] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Revised: 08/20/2013] [Accepted: 11/11/2013] [Indexed: 02/07/2023] Open
Abstract
Introduction Impaction allograft with cement is a common technique used in revision hip surgeries for the last 20 years. However, its clinical results are inconsistent. Recent studies have shown that mesenchymal stem cells (MSCs) seeded onto allograft can enhance bone formation. This in vitro study investigates whether the increase in temperature related to the polymerisation of bone cement will affect the viability of human MSCs. Methods The viability of human MSCs was measured after incubating them at temperatures of 38°C, 48°C and 58°C; durations 45 seconds, 80 seconds and 150 seconds. A control group was kept at 37°C and 5% carbon dioxide for the duration of the investigation (7 days). During the course of the study the human MSCs were analysed for cell metabolic activity using the alamarBlue™ assay, cell viability using both Trypan Blue dye exclusion and calcein staining under fluorescent microscopy, and necrosis and apoptosis using Annexin V and propidium iodide for flow cytometric analysis. A one-way analysis of variance with a priori Dunnett’s test was used to indicate the differences between the treatment groups, when analysed against the control. This identified conditions with a significant difference in cell metabolic activity (alamarBlue™) and cell viability (Trypan Blue). Results Results showed that cell metabolism was not severely affected up to 48°C/150 seconds, while cells in the 58°C group died. Similar results were shown using Trypan Blue and calcein analysis for cell viability. No significant difference in apoptosis and necrosis of the cells was observed when human MSCs treated at 48°C/150 seconds were compared with the control group. Conclusions The study suggests that human MSCs seeded onto allograft can be exposed to temperatures up to 48°C for 150 seconds. Exposure to this temperature for this time period is unlikely to occur during impaction allograft surgery when cement is used. Therefore, in many situations, the addition of human MSCs to cemented impaction grafting may be carried out without detrimental effects to the cells. Furthermore, previous studies have shown that this can enhance new bone formation and repair the defects in revision situations.
Collapse
|
105
|
Nickless A, Jackson E, Marasa J, Nugent P, Mercer RW, Piwnica-Worms D, You Z. Intracellular calcium regulates nonsense-mediated mRNA decay. Nat Med 2014; 20:961-6. [PMID: 25064126 PMCID: PMC4126864 DOI: 10.1038/nm.3620] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 02/23/2014] [Indexed: 12/17/2022]
Abstract
The nonsense-mediated mRNA decay (NMD) pathway selectively eliminates aberrant transcripts containing premature translation termination codons and regulates the levels of a number of physiological mRNAs. NMD modulates the clinical outcome of a variety of human diseases, including cancer and many genetic disorders, and may represent a target for therapeutic intervention. Here, we have developed a new multicolored bioluminescence-based reporter system that can specifically and effectively assay NMD in live human cells. Using this reporter system, we conducted a robust high-throughput small-molecule screen in human cells and, unpredictably, identified a group of cardiac glycosides, including ouabain and digoxin, as potent inhibitors of NMD. Cardiac glycoside-mediated effects on NMD are dependent on binding and inhibiting the sodium-potassium ATPase on the plasma membrane and subsequent elevation of intracellular calcium levels. Induction of calcium release from the endoplasmic reticulum also leads to inhibition of NMD. Thus, this study reveals intracellular calcium as a key regulator of NMD and has implications for exploiting NMD in the treatment of disease.
Collapse
Affiliation(s)
- Andrew Nickless
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Erin Jackson
- BRIGHT Institute, Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Jayne Marasa
- BRIGHT Institute, Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Patrick Nugent
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Robert W. Mercer
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - David Piwnica-Worms
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
- BRIGHT Institute, Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110
- Department of Cancer Systems Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Zhongsheng You
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
106
|
Li C, Dai Y, Zhang SX, Duan YH, Liu ML, Chen LY, Yao XS. Quinoid glycosides from Forsythia suspensa. PHYTOCHEMISTRY 2014; 104:105-13. [PMID: 24833035 DOI: 10.1016/j.phytochem.2014.04.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 02/26/2014] [Accepted: 04/11/2014] [Indexed: 05/21/2023]
Abstract
Phytochemical investigation on Forsythia suspensa (Thunb.) Vahl afforded 10 compounds, including quinoid glycosides, lignan glycosides, phenylethanoid glycoside and allylbenzene glycoside together with 13 known ones. Their structures were established based on extensive spectroscopic data analyses, including IR, UV, HRESIMS, 1D NMR and 2D NMR. Absolute configurations were determined by ECD calculation method and chemical degradation. In addition, all compounds were evaluated for their antiviral activity against influenza A (H1N1) virus and several were further evaluated against respiratory syncytial virus (RSV) in vitro. Among them, two previously known compounds showed significant activities against RSV with EC50 values of 3.43 and 6.72 μM.
Collapse
Affiliation(s)
- Chang Li
- College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yi Dai
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People's Republic of China
| | - Shu-Xiang Zhang
- Beijing American Oriental Pharmaceutical Institute, Beijing 100176, People's Republic of China
| | - Ying-Hui Duan
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People's Republic of China
| | - Ming-Li Liu
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People's Republic of China
| | - Liu-Yuan Chen
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People's Republic of China
| | - Xin-Sheng Yao
- College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, People's Republic of China.
| |
Collapse
|
107
|
Yang X, Yang J, Li L, Sun L, Yi X, Han X, Si W, Yan R, Chen Z, Xie G, Li W, Shang Y, Liang J. PAAT, a novel ATPase and trans-regulator of mitochondrial ABC transporters, is critically involved in the maintenance of mitochondrial homeostasis. FASEB J 2014; 28:4821-34. [PMID: 25063848 DOI: 10.1096/fj.14-254045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
ATP-binding cassette (ABC) transporters are implicated in a diverse range of physiological and pathophysiological processes, such as cholesterol and lipid transportation and multidrug resistance. Despite the considerable efforts made in understanding of the cellular function of ABC proteins, the regulation mechanism of this type of protein is still poorly defined. Here we report the identification and functional characterization of a novel ATPase protein, protein associated with ABC transporters (PAAT), in humans. PAAT contains a nucleotide-binding domain (NBD)-like domain and a signal for intramitochondrial sorting. We showed that PAAT is localized in both the cytoplasm and the mitochondria and has an intrinsic ATPase activity. PAAT physically interacts with the 3 known mitochondrial inner membrane ABC proteins, ABCB7, ABCB8, and ABCB10, but not ABCB1, ABCB6, or ABCG2, and functionally regulates the transport of ferric nutrients and heme biosynthesis. Significantly, PAAT deficiency promotes cell death, reduces mitochondrial potential, and sensitizes mitochondria to oxidative stress-induced DNA damages. Our experiments revealed that PAAT is a novel ATPase and a trans-regulator of mitochondrial ABC transporters that plays an important role in the maintenance of mitochondrial homeostasis and cell survival.
Collapse
Affiliation(s)
- Xiaohan Yang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| | - Jianguo Yang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| | - Lei Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| | - Luyang Sun
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| | - Xia Yi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| | - Xiao Han
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| | - Wenzhe Si
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and Tianjin Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China
| | - Ruorong Yan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| | - Zhe Chen
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| | - Guojia Xie
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| | - Wanjin Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| | - Yongfeng Shang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and Tianjin Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, China
| | - Jing Liang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China; and
| |
Collapse
|
108
|
Munshi S, Twining RC, Dahl R. Alamar blue reagent interacts with cell-culture media giving different fluorescence over time: potential for false positives. J Pharmacol Toxicol Methods 2014; 70:195-8. [PMID: 24933394 DOI: 10.1016/j.vascn.2014.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/28/2014] [Accepted: 06/04/2014] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The cell viability assay by alamar blue is based on the principle of reduction of the non-fluorescent reagent (resazurin) to a fluorescent compound (resarufin) by the intracellular reducing environment of living cells over time. In the present study, we have for the first time shown that even in the absence of cells, there occurs significant interaction between alamar blue and cell-culture media causing an increase in fluorescence. METHODS We have used Opti-MEM, DMEM and 1:1 DMEM:Opti-MEM as three different media and determined the changes in their relative fluorescence units (RFUs) over time after the addition of 10% (v/v) alamar blue using two-way repeated measures analysis of variance (RM-ANOVA) followed by Tukey's post-hoc test. RESULTS Our results show that upon the addition of alamar blue, there occurs a significant increase in RFUs in all the three media over time along with a significantly higher RFU for the Opti-MEM overall (p<0.05). We also show that the time-dependent change in RFU of 1:1 DMEM:Opti-MEM was more gradual compared to that of the other two media. DISCUSSION These findings indicate that the reagent can itself interact with the media causing significantly different fluorescence over time in a manner independent from the effect of intracellular reducing environment of living cells on alamar blue. In addition our results indicate that fluorescence varies as a function of incubation time with the reagent. These findings signify the need for routine subtraction of the background fluorescence of media-only with alamar blue reagent during measurement of cell viability by this method in order to determine an accurate measurement of cell viability.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| | - Robert C Twining
- Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| | - Russell Dahl
- Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| |
Collapse
|
109
|
In vitro antiplasmodial activities and synergistic combinations of differential solvent extracts of the polyherbal product, Nefang. BIOMED RESEARCH INTERNATIONAL 2014; 2014:835013. [PMID: 24877138 PMCID: PMC4022163 DOI: 10.1155/2014/835013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/04/2014] [Indexed: 11/17/2022]
Abstract
Nefang, a polyherbal product composed of Mangifera indica (bark and leaf), Psidium guajava, Carica papaya, Cymbopogon citratus, Citrus sinensis, and Ocimum gratissimum (leaves), is a potential therapy against P. falciparum malaria. In vitro antiplasmodial activities of its constituent solvent extracts were analyzed on CQ-sensitive (3D7) and multidrug resistant (Dd2) P. falciparum strains. The interactions involving the differential solvent extracts were further analyzed using a variable potency ratio drug combination approach. Effective concentration 50 (EC50) values were determined by nonlinear regression curve-fitting of the dose-response data and used in calculating the fractional inhibitory concentration 50 (FIC50) and combination indices (CI) for each pair. The derived EC50 values (3D7/Dd2, μg/mL) are Nefang-96.96/55.08, MiB-65.33/34.58, MiL-82.56/40.04, Pg-47.02/25.79, Cp-1188/317.5, Cc-723.3/141, Cs-184.4/105.1, and Og-778.5/118.9. Synergism was obtained with MiB/Pg (CI = 0.351), MiL/Pg (0.358), MiB/Cs (0.366), MiL/Cs (0.482), Pg/Cs (0.483), and Cs/Og (0.414) when analyzed at equipotency ratios. Cytotoxicity testing of Nefang and the solvent extracts on two human cell lines (Hep G2 and U2OS) revealed no significant toxicity relative to their antiplasmodial activities (SI > 20). Taken together, our data confirm the antimalarial activities of Nefang and its
constituent plant extracts and identified extract pairs with promising synergistic interactions for exploitation towards a rational phytotherapeutic and evidence-based antimalarial drug discovery.
Collapse
|
110
|
Wucherpfennig T, Schulz A, Pimentel JA, Corkidi G, Sieblitz D, Pump M, Gorr G, Schütte K, Wittmann C, Krull R. Viability characterization of Taxus chinensis plant cell suspension cultures by rapid colorimetric- and image analysis-based techniques. Bioprocess Biosyst Eng 2014; 37:1799-1808. [DOI: 10.1007/s00449-014-1153-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/07/2014] [Indexed: 11/29/2022]
|
111
|
Bohanon FJ, Wang X, Ding C, Ding Y, Radhakrishnan GL, Rastellini C, Zhou J, Radhakrishnan RS. Oridonin inhibits hepatic stellate cell proliferation and fibrogenesis. J Surg Res 2014; 190:55-63. [PMID: 24742622 DOI: 10.1016/j.jss.2014.03.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/07/2014] [Accepted: 03/12/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Liver fibrosis is a common response to liver injury and, in severe cases, leads to cirrhosis. The hepatic stellate cells (HSCs) become activated after liver injury and play a significant role in fibrogenesis. The activated HSC is characterized by increased proliferation, overexpression of α smooth muscle actin, and excessive production of extracellular matrix (ECM) proteins. Oridonin, a naturally occurring diterpenoid, has been shown to induce apoptosis in liver and gastric cancer cells. However, its effects on the HSC are unknown. METHODS We tested the effects of oridonin on the activated human and rat HSC lines LX-2 and HSC-T6, and the human hepatocyte cell line C3A. Transforming growth factor β1 (TGF-β1) was used to stimulate LX-2 cells. RESULTS Oridonin significantly inhibited LX-2 and HSC-T6 proliferation. In contrast, oridonin had no antiproliferative effect on C3A cells at our tested range. Oridonin induced apoptosis and S-phase arrest in LX-2 cells. These findings were associated with an increase in p53, p21, p16, and cleaved Poly (ADP-ribose) Polymerase (PARP), and with a decrease in Cyclin-dependent kinase 4 (Cdk4). Oridonin markedly decreased expression of α smooth muscle actin and ECM protein type I collagen and fibronectin, blocked TGF-β1-induced Smad2/3 phosphorylation and type I collagen expression. CONCLUSIONS Oridonin induces apoptosis and cell cycle arrest involving the p53-p21 pathway in HSC and appears to be nontoxic to hepatocytes. In addition, oridonin suppressed endogenous and TGF-β1-induced ECM proteins. Thus, oridonin may act as a novel agent to prevent hepatic fibrosis.
Collapse
Affiliation(s)
- Fredrick J Bohanon
- Department of Surgery, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Xiaofu Wang
- Department of Surgery, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Chunyong Ding
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Ye Ding
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Geetha L Radhakrishnan
- Department of Pediatrics, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Cristiana Rastellini
- Department of Surgery, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Ravi S Radhakrishnan
- Department of Surgery, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.,Department of Pediatrics, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| |
Collapse
|
112
|
High-throughput screening identifies idarubicin as a preferential inhibitor of smooth muscle versus endothelial cell proliferation. PLoS One 2014; 9:e89349. [PMID: 24586708 PMCID: PMC3933427 DOI: 10.1371/journal.pone.0089349] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 01/20/2014] [Indexed: 01/21/2023] Open
Abstract
Intimal hyperplasia is the cause of the recurrent occlusive vascular disease (restenosis). Drugs currently used to treat restenosis effectively inhibit smooth muscle cell (SMC) proliferation, but also inhibit the growth of the protective luminal endothelial cell (EC) lining, leading to thrombosis. To identify compounds that selectively inhibit SMC versus EC proliferation, we have developed a high-throughput screening (HTS) format using human cells and have employed this to screen a multiple compound collection (NIH Clinical Collection). We developed an automated, accurate proliferation assay in 96-well plates using human aortic SMCs and ECs. Using this HTS format we screened a 447-drug NIH Clinical Library. We identified 11 compounds that inhibited SMC proliferation greater than 50%, among which idarubicin exhibited a unique feature of preferentially inhibiting SMC versus EC proliferation. Concentration-response analysis revealed this differential effect most evident over an ∼10 nM-5 µM window. In vivo testing of idarubicin in a rat carotid injury model at 14 days revealed an 80% reduction of intimal hyperplasia and a 45% increase of lumen size with no significant effect on re-endothelialization. Taken together, we have established a HTS assay of human vascular cell proliferation, and identified idarubicin as a selective inhibitor of SMC versus EC proliferation both in vitro and in vivo. Screening of larger and more diverse compound libraries may lead to the discovery of next-generation therapeutics that can inhibit intima hyperplasia without impairing re-endothelialization.
Collapse
|
113
|
Leusch FDL, Khan SJ, Laingam S, Prochazka E, Froscio S, Trinh T, Chapman HF, Humpage A. Assessment of the application of bioanalytical tools as surrogate measure of chemical contaminants in recycled water. WATER RESEARCH 2014; 49:300-15. [PMID: 24355290 DOI: 10.1016/j.watres.2013.11.030] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/15/2013] [Accepted: 11/18/2013] [Indexed: 05/08/2023]
Abstract
The growing use of recycled water in large urban centres requires comprehensive public health risk assessment and management, an important aspect of which is the assessment and management of residual trace chemical substances. Bioanalytical methods such as in vitro bioassays may be ideal screening tools that can detect a wide range of contaminants based on their biological effect. In this study, we applied thirteen in vitro assays selected explicitly for their ability to detect molecular and cellular effects relevant to potential chemical exposure via drinking water as a means of screening for chemical contaminants from recycled water at 9 Australian water reclamation plants, in parallel to more targeted direct chemical analysis of 39 priority compounds. The selected assays provided measures of primary non-specific (cytotoxicity to various cell types), specific (inhibition of acetylcholinesterase and endocrine receptor-mediated effects) and reactive toxicity (mutagenicity and genotoxicity), as well as markers of adaptive stress response (modulation of cytokine production) and xenobiotic metabolism (liver enzyme induction). Chemical and bioassay analyses were in agreement and complementary to each other: the results show that source water (treated wastewater) contained high levels of biologically active compounds, with positive results in almost all bioassays. The quality of the product water (reclaimed water) was only marginally better after ultrafiltration or dissolved air floatation/filtration, but greatly improved after reverse osmosis often reducing biological activity to below detection limit. The bioassays were able to detect activity at concentrations below current chemical method detection limits and provided a sum measure of all biologically active compounds for that bioassay, thus providing an additional degree of confidence in water quality.
Collapse
Affiliation(s)
- Frederic D L Leusch
- Smart Water Research Centre, Griffith University Gold Coast Campus, Southport, Qld 4222, Australia.
| | - Stuart J Khan
- Water Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | | | - Erik Prochazka
- Smart Water Research Centre, Griffith University Gold Coast Campus, Southport, Qld 4222, Australia
| | - Suzanne Froscio
- Australian Water Quality Centre, SA Water, Adelaide, SA 5001, Australia
| | - Trang Trinh
- Water Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Heather F Chapman
- Smart Water Research Centre, Griffith University Gold Coast Campus, Southport, Qld 4222, Australia
| | - Andrew Humpage
- Australian Water Quality Centre, SA Water, Adelaide, SA 5001, Australia
| |
Collapse
|
114
|
Dessì M, Alvarez-Perez MA, De Santis R, Ginebra MP, Planell JA, Ambrosio L. Bioactivation of calcium deficient hydroxyapatite with foamed gelatin gel. A new injectable self-setting bone analogue. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2014; 25:283-295. [PMID: 24136082 DOI: 10.1007/s10856-013-5071-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 10/09/2013] [Indexed: 06/02/2023]
Abstract
An alternative approach to bone repair for less invasive surgical techniques, involves the development of biomaterials directly injectable into the injury sites and able to replicate a spatially organized platform with features of bone tissue. Here, the preparation and characterization of an innovative injectable bone analogue made of calcium deficient hydroxyapatite and foamed gelatin is presented. The biopolymer features and the cement self-setting reaction were investigated by rheological analysis. The porous architecture, the evolution of surface morphology and the grains dimension were analyzed with electron microscopy (SEM/ESEM/TEM). The physico-chemical properties were characterized by X-ray diffraction and FTIR analysis. Moreover, an injection test was carried out to prove the positive effect of gelatin on the flow ensuing that cement is fully injectable. The cement mechanical properties are adequate to function as temporary substrate for bone tissue regeneration. Furthermore, MG63 cells and bone marrow-derived human mesenchymal stem cells (hMSCs) were able to migrate and proliferate inside the pores, and hMSCs differentiated to the osteoblastic phenotype. The results are paving the way for an injectable bone substitute with properties that mimic natural bone tissue allowing the successful use as bone filler for craniofacial and orthopedic reconstructions in regenerative medicine.
Collapse
Affiliation(s)
- M Dessì
- Institute of Composite and Biomedical Materials, National Research Council of Italy, P.le Tecchio 80, 80125, Naples, Italy,
| | | | | | | | | | | |
Collapse
|
115
|
Wolpaw AJ, Stockwell BR. Multidimensional profiling in the investigation of small-molecule-induced cell death. Methods Enzymol 2014; 545:265-302. [PMID: 25065894 DOI: 10.1016/b978-0-12-801430-1.00011-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Numerous morphological variations of cell death have been described. These processes depend on a complex and overlapping cellular signaling network, making molecular definition of the pathways challenging. This review describes one solution to this problem for small-molecule-induced death, the creation of high-dimensionality profiles for compounds that can be used to define and compare pathways. Such profiles have been assembled from gene expression measurements, protein quantification, chemical-genetic interactions, chemical combination interactions, cancer cell line sensitivity profiling, quantitative imaging, and modulatory profiling. We discuss the advantages and limitations of these techniques in the study of cell death.
Collapse
Affiliation(s)
- Adam J Wolpaw
- Residency Program in Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, USA; Department of Chemistry, Columbia University, New York, USA; Howard Hughes Medical Institute, Columbia University, New York, USA.
| |
Collapse
|
116
|
García-Gareta E, Ravindran N, Sharma V, Samizadeh S, Dye JF. A novel multiparameter in vitro model of three-dimensional cell ingress into scaffolds for dermal reconstruction to predict in vivo outcome. Biores Open Access 2013; 2:412-20. [PMID: 24380051 PMCID: PMC3869440 DOI: 10.1089/biores.2013.0043] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The clinical demand for effective dermal substitutes continues as current commercially available products present limitations. However, there are no definitive in vitro methods to predict in vivo outcomes such as integration, cellularization and contraction, which may help the development of new dermal scaffolds. This study aimed to develop a multiparameter in vitro model of three-dimensional (3D) cell ingress into dermal scaffolds to predict in vivo outcomes of new dermal scaffolds under development. A new dermal scaffold, Smart Matrix, was compared to the scar-forming contractile collagen gel model and to the clinically well-established Integra® and Matriderm®. Parameters studied were cell viability and proliferation, apoptosis, matrix contraction, cell morphology, α-smooth muscle actin, and growth factor expression. Combinatorial evaluation of the results in a scoring matrix showed that Smart Matrix could offer an advantage over existing products. This method would be useful as an international golden scoring matrix to develop new dermal scaffolds that effectively improve the existing products, thus enabling better treatments for burns or chronic wounds.
Collapse
Affiliation(s)
- Elena García-Gareta
- RAFT Institute of Plastic Surgery , Mount Vernon Hospital, Northwood, United Kingdom
| | - Nivedita Ravindran
- RAFT Institute of Plastic Surgery , Mount Vernon Hospital, Northwood, United Kingdom
| | - Vaibhav Sharma
- RAFT Institute of Plastic Surgery , Mount Vernon Hospital, Northwood, United Kingdom
| | - Sorousheh Samizadeh
- RAFT Institute of Plastic Surgery , Mount Vernon Hospital, Northwood, United Kingdom
| | - Julian F Dye
- RAFT Institute of Plastic Surgery , Mount Vernon Hospital, Northwood, United Kingdom
| |
Collapse
|
117
|
Sulphoxythiocarbamates modify cysteine residues in HSP90 causing degradation of client proteins and inhibition of cancer cell proliferation. Br J Cancer 2013; 110:71-82. [PMID: 24322890 PMCID: PMC3887302 DOI: 10.1038/bjc.2013.710] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/11/2013] [Accepted: 10/17/2013] [Indexed: 12/15/2022] Open
Abstract
Background: Heat shock protein 90 (HSP90) has a key role in the maintenance of the cellular proteostasis. However, HSP90 is also involved in stabilisation of oncogenic client proteins and facilitates oncogene addiction and cancer cell survival. The development of HSP90 inhibitors for cancer treatment is an area of growing interest as such agents can affect multiple pathways that are linked to all hallmarks of cancer. This study aimed to test the hypothesis that targeting cysteine residues of HSP90 will lead to degradation of client proteins and inhibition of cancer cell proliferation. Methods: Combining chemical synthesis, biological evaluation, and structure–activity relationship analysis, we identified a new class of HSP90 inhibitors. Click chemistry and protease-mass spectrometry established the sites of modification of the chaperone. Results: The mildly electrophilic sulphoxythiocarbamate alkyne (STCA) selectively targets cysteine residues of HSP90, forming stable thiocarbamate adducts. Without interfering with the ATP-binding ability of the chaperone, STCA destabilises the client proteins RAF1, HER2, CDK1, CHK1, and mutant p53, and decreases proliferation of breast cancer cells. Addition of a phenyl or a tert-butyl group in tandem with the benzyl substituent at nitrogen increased the potency. A new compound, S-4, was identified as the most robust HSP90 inhibitor within a series of 19 derivatives. Conclusion: By virtue of their cysteine reactivity, sulphoxythiocarbamates target HSP90, causing destabilisation of its client oncoproteins and inhibiting cell proliferation.
Collapse
|
118
|
Hutin M, Burakowska-Meise E, Appel WPJ, Dankers PYW, Meijer EW. From Molecular Structure to Macromolecular Organization: Keys to Design Supramolecular Biomaterials. Macromolecules 2013. [DOI: 10.1021/ma401552e] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Marie Hutin
- Institute for Complex Molecular Systems & Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Ewelina Burakowska-Meise
- Institute for Complex Molecular Systems & Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Wilco P. J. Appel
- Institute for Complex Molecular Systems & Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Patricia Y. W. Dankers
- Institute for Complex Molecular Systems & Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - E. W. Meijer
- Institute for Complex Molecular Systems & Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
119
|
Pereira-Fernandes A, Demaegdt H, Vandermeiren K, Hectors TLM, Jorens PG, Blust R, Vanparys C. Evaluation of a screening system for obesogenic compounds: screening of endocrine disrupting compounds and evaluation of the PPAR dependency of the effect. PLoS One 2013; 8:e77481. [PMID: 24155963 PMCID: PMC3796469 DOI: 10.1371/journal.pone.0077481] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/02/2013] [Indexed: 12/21/2022] Open
Abstract
Recently the environmental obesogen hypothesis has been formulated, proposing a role for endocrine disrupting compounds (EDCs) in the development of obesity. To evaluate this hypothesis, a screening system for obesogenic compounds is urgently needed. In this study, we suggest a standardised protocol for obesogen screening based on the 3T3-L1 cell line, a well-characterised adipogenesis model, and direct fluorescent measurement using Nile red lipid staining technique. In a first phase, we characterised the assay using the acknowledged obesogens rosiglitazone and tributyltin. Based on the obtained dose-response curves for these model compounds, a lipid accumulation threshold value was calculated to ensure the biological relevance and reliability of statistically significant effects. This threshold based method was combined with the well described strictly standardized mean difference (SSMD) method for classification of non-, weak- or strong obesogenic compounds. In the next step, a range of EDCs, used in personal and household care products (parabens, musks, phthalates and alkylphenol compounds), were tested to further evaluate the obesogenicity screening assay for its discriminative power and sensitivity. Additionally, the peroxisome proliferator activated receptor γ (PPARγ) dependency of the positive compounds was evaluated using PPARγ activation and antagonist experiments. Our results showed the adipogenic potential of all tested parabens, several musks and phthalate compounds and bisphenol A (BPA). PPARγ activation was associated with adipogenesis for parabens, phthalates and BPA, however not required for obesogenic effects induced by Tonalide, indicating the role of other obesogenic mechanisms for this compound.
Collapse
Affiliation(s)
- Anna Pereira-Fernandes
- Systemic Physiological and Ecotoxicological Research (SPHERE), Department of Biology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | |
Collapse
|
120
|
In vitro toxicity of Stearoyl-poly(glycerol adipate) nanoparticles. J Appl Biomater Funct Mater 2013; 10:163-9. [PMID: 23242880 DOI: 10.5301/jabfm.2012.10294] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2012] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Poly(glycerol adipate) (PGA)-based nanoparticles are promising carriers for drug delivery with a wide range of available structures. The biodegradable polymer with pendant free hydroxyl groups can be diversely functionalized. In this study, the toxicity of different Stearoyl-PGA nanoparticles with respect to erythrocytes and HepG2 cells was assessed. These cells are crucial test systems for intravenously injected biomedical particles. METHODS For this work, a series of PGA polyesters with 0, 20, 50 and 65 mol% of converted hydroxyl groups was synthesized with stearic acid (PGABB, S20, S50, S65). Nanoparticles were prepared with these polymers by an optimized nanoprecipitation method. Physicochemical characterization was performed by photon correlation spectroscopy and zeta potential measurement. Cell compatibility was studied by a hemolysis assay with separated red blood cells as well as a QBlue viability test and a modified LDH cytotoxicity assay with HepG2 cells. RESULTS AND CONCLUSIONS Different self-stabilizing nanoparticles with narrow size distributions in the range of 100-140 nm were prepared. All tested nanoparticle samples were nontoxic for HepG2 cells. In fact, increased metabolic activity and proliferation was observed after 24 h incubation with the Stearoyl-PGA particles. Apart from PGAS20, all samples did not show any hemolytic effect. Hemolysis of PGAS20 particles could be considerably decreased by adding Poloxamer 188 during the preparation process.
Collapse
|
121
|
Heredero-Bermejo I, Copa-Patiño JL, Soliveri J, Gómez R, de la Mata FJ, Pérez-Serrano J. In vitro comparative assessment of different viability assays in Acanthamoeba castellanii and Acanthamoeba polyphaga trophozoites. Parasitol Res 2013; 112:4087-95. [PMID: 24026387 DOI: 10.1007/s00436-013-3599-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 08/25/2013] [Indexed: 01/30/2023]
Abstract
The species of the genus Acanthamoeba are opportunistic protozoan parasites that cause different diseases in humans, such as amoebic keratitis and granulomatous encephalitis. The rise in the rate of Acanthamoeba keratitis, mainly due to the increase in contact lens wearers, turns the development of viability assays using a multi-well plate reader as a tool for screening new antiamoebic agents in vitro into an important goal. In our study, the viability assays PrestoBlue®, resazurin sodium salt, 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide (MTT) and CellTiter96® were tested for their suitability as time-saving alternatives to the classical manual or direct-counting method, assessing the effect of the antiamoebic agent chlorhexidine digluconate and temperature on Acanthamoeba castellanii (ATCC® 30234™) and Acanthamoeba polyphaga 2961. Although resazurin and MTT have already been previously used in amoeba viability assays to test the activities of antiamoebic agents in vitro, it is the first time that PrestoBlue® and CellTiter96® are used for this purpose. Results indicated that the viability assays were strain-dependent leading in some cases to an overestimation of the real situation of viable cells. This implies that each viability assay ought to be set up for each amoeba strain studied.
Collapse
Affiliation(s)
- I Heredero-Bermejo
- Department of Biomedicine and Biotechnology, School of Pharmacy, University of Alcalá, Ctra. Madrid-Barcelona (Autovía A2) Km. 33 600, 28871, Alcalá de Henares, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
122
|
Zachari MA, Chondrou PS, Pouliliou SE, Mitrakas AG, Abatzoglou I, Zois CE, Koukourakis MI. Evaluation of the alamarblue assay for adherent cell irradiation experiments. Dose Response 2013; 12:246-58. [PMID: 24910583 DOI: 10.2203/dose-response.13-024.koukourakis] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The AlamarBlue assay is based on fluorometric detection of metabolic mitochondrial activity of cells. In this study, we determined the methodology for application of the assay to radiation response experiments in 96-well plates. AlamarBlue was added and its reduction measured 7 hours later. Selection of the initial number of plated cells was important so that the number of proliferating cells remains lower than the critical number that produced full AlamarBlue reduction (plateau phase) at the time points of measurements. Culture medium was replaced twice a week to avoid suppression of viability due to nutrient competition and metabolic waste accumulation. There was no need to replace culture medium before adding AlamarBlue. Cell proliferation continued after irradiation and the suppression effect on cell viability was most evident on day 8. At this time point, by comparing measurements from irradiated vs. non-irradiated cells, for various dose levels, a viability dose response curve was plotted. Immediately after the 8(th) day (nadir), cells started to re-grow at a rate inversely related to the radiation dose. By comparing measurements at the time point of nadir vs. a convenient subsequent time point, re-growth dose response abilities were plotted, simulating clonogenic assays.
Collapse
Affiliation(s)
- Maria A Zachari
- Department of Radiotherapy / Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace, Alexandroupolis, Greece
| | - Panagiota S Chondrou
- Department of Radiotherapy / Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace, Alexandroupolis, Greece
| | - Stamatia E Pouliliou
- Department of Radiotherapy / Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace, Alexandroupolis, Greece
| | - Achilleas G Mitrakas
- Department of Radiotherapy / Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioannis Abatzoglou
- Department of Radiotherapy / Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace, Alexandroupolis, Greece
| | - Christos E Zois
- Department of Radiotherapy / Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace, Alexandroupolis, Greece
| | - Michael I Koukourakis
- Department of Radiotherapy / Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
123
|
Yang X, Li L, Liang J, Shi L, Yang J, Yi X, Zhang D, Han X, Yu N, Shang Y. Histone acetyltransferase 1 promotes homologous recombination in DNA repair by facilitating histone turnover. J Biol Chem 2013; 288:18271-82. [PMID: 23653357 PMCID: PMC3689969 DOI: 10.1074/jbc.m113.473199] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 05/07/2013] [Indexed: 01/05/2023] Open
Abstract
Faithful repair of DNA double-strand breaks is vital to the maintenance of genome integrity and proper cell functions. Histone modifications, such as reversible acetylation, phosphorylation, methylation, and ubiquitination, which collectively contribute to the establishment of distinct chromatin states, play important roles in the recruitment of repair factors to the sites of double-strand breaks. Here we report that histone acetyltransferase 1 (HAT1), a classical B type histone acetyltransferase responsible for acetylating the N-terminal tail of newly synthesized histone H4 in the cytoplasm, is a key regulator of DNA repair by homologous recombination in the nucleus. We found that HAT1 is required for the incorporation of H4K5/K12-acetylated H3.3 at sites of double-strand breaks through its HIRA-dependent histone turnover activity. Incorporated histones with specific chemical modifications facilitate subsequent recruitment of RAD51, a key repair factor in mammalian cells, to promote efficient homologous recombination. Significantly, depletion of HAT1 sensitized cells to DNA damage compromised the global chromatin structure, inhibited cell proliferation, and induced cell apoptosis. Our experiments uncovered a role for HAT1 in DNA repair in higher eukaryotic organisms and provide a mechanistic insight into the regulation of histone dynamics by HAT1.
Collapse
Affiliation(s)
- Xiaohan Yang
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China and
| | - Lei Li
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China and
| | - Jing Liang
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China and
| | - Lei Shi
- Tianjin Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin 300070, China
| | - Jianguo Yang
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China and
| | - Xia Yi
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China and
| | - Di Zhang
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China and
| | - Xiao Han
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China and
| | - Na Yu
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China and
| | - Yongfeng Shang
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China and
- Tianjin Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
124
|
D'Antò V, Raucci MG, Guarino V, Martina S, Valletta R, Ambrosio L. Behaviour of human mesenchymal stem cells on chemically synthesized HA-PCL scaffolds for hard tissue regeneration. J Tissue Eng Regen Med 2013; 10:E147-54. [PMID: 23723157 DOI: 10.1002/term.1768] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 03/13/2013] [Accepted: 04/13/2013] [Indexed: 01/09/2023]
Abstract
Our goal was to characterize the response of human mesenchymal stem cells (hMSCs) to a novel composite scaffold for bone tissue engineering. The hydroxyapatite-polycaprolactone (HA-PCL) composite scaffolds were prepared by a sol-gel method at room temperature and the scaffold morphology was investigated by scanning electron microscopy (SEM)/energy-dispersive spectroscopy (EDS) to validate the synthesis process. The response of two different lines of hMSCs, bone-marrow-derived human mesenchymal stem cells (BMSCs) and dental pulp stem cells (DPSCs) in terms of cell proliferation and differentiation into the osteoblastic phenotype, was evaluated using Alamar blue assay, SEM, histology and alkaline phosphatase activity. Our results indicate that tissue engineering by means of composite HA-PCL scaffolds may represent a new therapeutic strategy to repair craniofacial bone defects.
Collapse
Affiliation(s)
- Vincenzo D'Antò
- Institute of Composite and Biomedical Materials (IMCB), National Research Council of Italy (CNR), Naples, Italy.,Department of Neurosciences, Reproductive Sciences and Oral Sciences, University of Naples 'Federico II', Italy.,Department of Pediatric Surgery and Transplantation, Bambino Gesù Children's Hospital, Rome, Italy
| | - Maria Grazia Raucci
- Institute of Composite and Biomedical Materials (IMCB), National Research Council of Italy (CNR), Naples, Italy
| | - Vincenzo Guarino
- Institute of Composite and Biomedical Materials (IMCB), National Research Council of Italy (CNR), Naples, Italy
| | - Stefano Martina
- Department of Neurosciences, Reproductive Sciences and Oral Sciences, University of Naples 'Federico II', Italy
| | - Rosa Valletta
- Department of Neurosciences, Reproductive Sciences and Oral Sciences, University of Naples 'Federico II', Italy
| | - Luigi Ambrosio
- Institute of Composite and Biomedical Materials (IMCB), National Research Council of Italy (CNR), Naples, Italy
| |
Collapse
|
125
|
Ghosh M, Chakraborty A, Mukherjee A. Cytotoxic, genotoxic and the hemolytic effect of titanium dioxide (TiO2) nanoparticles on human erythrocyte and lymphocyte cellsin vitro. J Appl Toxicol 2013; 33:1097-110. [DOI: 10.1002/jat.2863] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Revised: 12/26/2012] [Accepted: 01/13/2013] [Indexed: 11/12/2022]
Affiliation(s)
- Manosij Ghosh
- Cell Biology and Genetic Toxicology Laboratory, Centre of Advanced Study, Department of Botany; University of Calcutta; Kolkata; India
| | | | - Anita Mukherjee
- Cell Biology and Genetic Toxicology Laboratory, Centre of Advanced Study, Department of Botany; University of Calcutta; Kolkata; India
| |
Collapse
|
126
|
Ganly S, Hynes SO, Sharif F, Aied A, Barron V, McCullagh K, McMahon J, McHugh P, Crowley J, Wang W, O'Brien T, Greiser U. Liposomal surface coatings of metal stents for efficient non-viral gene delivery to the injured vasculature. J Control Release 2013; 167:109-19. [DOI: 10.1016/j.jconrel.2013.01.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 01/14/2013] [Accepted: 01/31/2013] [Indexed: 11/16/2022]
|
127
|
Zhou X, Holsbeeks I, Impens S, Sonnaert M, Bloemen V, Luyten F, Schrooten J. Noninvasive real-time monitoring by alamarBlue(®) during in vitro culture of three-dimensional tissue-engineered bone constructs. Tissue Eng Part C Methods 2013; 19:720-9. [PMID: 23327780 DOI: 10.1089/ten.tec.2012.0601] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Bone tissue engineering (TE) aims to develop reproducible and predictive three-dimensional (3D) TE constructs, defined as cell-seeded scaffolds produced by a controlled in vitro process, to heal or replace damaged and nonfunctional bone. To control and assure the quality of the bone TE constructs, a prerequisite for regulatory authorization, there is a need to develop noninvasive analysis techniques to evaluate TE constructs and to monitor their behavior in real time during in vitro culturing. Most analysis techniques, however, are limited to destructive end-point analyses. This study investigates the use of the nontoxic alamarBlue(®) (AB) reagent, which is an indicator for metabolic cell activity, for monitoring the cellularity of 3D TE constructs in vitro as part of a bioreactor culturing processes. Within the field of TE, bioreactors have a huge potential in the translation of TE concepts to the clinic. Hence, the use of the AB reagent was evaluated not only in static cultures, but also in dynamic cultures in a perfusion bioreactor setup. Hereto, the AB assay was successfully integrated in the bioreactor-driven TE construct culture process in a noninvasive way. The obtained results indicate a linear correlation between the overall metabolic activity and the total DNA content of a scaffold upon seeding as well as during the initial stages of cell proliferation. This makes the AB reagent a powerful tool to follow-up bone TE constructs in real-time during static as well as dynamic 3D cultures. Hence, the AB reagent can be successfully used to monitor and predict cell confluence in a growing 3D TE construct.
Collapse
Affiliation(s)
- Xiaohua Zhou
- Biomedical Engineering Research Team, Groep T, Leuven Engineering College, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
128
|
Bénardais K, Kotsiari A, Skuljec J, Koutsoudaki PN, Gudi V, Singh V, Vulinović F, Skripuletz T, Stangel M. Cuprizone [bis(cyclohexylidenehydrazide)] is selectively toxic for mature oligodendrocytes. Neurotox Res 2013; 24:244-50. [PMID: 23392957 DOI: 10.1007/s12640-013-9380-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Revised: 01/18/2013] [Accepted: 01/23/2013] [Indexed: 11/26/2022]
Abstract
Cuprizone [bis(cyclohexylidenehydrazide)]-induced toxic demyelination is an experimental animal model commonly used to study de- and remyelination in the central nervous system. In this model, mice are fed with the copper chelator cuprizone which leads to oligodendrocyte death with subsequent demyelination. The underlying mechanisms of cuprizone-induced oligodendrocyte death are still unknown, and appropriate in vitro investigations to study these mechanisms are not available. Thus, we studied cuprizone effects on rat primary glial cell cultures and on the neuroblastoma cell line SH-SY5Y. Treatment of cells with different concentrations of cuprizone failed to show effects on the proliferation and survival of SH-SY5Y cells, microglia, astrocytes, and oligodendrocyte precursor cells (OPC). In contrast, differentiated mature oligodendrocytes (OL) were found to be significantly affected by cuprizone treatment. This was accompanied by a reduced mitochondrial potential in cuprizone-treated OL. These results demonstrate that the main toxic target for cuprizone is mature OL, whilst other glial cells including OPC are not or only marginally affected. This explains the selective demyelination induced by cuprizone in vivo.
Collapse
Affiliation(s)
- Karelle Bénardais
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str-1, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Wyrsch P, Blenn C, Pesch T, Beneke S, Althaus FR. Cytosolic Ca2+ shifts as early markers of cytotoxicity. Cell Commun Signal 2013; 11:11. [PMID: 23384168 PMCID: PMC3762065 DOI: 10.1186/1478-811x-11-11] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 01/30/2013] [Indexed: 01/07/2023] Open
Abstract
The determination of the cytotoxic potential of new and so far unknown compounds as well as their metabolites is fundamental in risk assessment. A variety of strategic endpoints have been defined to describe toxin-cell interactions, leading to prediction of cell fate. They involve measurement of metabolic endpoints, bio-energetic parameters or morphological cell modifications. Here, we evaluated alterations of the free cytosolic Ca2+ homeostasis using the Fluo-4 dye and compared results with the metabolic cell viability assay Alamar Blue. We investigated a panel of toxins (As2O3, gossypol, H2O2, staurosporine, and titanium(IV)-salane complexes) in four different mammalian cell lines covering three different species (human, mouse, and African green monkey). All tested compounds induced an increase in free cytosolic Ca2+ within the first 5 s after toxin application. Cytosolic Ca2+ shifts occurred independently of the chemical structure in all tested cell systems and were persistent up to 3 h. The linear increase of free cytosolic Ca2+ within the first 5 s of drug treatment correlates with the EC25 and EC75 values obtained in Alamar Blue assays one day after toxin exposure. Moreover, a rise of cytosolic Ca2+ was detectable independent of induced cell death mode as assessed by caspase and poly(ADP-ribose) polymerase (PARP) activity in HeLa versus MCF-7 cells at very low concentrations. In conclusion, a cytotoxicity assay based on Ca2+ shifts has a low limit of detection (LOD), is less time consuming (at least 24 times faster) compared to the cell viability assay Alamar Blue and is suitable for high-troughput-screening (HTS).
Collapse
Affiliation(s)
- Philippe Wyrsch
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, Zurich, CH-8057, Switzerland.
| | | | | | | | | |
Collapse
|
130
|
Gloria A, Russo T, D'Amora U, Zeppetelli S, D'Alessandro T, Sandri M, Bañobre-López M, Piñeiro-Redondo Y, Uhlarz M, Tampieri A, Rivas J, Herrmannsdörfer T, Dediu VA, Ambrosio L, De Santis R. Magnetic poly(ε-caprolactone)/iron-doped hydroxyapatite nanocomposite substrates for advanced bone tissue engineering. J R Soc Interface 2013; 10:20120833. [PMID: 23303218 DOI: 10.1098/rsif.2012.0833] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In biomedicine, magnetic nanoparticles provide some attractive possibilities because they possess peculiar physical properties that permit their use in a wide range of applications. The concept of magnetic guidance basically spans from drug delivery and hyperthermia treatment of tumours, to tissue engineering, such as magneto-mechanical stimulation/activation of cell constructs and mechanosensitive ion channels, magnetic cell-seeding procedures, and controlled cell proliferation and differentiation. Accordingly, the aim of this study was to develop fully biodegradable and magnetic nanocomposite substrates for bone tissue engineering by embedding iron-doped hydroxyapatite (FeHA) nanoparticles in a poly(ε-caprolactone) (PCL) matrix. X-ray diffraction analyses enabled the demonstration that the phase composition and crystallinity of the magnetic FeHA were not affected by the process used to develop the nanocomposite substrates. The mechanical characterization performed through small punch tests has evidenced that inclusion of 10 per cent by weight of FeHA would represent an effective reinforcement. The inclusion of nanoparticles also improves the hydrophilicity of the substrates as evidenced by the lower values of water contact angle in comparison with those of neat PCL. The results from magnetic measurements confirmed the superparamagnetic character of the nanocomposite substrates, indicated by a very low coercive field, a saturation magnetization strictly proportional to the FeHA content and a strong history dependence in temperature sweeps. Regarding the biological performances, confocal laser scanning microscopy and AlamarBlue assay have provided qualitative and quantitative information on human mesenchymal stem cell adhesion and viability/proliferation, respectively, whereas the obtained ALP/DNA values have shown the ability of the nanocomposite substrates to support osteogenic differentiation.
Collapse
Affiliation(s)
- A Gloria
- Institute of Composite and Biomedical Materials, National Research Council, Naples 80125, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Fei Z, Wu Y, Sharma S, Gallego-Perez D, Higuita-Castro N, Hansford D, Lannutti JJ, Lee LJ. Gene Delivery to Cultured Embryonic Stem Cells Using Nanofiber-Based Sandwich Electroporation. Anal Chem 2013; 85:1401-7. [DOI: 10.1021/ac302140p] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Zhengzheng Fei
- William G. Lowrie
Department
of Chemical and Biomolecular Engineering, The Ohio State University, 125A Koffolt Laboratories, 140 West 19th
Avenue, Columbus, Ohio 43210, United States
- NSF Nanoscale Science and Engineering
Center for Affordable Nanoengineering of Polymer Biomedical Devices, The Ohio State University, 174 W 18th Avenue, Room
1012, Columbus, Ohio 43210, United States
| | - Yun Wu
- NSF Nanoscale Science and Engineering
Center for Affordable Nanoengineering of Polymer Biomedical Devices, The Ohio State University, 174 W 18th Avenue, Room
1012, Columbus, Ohio 43210, United States
| | - Sadhana Sharma
- NSF Nanoscale Science and Engineering
Center for Affordable Nanoengineering of Polymer Biomedical Devices, The Ohio State University, 174 W 18th Avenue, Room
1012, Columbus, Ohio 43210, United States
| | - Daniel Gallego-Perez
- NSF Nanoscale Science and Engineering
Center for Affordable Nanoengineering of Polymer Biomedical Devices, The Ohio State University, 174 W 18th Avenue, Room
1012, Columbus, Ohio 43210, United States
| | - Natalia Higuita-Castro
- NSF Nanoscale Science and Engineering
Center for Affordable Nanoengineering of Polymer Biomedical Devices, The Ohio State University, 174 W 18th Avenue, Room
1012, Columbus, Ohio 43210, United States
- Department of Biomedical Engineering, The Ohio State University, 270 Bevis Hall, 1080 Carmack
Road, Columbus, Ohio 43210, United States
| | - Derek Hansford
- NSF Nanoscale Science and Engineering
Center for Affordable Nanoengineering of Polymer Biomedical Devices, The Ohio State University, 174 W 18th Avenue, Room
1012, Columbus, Ohio 43210, United States
- Department of Biomedical Engineering, The Ohio State University, 270 Bevis Hall, 1080 Carmack
Road, Columbus, Ohio 43210, United States
| | - John J. Lannutti
- NSF Nanoscale Science and Engineering
Center for Affordable Nanoengineering of Polymer Biomedical Devices, The Ohio State University, 174 W 18th Avenue, Room
1012, Columbus, Ohio 43210, United States
- Department of Materials Science
and Engineering, The Ohio State University, 477 W Hall, 2041 College Road, Columbus, Ohio 43210, United States
| | - Ly James Lee
- William G. Lowrie
Department
of Chemical and Biomolecular Engineering, The Ohio State University, 125A Koffolt Laboratories, 140 West 19th
Avenue, Columbus, Ohio 43210, United States
- NSF Nanoscale Science and Engineering
Center for Affordable Nanoengineering of Polymer Biomedical Devices, The Ohio State University, 174 W 18th Avenue, Room
1012, Columbus, Ohio 43210, United States
| |
Collapse
|
132
|
Nimesh S. Characterization of nanoparticles: in vitro and in vivo. Gene Ther 2013. [DOI: 10.1533/9781908818645.65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
133
|
Wu X, Sooman L, Lennartsson J, Bergström S, Bergqvist M, Gullbo J, Ekman S. Microtubule inhibition causes epidermal growth factor receptor inactivation in oesophageal cancer cells. Int J Oncol 2012; 42:297-304. [PMID: 23174948 DOI: 10.3892/ijo.2012.1710] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 11/02/2012] [Indexed: 11/06/2022] Open
Abstract
Drugs that interfere with microtubule function can prevent cells from mitosis and may cause cell cycle arrest or apoptosis. Various microtubule targeting agents, both stabilizers and inhibitors, are used in a clinical setting to treat cancer. In the current study, we investigated the sensitivity of oesophageal cancer cells to different microtubule targeting agents. The current study demonstrated that different microtubule targeting agents disrupted the microtubule network and inhibited survival of oesophageal cancer cells in a dose-dependent manner. Interestingly, an additional cellular effect with inhibition of tyrosine phosphorylation of the EGFR and subsequent downregulation of EGFR-induced signalling was also observed, suggesting an additional mechanism of action for microtubule destabilising agents. A tyrosine phosphatase inhibitor, sodium orthovanadate, could reverse the EGFR dephosphorylation effects induced by microtubule targeting agents. The EGFR dephosphorylation could be reversed by a tyrosine phosphatase inhibitor, indicating that disruption of the microtubule network may lead to activation of a protein tyrosine phosphatase (PTP) that can regulate EGFR phosphorylation and activation, an effect of potential clinical relevance for combination therapies in patients.
Collapse
Affiliation(s)
- Xuping Wu
- The Second Hospital of Nanjing affiliated to Southeast University, Nanjing, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
134
|
Haider N, Nuß S. Weinreb amidation as the cornerstone of an improved synthetic route to A-ring-modified derivatives of luotonin A. Molecules 2012; 17:11363-78. [PMID: 23011278 PMCID: PMC6268516 DOI: 10.3390/molecules171011363] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 09/14/2012] [Accepted: 09/18/2012] [Indexed: 11/17/2022] Open
Abstract
Weinreb amidation of ethyl 4-oxo-3,4-dihydroquinazoline-2-carboxylate with aromatic amines provides a significantly improved route to anilide-type key intermediates for the synthesis of the anticancer alkaloid, luotonin A, and new A-ring-modified derivatives thereof. This method has advantages concerning overall yield, brevity, and versatility with regard to the aromatic amine component, even if the latter has less favourable nucleophilicity, solubility and/or stability properties. This is demonstrated by the concise synthesis of a small library of luotonin A analogues, including a novel thiophene isostere of the alkaloid.
Collapse
Affiliation(s)
- Norbert Haider
- Department of Drug and Natural Product Synthesis, Faculty of Life Sciences, University of Vienna, Althanstraße 14, A-1090 Vienna, Austria.
| | | |
Collapse
|
135
|
Rampersad SN. Multiple applications of Alamar Blue as an indicator of metabolic function and cellular health in cell viability bioassays. SENSORS (BASEL, SWITZERLAND) 2012; 12:12347-60. [PMID: 23112716 PMCID: PMC3478843 DOI: 10.3390/s120912347] [Citation(s) in RCA: 675] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/21/2012] [Accepted: 08/31/2012] [Indexed: 02/07/2023]
Abstract
Accurate prediction of the adverse effects of test compounds on living systems, detection of toxic thresholds, and expansion of experimental data sets to include multiple toxicity end-point analysis are required for any robust screening regime. Alamar Blue is an important redox indicator that is used to evaluate metabolic function and cellular health. The Alamar Blue bioassay has been utilized over the past 50 years to assess cell viability and cytotoxicity in a range of biological and environmental systems and in a number of cell types including bacteria, yeast, fungi, protozoa and cultured mammalian and piscine cells. It offers several advantages over other metabolic indicators and other cytotoxicity assays. However, as with any bioassay, suitability must be determined for each application and cell model. This review seeks to highlight many of the important considerations involved in assay use and design in addition to the potential pitfalls.
Collapse
Affiliation(s)
- Sephra N Rampersad
- Department of Life Sciences, The University of the West Indies, West Indies, St Augustine, Trinidad and Tobago.
| |
Collapse
|
136
|
Shukla NM, Salunke DB, Balakrishna R, Mutz CA, Malladi SS, David SA. Potent adjuvanticity of a pure TLR7-agonistic imidazoquinoline dendrimer. PLoS One 2012; 7:e43612. [PMID: 22952720 PMCID: PMC3429503 DOI: 10.1371/journal.pone.0043612] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 07/26/2012] [Indexed: 01/25/2023] Open
Abstract
Engagement of toll-like receptors (TLRs) serve to link innate immune responses with adaptive immunity and can be exploited as powerful vaccine adjuvants for eliciting both primary and anamnestic immune responses. TLR7 agonists are highly immunostimulatory without inducing dominant proinflammatory cytokine responses. We synthesized a dendrimeric molecule bearing six units of a potent TLR7/TLR8 dual-agonistic imidazoquinoline to explore if multimerization of TLR7/8 would result in altered activity profiles. A complete loss of TLR8-stimulatory activity with selective retention of the TLR7-agonistic activity was observed in the dendrimer. This was reflected by a complete absence of TLR8-driven proinflammatory cytokine and interferon (IFN)-γ induction in human PBMCs, with preservation of TLR7-driven IFN-α induction. The dendrimer was found to be superior to the imidazoquinoline monomer in inducing high titers of high-affinity antibodies to bovine α-lactalbumin. Additionally, epitope mapping experiments showed that the dendrimer induced immunoreactivity to more contiguous peptide epitopes along the amino acid sequence of the model antigen.
Collapse
Affiliation(s)
- Nikunj M. Shukla
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Deepak B. Salunke
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Rajalakshmi Balakrishna
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Cole A. Mutz
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Subbalakshmi S. Malladi
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Sunil A. David
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, United States of America
- * E-mail:
| |
Collapse
|
137
|
Membrane disruption by antimicrobial fatty acids releases low-molecular-weight proteins from Staphylococcus aureus. J Bacteriol 2012; 194:5294-304. [PMID: 22843840 DOI: 10.1128/jb.00743-12] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The skin represents an important barrier for pathogens and is known to produce fatty acids that are toxic toward gram-positive bacteria. A screen of fatty acids as growth inhibitors of Staphylococcus aureus revealed structure-specific antibacterial activity. Fatty acids like oleate (18:1Δ9) were nontoxic, whereas palmitoleate (16:1Δ9) was a potent growth inhibitor. Cells treated with 16:1Δ9 exhibited rapid membrane depolarization, the disruption of all major branches of macromolecular synthesis, and the release of solutes and low-molecular-weight proteins into the medium. Other cytotoxic lipids, such as glycerol ethers, sphingosine, and acyl-amines blocked growth by the same mechanisms. Nontoxic 18:1Δ9 was used for phospholipid synthesis, whereas toxic 16:1Δ9 was not and required elongation to 18:1Δ11 prior to incorporation. However, blocking fatty acid metabolism using inhibitors to prevent acyl-acyl carrier protein formation or glycerol-phosphate acyltransferase activity did not increase the toxicity of 18:1Δ9, indicating that inefficient metabolism did not play a determinant role in fatty acid toxicity. Nontoxic 18:1Δ9 was as toxic as 16:1Δ9 in a strain lacking wall teichoic acids and led to growth arrest and enhanced release of intracellular contents. Thus, wall teichoic acids contribute to the structure-specific antimicrobial effects of unsaturated fatty acids. The ability of poorly metabolized 16:1 isomers to penetrate the cell wall defenses is a weakness that has been exploited by the innate immune system to combat S. aureus.
Collapse
|
138
|
Hamalainen-Laanaya HK, Orloff MS. Analysis of cell viability using time-dependent increase in fluorescence intensity. Anal Biochem 2012; 429:32-8. [PMID: 22796501 DOI: 10.1016/j.ab.2012.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 06/14/2012] [Accepted: 07/03/2012] [Indexed: 10/28/2022]
Abstract
We report the use of resazurin (AlamarBlue) dye in a robust assay for cell viability of primary cells. Human mononuclear cells were used here for immunological studies, but the method can be applied to monitor reduction potential of any living cell. Reduction of AlamarBlue dye is widely used in several commercial assays of cell viability. Although it is fast and easy with immortal cell lines, the method is impractical for the primary cells due to their slower metabolic activity. We propose that the viability of human primary cells can be determined with AlamarBlue by monitoring the increase in fluorescence intensity in a matter of a few hours. In the presence of AlamarBlue, the dynamic increase in cellular reduction capacity is linear for several hours or, alternatively, the assay can be repeated to monitor the viability at any time point of cell culture. In addition to testing cellular growth rates and cytotoxicity, the application can be used to compare sample quality of cells that have been frozen or represent a pool of multiple donors. This application of the AlamarBlue cell viability assay is simple, rapid, and cost-effective, and therefore it is also well suited for high-throughput studies.
Collapse
Affiliation(s)
- H K Hamalainen-Laanaya
- Division of Solid Organ Transplantation, Department of Surgery, University of Rochester Medical Center, Rochester, NY 14623, USA.
| | | |
Collapse
|
139
|
Breuer S, Chang MW, Yuan J, Torbett BE. Identification of HIV-1 inhibitors targeting the nucleocapsid protein. J Med Chem 2012; 55:4968-77. [PMID: 22587465 DOI: 10.1021/jm201442t] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The HIV-1 nucleocapsid (NC) is a RNA/DNA binding protein encoded within the Gag polyprotein, which is critical for the selection and chaperoning of viral genomic RNA during virion assembly. RNA/DNA binding occurs through a highly conserved zinc-knuckle motif present in NC. Given the necessity of NC-viral RNA/DNA interaction for viral replication, identification of compounds that disrupt the NC-RNA/DNA interaction may have value as an antiviral strategy. To identify small molecules that disrupt NC-viral RNA/DNA binding, a high-throughput fluorescence polarization assay was developed and a library of 14,400 diverse, druglike compounds was screened. Compounds that disrupted NC binding to a fluorescence-labeled DNA tracer were next evaluated by differential scanning fluorimetry to identify compounds that must bind to NC or Gag to impart their effects. Two compounds were identified that inhibited NC-DNA interaction, specifically bound NC with nanomolar affinity, and showed modest anti-HIV-1 activity in ex vivo cell assays.
Collapse
Affiliation(s)
- Sebastian Breuer
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | | | | | |
Collapse
|
140
|
Dhandayuthapani B, Poulose AC, Nagaoka Y, Hasumura T, Yoshida Y, Maekawa T, Kumar DS. Biomimetic smart nanocomposite: in vitro biological evaluation of zein electrospun fluorescent nanofiber encapsulated CdS quantum dots. Biofabrication 2012; 4:025008. [PMID: 22592161 DOI: 10.1088/1758-5082/4/2/025008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
New hybrid quantum dot (QD)/nanofibers have potential applications in a variety of fields. A novel fluorescent nanocomposite nanofiber material, consisting of CdS and zein has been fabricated through the electrospinning process. A detailed optimization was carried out to fabricate continuous and uniform nanofibers without beads or droplets. The synthesized hybrid nanofibers were characterized by various state-of-the-art techniques such as scanning electron microscopy, transmission electron microscopy (TEM), TEM-energy dispersive spectrometry, atomic force microscopy and confocal fluorescence micrography. The optimization process was carried out to fabricate fibers ranging from 200 to 450 nm in diameter. The electrical conductivity of the zein-CdS hybrid nanofiber substrates was tested. The potential use of the electrospun CdS-encapsulated nanofibrous scaffold as substrates for cell/tissue culture was evaluated with two different cell types, i.e. mesenchymal stem cells and fibroblasts. The results showed that the electrospun fibrous scaffolds could support the attachment and the proliferation of cells. In addition, the cells cultured on the fibrous scaffolds exhibited normal cell shapes and integrated well with surrounding fibers. The obtained results confirmed the potential for the use of the electrospun QD-encapsulated fluorescent nanofiber mats as scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Brahatheeswaran Dhandayuthapani
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan
| | | | | | | | | | | | | |
Collapse
|
141
|
Dessì M, Raucci MG, Zeppetelli S, Ambrosio L. Design of injectable organic-inorganic hybrid for bone tissue repair. J Biomed Mater Res A 2012; 100:2063-70. [PMID: 22581691 DOI: 10.1002/jbm.a.34112] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 01/18/2012] [Accepted: 01/24/2012] [Indexed: 11/07/2022]
Abstract
Injectable bone substitutes are rapidly gained success in tissue engineering applications for their less invasive surgical aspect. Here, the design and the characterization of a novel degradable paste of PCL reinforced with nanocrystals of hydroxyapatite have been presented aiming to mimic natural tissue. Nanohydroxyapatite has been successfully synthesized via sol-gel technique. Dynamic and steady state viscoelastic properties of the solutions and paste were investigated to control the kinetic of phase transition. Correspondingly, the morphology and composition were characterized via TEM, EDAX, and thermal analysis. Injection test underlines the completely ability of the paste of being injected without altering its features. Preliminary biological study showed that the composite paste is not cytotoxic. The synergistic rheological and biological properties, combined with the positive effect of chemical synthesis method indicate that the composite paste is very suitable as local bone substitute in low-load areas.
Collapse
Affiliation(s)
- Mariagemiliana Dessì
- Institute of Composite and Biomedical Materials, National Research Council of Italy, Piazzale Tecchio 80, 80125 Naples, Italy
| | | | | | | |
Collapse
|
142
|
Huyck L, Ampe C, Van Troys M. The XTT cell proliferation assay applied to cell layers embedded in three-dimensional matrix. Assay Drug Dev Technol 2012; 10:382-92. [PMID: 22574651 DOI: 10.1089/adt.2011.391] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell proliferation, a main target in cancer therapy, is influenced by the surrounding three-dimensional (3D) extracellular matrix (ECM). In vitro drug screening is, thus, optimally performed under conditions in which cells are grown (embedded or trapped) in dense 3D matrices, as these most closely mimic the adhesive and mechanical properties of natural ECM. Measuring cell proliferation under these conditions is, however, technically more challenging compared with two-dimensional (2D) culture and other "3D culture conditions," such as growth on top of a matrix (pseudo-3D) or in spongy scaffolds with large pore sizes. Consequently, such measurements are only slowly applied on a wider scale. To advance this, we report on the equal quality (dynamic range, background, linearity) of measuring the proliferation of cell layers embedded in dense 3D matrices (collagen, Matrigel) compared with cells in 2D culture using the easy (one-step) and in 2D well-validated, 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide (XTT)-assay. The comparison stresses the differences in proliferation kinetics and drug sensitivity of matrix-embedded cells versus 2D culture. Using the specific cell-layer-embedded 3D matrix setup, quantitative measurements of cell proliferation and cell invasion are shown to be possible in similar assay conditions, and cytostatic, cytotoxic, and anti-invasive drug effects can thus be reliably determined and compared in physiologically relevant settings. This approach in the 3D matrix holds promise for improving early-stage, high-throughput drug screening, targeting either highly invasive or highly proliferative subpopulations of cancers or both.
Collapse
Affiliation(s)
- Lynn Huyck
- Department of Medical Protein Research, VIB, Ghent, Belgium
| | | | | |
Collapse
|
143
|
Franklin SP, Stoker AM, Cockrell MK, Pfeiffer FM, Sonny Bal B, Cook JL. Effects of low-temperature hydrogen peroxide gas plasma sterilization on in vitro cytotoxicity of poly(ϵ-caprolactone) (PCL). JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2012; 23:2197-206. [PMID: 22126862 DOI: 10.1163/092050611x612296] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Our objective was to determine whether low-temperature hydrogen peroxide (H2O2) gas plasma sterilization of porous three-dimensional poly(ϵ-caprolactone) (PCL) constructs significantly inhibits cellular metabolism of canine chondrocytes. Porous cylindrical constructs were fabricated using fused deposition modeling and divided into four sterilization groups. Two groups were sterilized with low-temperature H2O2 gas plasma (LTGP) and constructs from one of those groups were subsequently rinsed with Dulbecco's Modified Essential Media (LTGPDM). Constructs in the other two groups were disinfected with either 70% isopropyl alcohol or exposure to UV light. Canine chondrocytes were seeded in 6-well tissue-culture plates and allowed to adhere prior to addition of PCL. Cellular metabolism was assessed by adding resazurin to the tissue-culture wells and assessing conversion of this substrate by viable cells to the fluorescent die resorufin. This process was performed at three times prior to addition of PCL and at four times after addition of PCL to the tissue-culture wells. Metabolism was not significantly different among the different tissue-culture wells at any of the 3 times prior to addition of PCL. Metabolism was significantly different among the treatment groups at 3 of 4 times after addition of PCL to the tissue culture wells. Metabolism was significantly lower with constructs sterilized by LTGP than all other treatment groups at all 3 of these times. We conclude that LTGP sterilization of PCL constructs resulted in significant cytotoxicity to canine chondrocytes when compared to PCL constructs disinfected with either UV light exposure or 70% isopropyl alcohol.
Collapse
|
144
|
Dünnhaupt S, Barthelmes J, Rahmat D, Leithner K, Thurner CC, Friedl H, Bernkop-Schnürch A. S-protected thiolated chitosan for oral delivery of hydrophilic macromolecules: evaluation of permeation enhancing and efflux pump inhibitory properties. Mol Pharm 2012; 9:1331-41. [PMID: 22489677 DOI: 10.1021/mp200598j] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The objective of this study was the investigation of permeation enhancing and P-glycoprotein (P-gp) inhibition effects of a novel thiolated chitosan, the so-named S-protected thiolated chitosan. Mediated by a carbodiimide, increasing amounts of thioglycolic acid (TGA) were covalently bound to chitosan (CS) in the first step of modification. In the second step, these thiol groups of thiolated chitosan were protected by disulfide bond formation with the thiolated aromatic residue 6-mercaptonicotinamide (6-MNA). Mucoadhesive properties of all conjugates were evaluated in vitro on porcine intestinal mucosa based on tensile strength investigations. Permeation enhancing effects were evaluated ex vivo using rat intestinal mucosa and in vitro via Caco-2 cells using the hydrophilic macromolecule FD(4) as the model drug. Caco-2 cells were further used to show P-gp inhibition effects by using Rho-123 as P-gp substrate. Apparent permeability coefficients (P(app)) were calculated and compared to values obtained from each buffer control. Three different thiolated chitosans were generated in the first step of modification, which displayed increasing amounts of covalently attached free thiol groups on the polymer backbone. In the second modification step, more than 50% of these free thiol groups were covalently linked with 6-MNA. Within 3 h of permeation studies on excised rat intestine, P(app) values of all S-protected chitosans were at least 1.3-fold higher compared to those of corresponding thiomers and more than twice as high as that of unmodified chitosan. Additional permeation studies on Caco-2 cells confirmed these results. Because of the chemical modification and higher amount of reactive thiol groups, all S-protected thiolated chitosans exhibit at least 1.4-fold pronounced P-gp inhibition effects in contrast to their corresponding thiomers. These features approve S-protected thiolated chitosan as a promising excipient for various drug delivery systems providing improved permeation enhancing and efflux inhibition effects.
Collapse
Affiliation(s)
- Sarah Dünnhaupt
- Department of Pharmaceutical Technology, Institute of Pharmacy, Leopold-Franzenz-University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
145
|
Gosau M, Bürgers R, Vollkommer T, Holzmann T, Prantl L. Effectiveness of antibacterial copper additives in silicone implants. J Biomater Appl 2012; 28:187-98. [DOI: 10.1177/0885328212441957] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Staphylococcus epidermidis plays a major role in capsular contractures of silicone breast implants. This in vitro study evaluates the antibacterial effect of copper on S. epidermidis in silicone implants. Specimens of a silicone material used for breast augmentation (Cu0) and specimens coated with different copper concentrations (Cu1, Cu2) were artificially aged. Surface roughness and surface free energy were assessed. The specimens were incubated in an S. epidermidis suspension. We assessed the quantification and the viability of adhering bacteria by live/dead cell labeling with fluorescence microscopy. Additionally, inhibition of bacterial growth was evaluated by agar diffusion, broth culture, and quantitative culture of surface bacteria. No significant differences in surface roughness and surface free energy were found between Cu0, Cu1 and Cu2. Aging did not change surface characteristics and the extent of bacterial adhesion. Fluorescence microscopy showed that the quantity of bacteria on Cu0 was significantly higher than that on Cu1 and Cu2. The ratio of dead to total adhering bacteria was significantly lower on Cu0 than on Cu1 and Cu2, and tended to be higher for Cu2 than for Cu1. Quantitative culture showed equal trends. Copper additives seem to have anti-adherence and bactericidal effects on S. epidermidis in vitro.
Collapse
Affiliation(s)
- Martin Gosau
- Department of Cranio-Maxillo-Facial Surgery, University Medical Center Regensburg, Germany
| | - Ralf Bürgers
- Department of Prosthetic Dentistry, University Medical Center Regensburg, Germany
| | - Tobias Vollkommer
- Department of Cranio-Maxillo-Facial Surgery, University Medical Center Regensburg, Germany
| | - Thomas Holzmann
- Institute for Medical Microbiology and Hygiene, University Medical Center Regensburg, Germany
| | - Lukas Prantl
- Department of Trauma and Plastic Surgery, University Medical Center Regensburg, Germany
| |
Collapse
|
146
|
Bayer CL, Trenchard IJ, Peppas NA. Analyzing Polyaniline-poly(2-acrylamido-2-methylpropane sulfonic acid) Biocompatibility with 3T3 Fibroblasts. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2012; 21:623-34. [DOI: 10.1163/156856209x434647] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Carolyn L. Bayer
- a Department of Biomedical Engineering, Center on Biomaterials, Drug Delivery, Bionanotechnology and Molecular Recognition, 1 University Station, C-0400, The University of Texas at Austin, Austin, TX 78712-0231, USA
| | - Isis J. Trenchard
- b Department of Biomedical Engineering, Center on Biomaterials, Drug Delivery, Bionanotechnology and Molecular Recognition, 1 University Station, C-0400, The University of Texas at Austin, Austin, TX 78712-0231, USA
| | - Nicholas A. Peppas
- c Department of Biomedical Engineering, Department of Chemical Engineering, Department of Pharmaceutics, Center on Biomaterials, Drug Delivery, Bionanotechnology and Molecular Recognition, 1 University Station, C-0400, The University of Texas at Austin, Austin, TX 78712-0231, USA;,
| |
Collapse
|
147
|
Tachibana S, Shimomura A, Inadera H. Toxicity monitoring with primary cultured hepatocytes underestimates the acetaminophen-induced inflammatory responses of the mouse liver. TOHOKU J EXP MED 2012; 225:263-72. [PMID: 22083109 DOI: 10.1620/tjem.225.263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In vitro gene expression profiling with isolated hepatocytes has been used to assess the hepatotoxicity of certain chemicals because of animal welfare issues. However, whether an in vitro system can completely replace the in vivo system has yet to be elucidated in detail. Using a focused microarray established in our laboratory, we examined gene expression profiles in the mouse liver and primary cultured hepatocytes after treatment with different doses of acetaminophen, a widely used analgesic that frequently causes liver injury. The acute hepatotoxicity of acetaminophen was confirmed by showing the induction of an oxidative stress marker, heme oxygenase-1, elevated levels of serum transaminase, and histopathological findings. In vivo microarray and network analysis showed that acetaminophen treatment provoked alterations in relation to the inflammatory response, and that tumor necrosis factor-α plays a central role in related pathway alterations. By contrast, pathway analyses in in vitro isolated hepatocytes did not find such prominent changes in the inflammation-related networks compared with the in vivo situation. Thus, although in vitro gene expression profiles are useful for evaluating the direct toxicity of chemicals, indirect toxicities including inflammatory responses mediated by cell-cell interactions or secondary toxicity due to pathophysiological changes in the whole body may be overlooked. Our results indicate that the in vitro hepatotoxicity prediction system using isolated hepatocytes does not fully reflect the in vivo cellular response. An in vitro system may be appropriate, therefore, for high throughput screening to detect the direct hepatotoxicity of a test compound.
Collapse
|
148
|
Karna P, Chagani S, Gundala SR, Rida PCG, Asif G, Sharma V, Gupta MV, Aneja R. Benefits of whole ginger extract in prostate cancer. Br J Nutr 2012; 107:473-84. [PMID: 21849094 PMCID: PMC3426621 DOI: 10.1017/s0007114511003308] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
It is appreciated far and wide that increased and regular consumption of fruits and vegetables is linked with noteworthy anticancer benefits. Extensively consumed as a spice in foods and beverages worldwide, ginger (Zingiber officinale Roscoe) is an excellent source of several bioactive phenolics, including non-volatile pungent compounds such as gingerols, paradols, shogaols and gingerones. Ginger has been known to display anti-inflammatory, antioxidant and antiproliferative activities, indicating its promising role as a chemopreventive agent. Here, we show that whole ginger extract (GE) exerts significant growth-inhibitory and death-inductory effects in a spectrum of prostate cancer cells. Comprehensive studies have confirmed that GE perturbed cell-cycle progression, impaired reproductive capacity, modulated cell-cycle and apoptosis regulatory molecules and induced a caspase-driven, mitochondrially mediated apoptosis in human prostate cancer cells. Remarkably, daily oral feeding of 100 mg/kg body weight of GE inhibited growth and progression of PC-3 xenografts by approximately 56 % in nude mice, as shown by measurements of tumour volume. Tumour tissue from GE-treated mice showed reduced proliferation index and widespread apoptosis compared with controls, as determined by immunoblotting and immunohistochemical methods. Most importantly, GE did not exert any detectable toxicity in normal, rapidly dividing tissues such as gut and bone marrow. To the best of our knowledge, this is the first report to demonstrate the in vitro and in vivo anticancer activity of whole GE for the management of prostate cancer.
Collapse
Affiliation(s)
- Prasanthi Karna
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Sharmeen Chagani
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Sushma R. Gundala
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | | | - Ghazia Asif
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Vibhuti Sharma
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | | | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
149
|
Cadieux JA, Zhang Z, Mattice M, Brownlie-Cutts A, Fu J, Ratkay LG, Kwan R, Thompson J, Sanghara J, Zhong J, Goldberg YP. Synthesis and biological evaluation of substituted pyrazoles as blockers of divalent metal transporter 1 (DMT1). Bioorg Med Chem Lett 2011; 22:90-5. [PMID: 22154351 DOI: 10.1016/j.bmcl.2011.11.069] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 11/14/2011] [Accepted: 11/18/2011] [Indexed: 10/15/2022]
Abstract
Three distinct series of substituted pyrazole blockers of divalent metal transporter 1 (DMT1) were elaborated from the high-throughput screening pyrazolone hit 1. Preliminary hit-to-lead efforts revealed a preference for electron-withdrawing substituents in the 4-amido-5-hydroxypyrazole series 6a-l. In turn, this preference was more pronounced in a series of 4-aryl-5-hydroxypyrazoles 8a-j. The representative analogs 6f and 12f were found to be efficacious in a rodent model of acute iron hyperabsorption. These three series represent promising starting points for lead optimization efforts aimed at the discovery of DMT1 blockers as iron overload therapeutics.
Collapse
Affiliation(s)
- Jay A Cadieux
- Department of Medicinal Chemistry, Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, British Columbia, Canada V5G 4W8.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Mao YW, Tseng HW, Liang WL, Chen IS, Chen ST, Lee MH. Anti-inflammatory and free radial scavenging activities of the constituents isolated from Machilus zuihoensis. Molecules 2011; 16:9451-66. [PMID: 22075574 PMCID: PMC6264439 DOI: 10.3390/molecules16119451] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 11/01/2011] [Accepted: 11/02/2011] [Indexed: 11/16/2022] Open
Abstract
A new biflavonol glycoside, quercetin-3-O-β-D-glucopyranoside-(3'→O-3''')-quercetin-3-O-β-D-galactopyranoside (9), together with eight known compounds was isolated for the first time from the leaves of Machilus zuihoensis Hayata (Lauraceae). The structure of compound 9 was elucidated by various types of spectroscopic data analysis. Analysis of the biological activity assay found that compound 9 showed significant superoxide anion scavenging activity (IC₅₀ is 30.4 μM) and markedly suppressed LPS-induced high mobility group box 1 (HMGB-1) protein secretion in RAW264.7 cells. In addition, the HMGB-1 protein secretion was also inhibited by quercitrin (3), ethyl caffeate (6), and ethyl 3-O-caffeoylquinate (7) treatment. In the LPS-stimulated inducible nitric oxide synthase (iNOS) activation analysis, two known compounds, quercetin (1) and ethyl caffeate (6), were found to markedly suppress nitric oxide (NO) production (IC₅₀ value, 27.6 and 42.9 μM, respectively) in RAW264.7 cells. Additionally, it was determined that ethyl caffeate (6) down-regulated mRNA expressions of iNOS, IL-1β, and IL-10 in the LPS-treatment of RAW264.7 cells via a suppressed NF-kB pathway. These results suggested for the first time that the new compound 9 and other constituents isolated from M. zuihoensis have potential anti-inflammatory and superoxide anion scavenging effects. These constituents may be useful for treating various inflammatory diseases.
Collapse
Affiliation(s)
- Yi-Wen Mao
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; (Y.-W.M.); (W.-L.L.)
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Hsiang-Wen Tseng
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 300, Taiwan; (H.-W.T.)
| | - Wen-Li Liang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; (Y.-W.M.); (W.-L.L.)
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Ih-Sheng Chen
- Graduate Institute of Pharmaceutical Sciences, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (I.-S.C.)
| | - Shui-Tein Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 106, Taiwan
- Authors to whom correspondence should be addressed; (S.-T.C.); (M.-H.L.); Tel.: +886-(2)-2732-6752 (S.-T.C.); +886-(2)-2736-1661 (ext. 6151) (M.-H.L.); Fax: +886-(2)-27883473 (S.-T.C.); +886-(2)-27357983(M.-H.L.)
| | - Mei-Hsien Lee
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Center for Reproductive Medicine & Sciences, Taipei Medical University Hospital, Taipei 110, Taiwan
- Authors to whom correspondence should be addressed; (S.-T.C.); (M.-H.L.); Tel.: +886-(2)-2732-6752 (S.-T.C.); +886-(2)-2736-1661 (ext. 6151) (M.-H.L.); Fax: +886-(2)-27883473 (S.-T.C.); +886-(2)-27357983(M.-H.L.)
| |
Collapse
|