Augmentation of bleomycin-induced DNA damage in intact cells

1989 ◽  
Vol 257 (5) ◽  
pp. C882-C887 ◽  
Author(s):  
P. L. Moseley

Bleomycin, an important cause of pulmonary fibrosis, is known to produce DNA damage. The mechanism for this damage in vitro is related to free radical production by a bleomycin and iron complex. To determine whether bleomycin causes damage to DNA in vivo by a similar mechanism, we used a viral minichromosome that is replicated in cultured cells. Bleomycin causes dose-dependent damage to intracellular DNA, and this damage is augmented by Fe2+ but not Fe3+. The augmentation of the bleomycin-induced DNA damage caused by Fe2+ is also dose dependent in that increasing DNA damage occurs with increasing amounts of Fe2+. These studies demonstrate that bleomycin causes damage to DNA in vivo and suggest that bleomycin must rely on Fe2+ to donate an electron for oxygen radical-induced DNA strand scission.

2020 ◽  
Vol 61 (6) ◽  
pp. 896-910 ◽  
Author(s):  
Eyad Naser ◽  
Stephanie Kadow ◽  
Fabian Schumacher ◽  
Zainelabdeen H. Mohamed ◽  
Christian Kappe ◽  
...  

Inhibition of acid sphingomyelinase (ASM), a lysosomal enzyme that catalyzes the hydrolysis of sphingomyelin into ceramide and phosphorylcholine, may serve as an investigational tool or a therapeutic intervention to control many diseases. Specific ASM inhibitors are currently not sufficiently characterized. Here, we found that 1-aminodecylidene bis-phosphonic acid (ARC39) specifically and efficiently (>90%) inhibits both lysosomal and secretory ASM in vitro. Results from investigating sphingomyelin phosphodiesterase 1 (SMPD1/Smpd1) mRNA and ASM protein levels suggested that ARC39 directly inhibits ASM’s catalytic activity in cultured cells, a mechanism that differs from that of functional inhibitors of ASM. We further provide evidence that ARC39 dose- and time-dependently inhibits lysosomal ASM in intact cells, and we show that ARC39 also reduces platelet- and ASM-promoted adhesion of tumor cells. The observed toxicity of ARC39 is low at concentrations relevant for ASM inhibition in vitro, and it does not strongly alter the lysosomal compartment or induce phospholipidosis in vitro. When applied intraperitoneally in vivo, even subtoxic high doses administered short-term induced sphingomyelin accumulation only locally in the peritoneal lavage without significant accumulation in plasma, liver, spleen, or brain. These findings require further investigation with other possible chemical modifications. In conclusion, our results indicate that ARC39 potently and selectively inhibits ASM in vitro and highlight the need for developing compounds that can reach tissue concentrations sufficient for ASM inhibition in vivo.


Toxins ◽  
2020 ◽  
Vol 12 (6) ◽  
pp. 348 ◽  
Author(s):  
Leticia Díez-Quijada ◽  
Concepción Medrano-Padial ◽  
María Llana-Ruiz-Cabello ◽  
Giorgiana M. Cătunescu ◽  
Rosario Moyano ◽  
...  

Cylindrospermopsin (CYN) and microcystins (MC) are cyanotoxins that can occur simultaneously in contaminated water and food. CYN/MC-LR mixtures previously investigated in vitro showed an induction of micronucleus (MN) formation only in the presence of the metabolic fraction S9. When this is the case, the European Food Safety Authority recommends a follow up to in vivo testing. Thus, rats were orally exposed to 7.5 + 75, 23.7 + 237, and 75 + 750 μg CYN/MC-LR/kg body weight (b.w.). The MN test in bone marrow was performed, and the standard and modified comet assays were carried out to measure DNA strand breaks or oxidative DNA damage in stomach, liver, and blood cells. The results revealed an increase in MN formation in bone marrow, at all the assayed doses. However, no DNA strand breaks nor oxidative DNA damage were induced, as shown in the comet assays. The histopathological study indicated alterations only in the highest dose group. Liver was the target organ showing fatty degeneration and necrotic hepatocytes in centrilobular areas, as well as a light mononuclear inflammatory periportal infiltrate. Additionally, the stomach had flaking epithelium and mild necrosis of epithelial cells. Therefore, the combined exposure to cyanotoxins may induce genotoxic and histopathological damage in vivo.


Blood ◽  
2005 ◽  
Vol 106 (13) ◽  
pp. 4131-4138 ◽  
Author(s):  
Miriam Erlacher ◽  
Ewa M. Michalak ◽  
Priscilla N. Kelly ◽  
Verena Labi ◽  
Harald Niederegger ◽  
...  

Numerous p53 target genes have been implicated in DNA damage–induced apoptosis signaling, but proapoptotic Bcl-2 (B-cell leukemia 2) family members of the BH3 (Bcl-2 homolog region [BH] 3)–only subgroup appear to play the critical initiating role. In various types of cultured cells, 3 BH3-only proteins, namely Puma (p53 up-regulated modulator of apoptosis), Noxa, and Bim (Bcl-2 interacting mediator of cell death), have been shown to initiate p53-dependent as well as p53-independent apoptosis in response to DNA damage and treatment with anticancer drugs or glucocorticoids. In particular, the absence of Puma or Bim renders thymocytes and mature lymphocytes refractory to varying degrees to death induced in vitro by growth factor withdrawal, DNA damage, or glucocorticoids. To assess the in vivo relevance of these findings, we subjected mice lacking Puma, Noxa, or Bim to whole-body γ-radiation or the glucocorticoid dexamethasone and compared lymphocyte survival with that in wild-type and BCL2–transgenic mice. Absence of Puma or Bcl-2 overexpression efficiently protected diverse types of lymphocytes from the effects of γ-radiation in vivo, and loss of Bim provided lower but significant protection in most lymphocytes, whereas Noxa deficiency had no impact. Furthermore, both Puma and Bim were found to contribute significantly to glucocorticoid-induced killing. Our results thus establish that Puma and Bim are key initiators of γ-radiation– and glucocorticoid-induced apoptosis in lymphoid cells in vivo.


2000 ◽  
Vol 53 (1) ◽  
pp. 7 ◽  
Author(s):  
Lianbo Zhang ◽  
Peter A. Lay

The reaction of chromium(VI) with L-ascorbic acid (AsA) in buffer solutions was investigated by e.p.r. spectroscopy. Chromium(V)/ascorbate complexes with signals at giso = 1.9791 and chromium(VI)/ascorbate/peroxo complexes with signals at giso = 1.9819 and giso = 1.9824 were observed in all buffers. New signals at giso values of 1.9765 and 1.9781 were observed in Tris–HCl [tris(hydroxymethyl)aminomethane hydrochloride] buffer and were assigned to a mixed-ligand ascorbate/Tris complex of chromium(V), [CrO(ascorbate)(Tris)]2– , and a Tris/peroxo species, [CrO(O2)(Tris)]2– , respectively. The speciation of the e.p.r.-active chromium(V) complexes detected from solutions with other buffers, such as HEPES, cacodylate and phosphate, was not influenced by the buffer type. Preincubation of catalase with ascorbate solutions inhibited the formation of the chromium(V) peroxo species in all buffers. Manganese(II) reduces the chromium(V) species produced in the reaction, which has shown that it is inappropriate to use manganeses(II) as a selective reagent for monitoring the concentrations of chromium(IV) in such reactions. In particular, manganeses(II) reacts more efficiently with the chromium(V) species that are most damaging to DNA in vitro, viz., the mixed-ligand chromium(V)/ascorbate/peroxo complexes. The correlation of the present results with those of in vitro DNA damage experiments reported in the literature has revealed that the chromium(V)/ascorbate/peroxo species are the major species responsible for the in vitro DNA strand breaks in all of the buffer systems. These species are not expected to be as important in vivo.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1371-1371
Author(s):  
Scott M. Portwood ◽  
Marianna C Cantella ◽  
Tara L. Cronin ◽  
Eunice S. Wang

Background CD33 (Siglec3) is a cell surface transmembrane receptor that is rapidly internalized and highly expressed on AML blasts but is absent on normal hematopoietic stem cells. Gemtuzumab ozogamicin (GO), a humanized anti-CD33 antibody conjugated to a DNA strand scission inducing agent (calicheamicin) was recently FDA approved for the treatment of newly-diagnosed or relapsed/refractory CD33-positive acute myeloid leukemia (AML). GO has been shown to exert clinical activity in leukemia patients. Poly (ADP-ribose) polymerase (PARP) inhibitors prevent the repair of single stranded DNA breaks by blocking the nicotinamide adenine dinucleotide (NAD) catalytic domain of the PARP protein and by preventing the dissociation of PARP from the DNA (PARP trapping). Talazoparib is unique among clinical PARP inhibitors in displaying 10,000-fold increased PARP trapping as compared to other agents . We hypothesized that combination therapy using GO and Talazoparib would result in synergistic anti-leukemic effects on human CD33+ AML cells due to the ability of the PARP inhibitor to enhance levels of DNA damage induced by GO therapy. Materials and Methods Human AML cell lines were characterized for CD33 expression using flow cytometry after staining with antibody-linked fluorescent QuantiBrite Beads. Cells were continuously exposed to varying doses of GO (10pM - 100mM) and PARP inhibitors (1nM - 100mM) for 96h alone and in combination. Cell viability was measured immediately following treatment using a WST colorimetric assay. Treatment-induced apoptosis (annexin/PI) and DNA damage (H2AX) were quantified by flow cytometric assays. Synergy reports were generated using Compusyn software. In vivo efficacy was assessed in NSG mice systematically engrafted with luciferase labeled human CD33+ AML cells following tail vein injection. Animals were treated with varying doses of vehicle, GO (1 and 50ug/kg 1x/week for 3 weeks), or talazoparib (0.1 and 0.33mg/kg 5 days/week) either alone or in combination. Treatment effects on leukemia burden, toxicity, and survival were determined by weekly whole animal bioluminescent imaging, total animal weights, and time to morbidity. Results Human AML cell lines (HEL, HL60) express high expression levels of CD33 molecules/cell (43,645 and 31,286 respectively) relative to negative controls. Continuous exposure to single agent GO and Talazoparib for 96h resulted in a dose dependent inhibition of human AML cell growth (HEL, HL60) . IC values for GO were 0.01 - 6.6μg/ml and for Talazoparib were 0.8-0.95μM. Combination in vitro therapy with GO (0.005 - 1μg/ml) and Talazoparib (fixed dose 100nM) resulted in synergistic anti-leukemic effects (p<0.01) significantly improving upon monotherapy. Software analyses yielded a combination Index (CI) <1 consistent with synergistic anti-leukemic effects. Combination GO and Talazoparib therapy also significantly enhanced AML cell apoptosis (p=0.0111) and levels of DNA damage (phosphorylated H2AX) (p=0.0054) over single agent activity. Evaluation of PARP trapping by western blot analysis is ongoing. In vivo administration of GO (1-50μg/kg) and Talazoparib (0.1-0.33mg/kg) in NSG mice with systemic engraftment of luciferase tagged human CD33+ AML cells was generally well tolerated with no significant weight loss or early morbidity. Single agent GO and Talazoparib therapy decreased systemic AML burden in a dose dependent manner and prolonged overall survival over vehicle treated mice (P<0.05). Concomitant GO (1μg/kg) and Talazoparib (0.33μg/kg) treatment was similarly well tolerated with no notable weight loss or toxicities. Combination GO and Talazoparib therapy significantly prolonged overall survival of leukemia xenografted mice over vehicle (p=0.0018) and single agent therapy with the same doses of GO (p=0.0018) and Talazoparib (p=0.0499), respectively). Conclusions In summary, our results demonstrate that the addition of the PARP inhibitor, Talazoparib, to the CD33 antibody drug conjugate, GO, results in potent in vitro and in vivo anti-tumor activity in human CD33+ AML preclinical models. Further studies investigating this novel combinatorial approach in AML are currently ongoing. Due to GO's FDA approval for CD33+ AML in 2018, this data strongly supports future clinical investigation using PARP inhibitors as a novel class of agents for combination therapy to significantly enhance the efficacy of ADCs. Figure 1 Disclosures Wang: Amgen: Other: Advisory role; Agios: Other: Advisory role; Stemline: Other: Advisory role, Speakers Bureau; Daiichi: Other: Advisory role; Abbvie: Other: Advisory role; Kite: Other: Advisory role; Jazz: Other: Advisory role; Astellas: Other: Advisory role, Speakers Bureau; celyad: Other: Advisory role; Pfizer: Other: Advisory role, Speakers Bureau.


1987 ◽  
Vol 8 (11) ◽  
pp. 1657-1662 ◽  
Author(s):  
Paul V. Woolley ◽  
Shailendra Kumar ◽  
Peter Fitzgerald ◽  
Robert T. Simpson

1988 ◽  
Vol 253 (2) ◽  
pp. 425-433 ◽  
Author(s):  
E J White ◽  
J B Clark

Menadione bisulphite increased endogenous oxygen-radical production by rat brain synaptosomes, as indicated by H2O2 generation. Increased oxygen-radical production was also demonstrated in synaptosomes prepared from menadione-treated rats and synaptosomes reoxygenated after an anoxic insult. Acetylcholine synthesis de novo was inhibited in synaptosomes incubated with menadione in vitro, in synaptosomes prepared from menadione-treated animals in vivo, and in depolarized post-anoxic synaptosomes. Intrasynaptosomal free Ca2+ was increased by menadione in vitro (50 microM), but this increase was not due to stimulation of Ca2+ entry into the nerve terminals. Acetylcholine release was stimulated by menadione in vitro, possibly as a consequence of the elevated intrasynaptosomal Ca2+ content. The Ca2+ contents of synaptosomes prepared from menadione (10 mg/kg)-treated animals in vivo and synaptosomes reoxygenated after anoxia were unchanged. In synaptosomes prepared from menadione-treated animals, acetylcholine release was no longer significantly stimulated by K+, whereas it was unchanged from control (normoxic) values in synaptosomes reoxygenated after anoxia. None of these treatments caused any measurable damage to the synaptic plasma membrane (as judged by the release of lactate dehydrogenase), or to synaptosomal phospholipases (as judged by choline release from membrane phospholipids). Synaptosomes prepared from menadione-treated rats were found to be a good model for the study of post-anoxic damage to nerve-terminal function.


2021 ◽  
Author(s):  
Likun Duan ◽  
Daniel E. Cooper ◽  
Grace Scheidemantle ◽  
Jason W. Locasale ◽  
David G. Kirsch ◽  
...  

Abstract13C tracing analysis is increasingly used to monitor cellular metabolism in vivo and in intact cells, but data interpretation is still the key element to unveil the complexity of metabolic activities. We have performed [U-13C]-glucose and [U-13C]-glutamine tracing in sarcoma-bearing mice (in vivo) and in cancer cell lines (in vitro). 13C enrichment of metabolites in cultured cells and tissues was determined by liquid chromatography coupled with high-resolution mass spectrometer (LC-HRMS). As expected, citrate M+2 or M+4 is the dominant mass isotopologue in vitro. However, citrate M+1 was unexpectedly the dominant isotopologue in mice receiving [U-13C]-glucose or [U-13C]-glutamine infusion. One plausible explanation is that 13CO2 produced from the oxidation of 13C tracers in vitro is negligible due to the dilution of HCO3- supplemented to cell culture when sodium bicarbonante is used and diffusible volume of CO2 in the culture incubator, while endogenous 13CO2 in vivo is substantial and is fixed into the TCA cycle, purine, and serine, resulting in M+1 isotopologues. A time course study shows the generation of high abundance citrate M+1 early in plasma, which may serve as a potent non-invasive biomarker of tissue pyruvate carboxylase activity. Altogether, our results show that recycling of endogenous CO2 is substantial in vivo and provides important insights into the experimental design and data interpretation of 13C tracing assays.


1989 ◽  
Vol 8 (7) ◽  
pp. 1275-1283 ◽  
Author(s):  
Andrew M. Standeven ◽  
Karen E. Wetterhahn

Much recent data supports the “uptake-reduction” model explaining the carcinogenicity of chromium(VI) compounds and the lack of carcinogenicity of chromium(III) com pounds. Cr(VI) readily enters cells by diffusion through a nonspecific anion channel, whereas cells are relatively impermeable to Cr(III). Glutathione appears to facilitate Cr(VI) uptake by reducing Cr(VI) to Cr(III) after it enters the cell, presumably keeping intracellular Cr(VI) concentration low and allowing for further Cr(VI) uptake. Some other nonenzymatic factors, for example, ascorbate and riboflavin, as well as enzymes, such as cytochrome P-450, DT-diaphorase, and the mitochondrial electron transport chain complexes, are capable of reducing Cr(VI) in vitro, but their contribution in vivo is not clear. Cr(VI), once reduced intracellularly, produces various forms of DNA damage including DNA interstrand crosslinks, DNA-protein crosslinks, DNA strand breaks, and Cr-DNA adducts. The pathway of Cr(VI) metabolism in different tissues appears to influence the type of “reactive intermediates” produced, for example, Cr(V) and radical species, and thus the nature and extent of DNA damage. This DNA damage presumably accounts for observed functional changes in DNA replication and transcription which may be crucial to the carcinogenicity of chromium(VI) compounds.


Author(s):  
M.J. Murphy ◽  
R.R. Price ◽  
J.C. Sloman

The in vitro human tumor cloning assay originally described by Salmon and Hamburger has been applied recently to the investigation of differential anti-tumor drug sensitivities over a broad range of human neoplasms. A major problem in the acceptance of this technique has been the question of the relationship between the cultured cells and the original patient tumor, i.e., whether the colonies that develop derive from the neoplasm or from some other cell type within the initial cell population. A study of the ultrastructural morphology of the cultured cells vs. patient tumor has therefore been undertaken to resolve this question. Direct correlation was assured by division of a common tumor mass at surgical resection, one biopsy being fixed for TEM studies, the second being rapidly transported to the laboratory for culture.


Sign in / Sign up

Export Citation Format

Share Document