Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(7): e0307297, 2024.
Article in English | MEDLINE | ID: mdl-39037974

ABSTRACT

Haemaphysalis longicornis is a common Ixodida tick species found in temperate areas of Asian countries. An anti-tick assay was conducted on adult female H. longicornis ticks. Plant extract solutions were prepared at concentrations of 50, 25, and 10 mg/mL. Tick survival and mortality were assessed by counting the number of dead and live ticks at 24 h, 48 h, 72 h, and 96 h posttreatment. Out of 11 plant extracts screened, Artemisia judaica extract exhibited the highest potency with 100% mortality (5/5) at 48 h when applied at high and moderate concentrations (50 and 25 mg/mL). Similar results were observed at 96 h for the 10 mg/mL group compared to the untreated ticks. Cleome droserifolia extract demonstrated partial activity with 60% (3/5) and 20% (1/5) mortality at 96 h posttreatment at concentrations of 50 and 25 mg/mL, respectively. Forsskaolea tenacissima extract showed a weak effect with 100% tick mortality (5/5) only at the highest treatment concentration after 96 h. To confirm the activity of A. judaica, trial 2 was conducted. A. judaica demonstrated potency within 48 h in high dose and 72 h in moderate dose, with 100% mortality (15/15) at 96 h posttreatment compared to untreated ticks. The median lethal time 50 (LT50) values were 30.37 h for the high and 55.08 h for the moderate doses. Liquid chromatography‒mass spectrometry was performed on the most potent candidate (A. judaica) to identify its phytochemical components. The results revealed the presence of 9 compounds identified through manual annotation and 74 compounds from the Global Natural Products Social library. These compounds included terpenoids, steroids, phenylpropanoids, flavonoid glycosides, flavonoids, and benzenoids. Camphor was identified as the major component via both approaches. These findings suggest the potential use of A. judaica extract in the future development of acaricidal therapeutics.


Subject(s)
Acaricides , Ixodidae , Plant Extracts , Animals , Plant Extracts/pharmacology , Plant Extracts/chemistry , Ixodidae/drug effects , Acaricides/pharmacology , Acaricides/chemistry , Female , Egypt , Haemaphysalis longicornis
2.
Arch Toxicol ; 98(5): 1533-1542, 2024 May.
Article in English | MEDLINE | ID: mdl-38466352

ABSTRACT

Acetaminophen (APAP) is known to cause a breach of the blood-bile barrier in mice that, via a mechanism called futile bile acid (BA) cycling, increases BA concentrations in hepatocytes above cytotoxic thresholds. Here, we compared this mechanism in mice and rats, because both species differ massively in their susceptibility to APAP and compared the results to available human data. Dose and time-dependent APAP experiments were performed in male C57BL6/N mice and Wistar rats. The time course of BA concentrations in liver tissue and in blood was analyzed by MALDI-MSI and LC-MS/MS. APAP and its derivatives were measured in the blood by LC-MS. APAP-induced liver damage was analyzed by histopathology, immunohistochemistry, and by clinical chemistry. In mice, a transient increase of BA in blood and in peri-central hepatocytes preceded hepatocyte death. The BA increase coincided with oxidative stress in liver tissue and a compromised morphology of bile canaliculi and immunohistochemically visualized tight junction proteins. Rats showed a reduced metabolic activation of APAP compared to mice. However, even at very high doses that caused cell death of hepatocytes, no increase of BA concentrations was observed neither in liver tissue nor in the blood. Correspondingly, no oxidative stress was detectable, and the morphology of bile canaliculi and tight junction proteins remained unaltered. In conclusion, different mechanisms cause cell death in rats and mice, whereby oxidative stress and a breach of the blood-bile barrier are seen only in mice. Since transient cholestasis also occurs in human patients with APAP overdose, mice are a clinically relevant species to study APAP hepatotoxicity but not rats.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Mice , Rats , Humans , Male , Animals , Acetaminophen/toxicity , Acetaminophen/metabolism , Bile/metabolism , Chromatography, Liquid , Chemical and Drug Induced Liver Injury/pathology , Rats, Wistar , Tandem Mass Spectrometry , Liver/metabolism , Hepatocytes/metabolism , Mice, Inbred C57BL , Tight Junction Proteins/metabolism
3.
J Hepatol ; 80(2): 268-281, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37939855

ABSTRACT

BACKGROUND & AIMS: Cholemic nephropathy (CN) is a severe complication of cholestatic liver diseases for which there is no specific treatment. We revisited its pathophysiology with the aim of identifying novel therapeutic strategies. METHODS: Cholestasis was induced by bile duct ligation (BDL) in mice. Bile flux in kidneys and livers was visualized by intravital imaging, supported by MALDI mass spectrometry imaging and liquid chromatography-tandem mass spectrometry. The effect of AS0369, a systemically bioavailable apical sodium-dependent bile acid transporter (ASBT) inhibitor, was evaluated by intravital imaging, RNA-sequencing, histological, blood, and urine analyses. Translational relevance was assessed in kidney biopsies from patients with CN, mice with a humanized bile acid (BA) spectrum, and via analysis of serum BAs and KIM-1 (kidney injury molecule 1) in patients with liver disease and hyperbilirubinemia. RESULTS: Proximal tubular epithelial cells (TECs) reabsorbed and enriched BAs, leading to oxidative stress and death of proximal TECs, casts in distal tubules and collecting ducts, peritubular capillary leakiness, and glomerular cysts. Renal ASBT inhibition by AS0369 blocked BA uptake into TECs and prevented kidney injury up to 6 weeks after BDL. Similar results were obtained in mice with humanized BA composition. In patients with advanced liver disease, serum BAs were the main determinant of KIM-1 levels. ASBT expression in TECs was preserved in biopsies from patients with CN, further highlighting the translational potential of targeting ASBT to treat CN. CONCLUSIONS: BA enrichment in proximal TECs followed by oxidative stress and cell death is a key early event in CN. Inhibiting renal ASBT and consequently BA enrichment in TECs prevents CN and systemically decreases BA concentrations. IMPACT AND IMPLICATIONS: Cholemic nephropathy (CN) is a severe complication of cholestasis and an unmet clinical need. We demonstrate that CN is triggered by the renal accumulation of bile acids (BAs) that are considerably increased in the systemic blood. Specifically, the proximal tubular epithelial cells of the kidney take up BAs via the apical sodium-dependent bile acid transporter (ASBT). We developed a therapeutic compound that blocks ASBT in the kidneys, prevents BA overload in tubular epithelial cells, and almost completely abolished all disease hallmarks in a CN mouse model. Renal ASBT inhibition represents a potential therapeutic strategy for patients with CN.


Subject(s)
Carrier Proteins , Cholestasis , Kidney Diseases , Liver Diseases , Membrane Glycoproteins , Organic Anion Transporters, Sodium-Dependent , Symporters , Humans , Mice , Animals , Cholestasis/complications , Cholestasis/metabolism , Kidney/metabolism , Symporters/metabolism , Bile Acids and Salts/metabolism , Liver/metabolism , Bile Ducts/metabolism , Liver Diseases/metabolism , Sodium
4.
Front Pharmacol ; 14: 1279357, 2023.
Article in English | MEDLINE | ID: mdl-38053838

ABSTRACT

Rationale: Liver cirrhosis is known to affect drug pharmacokinetics, but the functional assessment of the underlying pathophysiological alterations in drug metabolism is difficult. Methods: Cirrhosis in mice was induced by repeated treatment with carbon tetrachloride for 12 months. A cocktail of six drugs was administered, and parent compounds as well as phase I and II metabolites were quantified in blood, bile, and urine in a time-dependent manner. Pharmacokinetics were modeled in relation to the altered expression of metabolizing enzymes. In discrepancy with computational predictions, a strong increase of glucuronides in blood was observed in cirrhotic mice compared to vehicle controls. Results: The deviation between experimental findings and computational simulations observed by analyzing different hypotheses could be explained by increased sinusoidal export and corresponded to increased expression of export carriers (Abcc3 and Abcc4). Formation of phase I metabolites and clearance of the parent compounds were surprisingly robust in cirrhosis, although the phase I enzymes critical for the metabolism of the administered drugs in healthy mice, Cyp1a2 and Cyp2c29, were downregulated in cirrhotic livers. RNA-sequencing revealed the upregulation of numerous other phase I metabolizing enzymes which may compensate for the lost CYP isoenzymes. Comparison of genome-wide data of cirrhotic mouse and human liver tissue revealed similar features of expression changes, including increased sinusoidal export and reduced uptake carriers. Conclusion: Liver cirrhosis leads to increased blood concentrations of glucuronides because of increased export from hepatocytes into the sinusoidal blood. Although individual metabolic pathways are massively altered in cirrhosis, the overall clearance of the parent compounds was relatively robust due to compensatory mechanisms.

5.
Arch Toxicol ; 97(10): 2763-2770, 2023 10.
Article in English | MEDLINE | ID: mdl-37401952

ABSTRACT

Chloroquine (CQ) and hydroxychloroquine (HCQ) are classical antimalarial drugs, and recently have been used for other applications including coronavirus disease 2019 (COVID-19). Although they are considered safe, cardiomyopathy may associate CQ and HCQ applications particularly at overdoses. The goal of the present study was to evaluate the potential protective effect of the nootropic agent vinpocetine against CQ and HCQ adverse effects with a specific focus on the heart. For this purpose, a mouse model of CQ (0.5 up to 2.5 g/kg)/HCQ (1 up to 2 g/kg) toxicity was used, and the effect of vinpocetine was evaluated by survival, biochemical, as well as histopathological analyses. Survival analysis revealed that CQ and HCQ caused dose-dependent lethality, which was prevented by co-treatment with vinpocetine (100 mg/kg, oral or intraperitoneal). To gain deeper understanding, a dose of 1 g/kg CQ-which did not cause death within the first 24 h after administration-was applied with and without vinpocetine administration (100 mg/kg, intraperitoneal). The CQ vehicle group showed marked cardiotoxicity as evidenced by significant alterations of blood biomarkers including troponione-1, creatine phosphokinase (CPK), creatine kinase-myocardial band (CK-MB), ferritin, and potassium levels. This was confirmed at the tissue level by massive alteration of the heart tissue morphology and coincided with massive oxidative stress. Interestingly, co-administration of vinpocetine strongly ameliorated CQ-induced alterations and restored the antioxidant-defense system of the heart. These data suggest that vinpocetine could be used as an adjuvant therapy together with CQ/HCQ applications.


Subject(s)
COVID-19 , Chloroquine , Animals , Mice , Chloroquine/toxicity , Cardiotoxicity/prevention & control , SARS-CoV-2 , COVID-19 Drug Treatment , Hydroxychloroquine/toxicity , Hydroxychloroquine/therapeutic use , Oxidative Stress
6.
J Vet Med Sci ; 84(7): 1034-1040, 2022 Jul 25.
Article in English | MEDLINE | ID: mdl-35661076

ABSTRACT

Wild medicinal plants have been traditionally used as antimicrobial agents. Here, we evaluated the in vitro activity of extracts from wild Egyptian desert plants against Toxoplasma gondii and Neospora caninum. From 12 plant extracts tested, the methanolic extracts from Artemisia judaica, Cleome droserifolia, Trichodesma africanum, and Vachellia tortilis demonstrated potent activity against the growth of T. gondii, with half-maximal inhibitory concentrations (IC50s) of 2.1, 12.5, 21.8, and 24.5 µg/ml, respectively. C. droserifolia, an ethanolic extract of P. undulata, T. africanum, A. judaica, and V. tortilis demonstrated potent efficacy against N. caninum, with mean IC50s of 1.0, 3.0, 3.1, 8.6, and 17.2 µg/ml, respectively. Our data suggest these extracts could provide an alternative treatment for T. gondii and N. caninum infections.


Subject(s)
Coccidiosis , Neospora , Toxoplasma , Toxoplasmosis, Animal , Animals , Antibodies, Protozoan , Coccidiosis/drug therapy , Coccidiosis/veterinary , Egypt , Plant Extracts/pharmacology , Seroepidemiologic Studies
7.
BMC Complement Med Ther ; 22(1): 130, 2022 May 12.
Article in English | MEDLINE | ID: mdl-35550108

ABSTRACT

BACKGROUND: Medicinal plants have been successfully used as an alternative source of drugs for the treatment of microbial diseases. Finding a novel treatment for malaria is still challenging, and various extracts from different wild desert plants have been reported to have multiple medicinal uses for human public health, this study evaluated the antimalarial efficacy of several Egyptian plant extracts. METHODS: We assessed the cytotoxic potential of 13 plant extracts and their abilities to inhibit the in vitro growth of Plasmodium falciparum (3D7), and to treat infection with non-lethal Plasmodium yoelii 17XNL in an in vivo malaria model in BALB/c mice. RESULTS: In vitro screening identified four promising candidates, Trichodesma africanum, Artemisia judaica, Cleome droserifolia, and Vachellia tortilis, with weak-to-moderate activity against P. falciparum erythrocytic blood stages with mean half-maximal inhibitory concentration 50 (IC50) of 11.7 µg/ml, 20.0 µg/ml, 32.1 µg/ml, and 40.0 µg/ml, respectively. Their selectivity index values were 35.2, 15.8, 11.5, and 13.8, respectively. Among these four candidates, T. africanum crude extract exhibited the highest parasite suppression in a murine malaria model against P. yoelii. CONCLUSION: Our study identified novel natural antimalarial agents of plant origin that have potential for development into therapeutics for treating malaria.


Subject(s)
Antimalarials , Malaria , Plants, Medicinal , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Egypt , Malaria/drug therapy , Malaria/parasitology , Mice , Plant Extracts/therapeutic use
8.
EXCLI J ; 20: 550-561, 2021.
Article in English | MEDLINE | ID: mdl-34121971

ABSTRACT

Vinpocetine (Vin), a synthetic-derivative of Vincamine, monoterpenoid indole alkaloid, has been reported to have various medicinal benefits. The purpose of our study was to investigate the pivotal role of "nuclear factor erythroid 2-related factor-2" (Nrf2)-mediated antioxidant protection of Vin against H2O2 and paracetamol (APAP)-induced liver toxicity. For this purpose, a normal human hepatic cell line (L02 cells) was incubated with cytotoxic concentrations of H2O2 or APAP in the presence or absence of Vin. To evaluate the responses, MTS Cell Viability assay, immunoblotting, biochemical assays, and molecular docking approach were used. Viability analysis showed that treatment of L02 cells with Vin prevented the cytotoxicity induced by H2O2 and APAP. It was evidenced by the fact that Vin dumped H2O2- and APAP-cytotoxicity and reactive oxygen species (ROS) generation. The immunoblotting analysis shows that Vin increased Nrf2 expression along with the expression of target protein, heme oxygenase-1 (HO-1), and increased intracellular glutathione (GSH) level. Interestingly, we found that Vin could protect the protein expression-level of Nrf2, which indicated the prospective interaction between Vin and Keap1 protein. Additionally, molecular docking-study revealed that Vin competed with Nrf2 for Keap1-binding site, with hydrogen and stearic interactions. Collectively, Vin effectively protects against H2O2 and APAP-induced cytotoxicity via executing Nrf2-mediated restoration of antioxidative/oxidative balance. Meanwhile, Vin interrupts protein-protein interaction between Nrf2 and Keap1, which might also contribute to decrease Nrf2 degradation and stabilize protein expression. Thus, Vin-based adjuvant therapy may represent a smart drug regimen to mitigate drug-induced oxidative stress and liver injuries.

9.
J Lipid Res ; 60(9): 1590-1602, 2019 09.
Article in English | MEDLINE | ID: mdl-31363040

ABSTRACT

The combination of daunorubicin (dnr) and cytarabine (Ara-C) is a cornerstone of treatment for acute myelogenous leukemia (AML); resistance to these drugs is a major cause of treatment failure. Ceramide, a sphingolipid (SL), plays a critical role in cancer cell apoptosis in response to chemotherapy. Here, we investigated the effects of chemotherapy selection pressure with Ara-C and dnr on SL composition and enzyme activity in the AML cell line HL-60. Resistant cells, those selected for growth in Ara-C- and dnr-containing medium (HL-60/Ara-C and HL-60/dnr, respectively), demonstrated upregulated expression and activity of glucosylceramide synthase, acid ceramidase (AC), and sphingosine kinase 1 (SPHK1); were more resistant to ceramide than parental cells; and displayed sensitivity to inhibitors of SL metabolism. Lipidomic analysis revealed a general ceramide deficit and a profound upswing in levels of sphingosine 1-phosphate (S1P) and ceramide 1-phosphate (C1P) in HL-60/dnr cells versus parental and HL-60/Ara-C cells. Both chemotherapy-selected cells also exhibited comprehensive upregulations in mitochondrial biogenesis consistent with heightened reliance on oxidative phosphorylation, a property that was partially reversed by exposure to AC and SPHK1 inhibitors and that supports a role for the phosphorylation system in resistance. In summary, dnr and Ara-C selection pressure induces acute reductions in ceramide levels and large increases in S1P and C1P, concomitant with cell resilience bolstered by enhanced mitochondrial remodeling. Thus, strategic control of ceramide metabolism and further research to define mitochondrial perturbations that accompany the drug-resistant phenotype offer new opportunities for developing therapies that regulate cancer growth.


Subject(s)
Mitochondria/metabolism , Sphingolipids/metabolism , Amides/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Ceramidases/metabolism , Ceramides/metabolism , Fatty Acids, Unsaturated/pharmacology , Glucosyltransferases/metabolism , HL-60 Cells , Humans , Immunoblotting , Lysophospholipids/metabolism , Mass Spectrometry , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/analogs & derivatives , Sphingosine/metabolism
10.
Exp Cell Res ; 381(2): 256-264, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31112736

ABSTRACT

Acute myelogenous leukemia (AML) is a hematological malignancy marked by the accumulation of large numbers of immature myeloblasts in bone marrow. The overall prognosis in AML is poor; hence, there is a pressing need to improve treatment. Although the sphingolipid (SL) ceramide demonstrates known cancer suppressor properties, it's mechanism of action is multifaceted. Our studies in leukemia and other cancers have demonstrated that when combined with the antiestrogen, tamoxifen, the apoptosis-inducting effect of ceramide is greatly enhanced. The goal of the present study was to establish whether a ceramide-tamoxifen regimen also affects autophagic-driven cellular responses in leukemia. Using the human AML cell line KG-1, we demonstrate that, unlike exposure to the single agents, combination C6-ceramide-tamoxifen upregulated LC3-II expression, inhibited the mTOR signaling pathway, and synergistically induced KG-1 cell death in an Atg5-dependent manner. In addition, colocalization of autophagosome and mitochondria, indicative of mitophagosome formation and mitophagy, was observed. Versatility of the drug regimen was confirmed by experiments in MV4-11 cells, a FLT3-ITD AML mutant. These results indicate that the C6-ceramide-tamoxifen regimen plays a pivotal role inducing autophagy in AML, and thus constitutes a novel therapeutic design.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Ceramides/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mitophagy/physiology , Tamoxifen/administration & dosage , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5/physiology , Cell Death/drug effects , Cell Survival/drug effects , Drug Synergism , Humans , Mitophagy/drug effects , Signal Transduction/drug effects , Tumor Cells, Cultured
11.
Biomed Pharmacother ; 96: 417-425, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29031200

ABSTRACT

Oleanolic acid derivatives exhibit potent anticancer activities against numerous types of cancer. However, the antitumor activity of oleanolic acid methylester (OAME), an oleanolic acid derivative, against prostate cancer has not been studied. Hence, the present work was conducted to study the anticancer activities of OAME. Viability assay showed that treatment of cancer cells with OAME induced a significant cell death in concentration- and time-dependent manner. Of note, OAME displayed a selective cytotoxicity against cancer cells compared to normal epithelial cells. Cells treated with OAME exhibited cell cycle arrest at both G1 and G2. Apoptotic induction potential of OAME was demonstrated using Annexin V assay, caspase activation, and DNA fragmentation methods Mechanistically, the results revealed that OAME strongly impacted the intrinsic apoptotic pathway in a concentration-dependent manner, as demonstrated by loss of mitochondrial membrane potential and release cytochrome c into the cytosol. ROS scavenger completely abrogated OAME-induced cell death. In vivo, OAME exerted concentration- dependent antiproliferative effect, associated with a significant level of apoptosis, potent antiangiogenic activity, and downregulation of survivin. This study provides significant insight into the therapeutic activities of OAME against prostate cancer in vitro and in vivo, suggesting that OAME might serve as a promising lead compound to treat hormonal-resistant prostate cancer.


Subject(s)
Cell Cycle Checkpoints/physiology , Cytotoxins/pharmacology , Oleanolic Acid/pharmacology , Prostatic Neoplasms, Castration-Resistant/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Chick Embryo , Cytotoxins/chemistry , Dose-Response Relationship, Drug , Humans , Male , Oleanolic Acid/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/pathology , Xenograft Model Antitumor Assays/methods
12.
Biochem Pharmacol ; 85(8): 1057-65, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23353700

ABSTRACT

Poor prognosis in patients with later stage colorectal cancer (CRC) necessitates the search for new treatment strategies. Ceramide, because of its role in orchestrating death cascades in cancer cells, is a versatile alternative. Ceramide can be generated by exposure to chemotherapy or ionizing radiation, or it can be administered in the form of short-chain analogs (C6-ceramide). Because intracellular P-glycoprotein (P-gp) plays a role in catalyzing the conversion of ceramide to higher sphingolipids, we hypothesized that administration of P-gp antagonists with C6-ceramide would magnify cell death cascades. Human CRC cell lines were employed, HCT-15, HT-29, and LoVo. The addition of either tamoxifen, VX-710, verapamil, or cyclosporin A, antagonists of P-gp, enhanced C6-ceramide cytotoxicity in all cell lines. In depth studies with C6-ceramide and tamoxifen in LoVo cells showed the regimen induced PARP cleavage, caspase-dependent apoptosis, mitochondrial membrane permeabilization (MMP), and cell cycle arrest at G1 and G2. At the molecular level, the regimen, but not single agents, induced time-dependent upregulation of tumor suppressor protein p53; however, introduction of a p53 inhibitor staved neither MMP nor apoptosis. Nanoliposomal formulations of C6-ceramide and tamoxifen were also effective, yielding synergistic cell kill. We conclude that tamoxifen is a favorable adjuvant for enhancing C6-ceramide cytotoxicity in CRC, and demonstrates uniquely integrated effects. The high frequency of expression of P-gp in CRC presents an adventitious target for complementing ceramide-based therapies, a strategy that could hold promise for treatment of resistant disease.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Ceramides/therapeutic use , Colorectal Neoplasms/drug therapy , Tumor Suppressor Protein p53/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Ceramides/metabolism , Drug Synergism , Humans , Piperidines/pharmacology , Pyridines/pharmacology , Tamoxifen/pharmacology
13.
Cancer Chemother Pharmacol ; 71(3): 635-45, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23263160

ABSTRACT

PURPOSE: Acid ceramidase (AC) occupies an important place in the control of cancer cell proliferation. We tested the influence of AC inhibition on the effects of PSC 833, a P-glycoprotein antagonist with potent ceramide-generating capacity, to determine whether AC could be a therapeutic target in pancreatic cancer. METHODS: Ceramide metabolism was followed using (3)H-palmitate, and molecular species were determined by mass spectroscopy. Apoptosis was measured by DNA fragmentation, autophagy by acridine orange staining, and cell cycle was assessed by flow cytometry and RB phosphorylation. AC was measured in intact cells using fluorescent substrate. RESULTS: Exposure of human PANC-1 or MIA-PaCa-2 cells to PSC 833 promoted increases in de novo (dihydro)ceramides, (dihydro)glucosylceramides, and (dihydro)sphingomyelins, demarking ceramide generation and robust metabolism. Despite the multifold increases in (dihydro)ceramide levels, cells were refractory to PSC 833. However, PSC 833 produced a dose-dependent decrease in DNA synthesis and dose- and time-dependent decreases in RB phosphorylation, consistent with cell cycle arrest as demonstrated at G1. Cytostatic effects of PSC 833 were converted to cytotoxic end-point by acid ceramidase inhibition. Cytotoxicity was accompanied by formation of acridine orange-stained acidic vesicles and an increase in LC3 expression, indicative of autophagic response. Cell death was not reversed by preexposure to myriocin, which blocks PSC 833-induced ceramide generation. CONCLUSION: Although the role of ceramide in end-point cytotoxicity is unclear, our results suggest that acid ceramidase is a viable target in pancreatic cancer. We propose that AC inhibition will be effective in concert with other anticancer therapies.


Subject(s)
Acid Ceramidase/antagonists & inhibitors , Acid Ceramidase/metabolism , Antineoplastic Agents/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Acridine Orange , Amides/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Chromatography, Thin Layer , Cyclosporins/pharmacology , DNA Fragmentation/drug effects , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Endpoint Determination , Fatty Acids, Unsaturated/pharmacology , Flow Cytometry , Humans , Mass Spectrometry , Sphingolipids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL