Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 22(2): 179-191, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36399631

ABSTRACT

The multi-receptor tyrosine kinase inhibitor XL092 has been developed to inhibit the activity of oncogenic targets, including MET, VEGFR2, and the TAM family of kinases TYRO3, AXL and MER. Presented here is a preclinical evaluation of XL092. XL092 causes a significant decrease in tumor MET and AXL phosphorylation (P < 0.01) in murine Hs 746T xenograft models relative to vehicle, and a 96% inhibition of VEGFR2 phosphorylation in murine lungs. Dose-dependent tumor growth inhibition with XL092 was observed in various murine xenograft models, with dose-dependent tumor regression seen in the NCI-H441 model. Tumor growth inhibition was enhanced with the combination of XL092 with anti-PD-1, anti-programmed death ligand-1 (PD-L1), or anti-CTLA-4 compared with any of these agents alone in the MC38 murine syngeneic model and with anti-PD-1 in the CT26 colorectal cancer survival model. In vivo, XL092 promoted a decrease in the tumor microvasculature and significant increases of peripheral CD4+ T cells and B cells and decreases in myeloid cells versus vehicle. Significant increases in CD8+ T cells were also observed with XL092 plus anti-PD-1 or anti-PD-L1 versus vehicle. In addition, XL092 promoted M2 to M1 repolarization of macrophages in vitro and inhibited primary human macrophage efferocytosis in a dose-dependent manner. In summary, XL092 was shown to have significant antitumor and immunomodulatory activity in animal models both alone and in combination with immune checkpoint inhibitors, supporting its evaluation in clinical trials.


Subject(s)
Neoplasms , Humans , Animals , Mice , Carrier Proteins , CD8-Positive T-Lymphocytes , Receptor Protein-Tyrosine Kinases , Disease Models, Animal , Cell Line, Tumor
2.
Mar Drugs ; 20(7)2022 Jun 27.
Article in English | MEDLINE | ID: mdl-35877711

ABSTRACT

The mandelalides are complex macrolactone natural products with distinct macrocycle motifs and a bioactivity profile that is heavily influenced by compound glycosylation. Mandelalides A and B are direct inhibitors of mitochondrial ATP synthase (complex V) and therefore more toxic to mammalian cells with an oxidative metabolic phenotype. To provide further insight into the pharmacology of the mandelalides, we studied the AMP-activated protein kinase (AMPK) energy stress pathway and report that mandelalide A is an indirect activator of AMPK. Wild-type mouse embryonic fibroblasts (MEFs) and representative human non-small cell lung cancer (NSCLC) cells showed statistically significant increases in phospho-AMPK (Thr172) and phospho-ACC (Ser79) in response to mandelalide A. Mandelalide L, which also harbors an A-type macrocycle, induced similar increases in phospho-AMPK (Thr172) and phospho-ACC (Ser79) in U87-MG glioblastoma cells. In contrast, MEFs co-treated with an AMPK inhibitor (dorsomorphin), AMPKα-null MEFs, or NSCLC cells lacking liver kinase B1 (LKB1) lacked this activity. Mandelalide A was significantly more cytotoxic to AMPKα-null MEFs than wild-type cells, suggesting that AMPK activation serves as a protective response to mandelalide-induced depletion of cellular ATP. However, LKB1 status alone was not predictive of the antiproliferative effects of mandelalide A against NSCLC cells. When EGFR status was considered, erlotinib and mandelalide A showed strong cytotoxic synergy in combination against erlotinib-resistant 11-18 NSCLC cells but not against erlotinib-sensitive PC-9 cells. Finally, prolonged exposures rendered mandelalide A, a potent and efficacious cytotoxin, against a panel of human glioblastoma cell types regardless of the underlying metabolic phenotype of the cell. These results add biological relevance to the mandelalide series and provide the basis for their further pre-clinical evaluation as ATP synthase inhibitors and secondary activators of AMPK.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Glioblastoma , Lung Neoplasms , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Erlotinib Hydrochloride , Fibroblasts/metabolism , Humans , Lung Neoplasms/drug therapy , Macrolides , Mammals/metabolism , Mice , Phosphorylation
3.
Cell Stress Chaperones ; 27(3): 223-239, 2022 05.
Article in English | MEDLINE | ID: mdl-35244890

ABSTRACT

Osteosarcoma is the most common type of bone cancer in dogs and humans, with significant numbers of patients experiencing treatment failure and disease progression. In our search for new approaches to treat osteosarcoma, we previously detected multiple chaperone proteins in the surface-exposed proteome of canine osteosarcoma cells. In the present study, we characterized expression of representative chaperones and find evidence for stress adaptation in canine osteosarcoma cells relative to osteogenic progenitors from normal bone. We compared the cytotoxic potential of direct (HA15) and putative (OSU-03012) inhibitors of Grp78 function and found canine POS and HMPOS osteosarcoma cells to be more sensitive to both compounds than normal cells. HA15 and OSU-03012 increased the thermal stability of Grp78 in intact POS cells at low micromolar concentrations, but each induced distinct patterns in Grp78 expression without significant change in Grp94. Both inhibitors were as effective alone as carboplatin and showed little evidence of synergy in combination treatment. However, HMPOS cells with acquired resistance to carboplatin were sensitive to inhibition of Grp78 (by HA15; OSU-03012), Hsp70 (by VER-155008), and Hsp90 (by 17-AAG) function. These results suggest that multiple nodes within the osteosarcoma chaperome may be relevant chemotherapeutic targets against platinum resistance.


Subject(s)
Bone Neoplasms , Osteosarcoma , Animals , Bone Neoplasms/drug therapy , Carboplatin , Cell Line, Tumor , Dogs , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/metabolism , Osteosarcoma/drug therapy
4.
Biochem Pharmacol ; 183: 114317, 2021 01.
Article in English | MEDLINE | ID: mdl-33152346

ABSTRACT

Coibamide A is a potent cancer cell toxin and one of a select group of natural products that inhibit protein entry into the secretory pathway via a direct inhibition of the Sec61 protein translocon. Many Sec61 client proteins are clinically relevant drug targets once trafficked to their final destination in or outside the cell, however the use of Sec61 inhibitors to block early biosynthesis of specific proteins is at a pre-clinical stage. In the present study we evaluated the action of coibamide A against human epidermal growth factor receptor (HER, ErbB) proteins in representative breast and lung cancer cell types. HERs were selected for this study as they represent a family of Sec61 clients that is frequently dysregulated in human cancers, including coibamide-sensitive cell types. Although coibamide A inhibits biogenesis of a broad range of Sec61 substrate proteins in a presumed substrate-nonselective manner, endogenous HER3 (ErbB-3) and EGFR (ErbB-1) proteins were more sensitive to coibamide A, and the related Sec61 inhibitor apratoxin A, than HER2 (ErbB-2). Despite this rank order of sensitivity (HER3 > EGFR > HER2), Sec61-dependent inhibition by coibamide A was sufficient to decrease cell surface expression of HER2. We report that coibamide A- or apratoxin A-mediated block of HER3 entry into the secretory pathway is unlikely to be mediated by the HER3 signal peptide alone. HER3 (G11L/S15L), that is fully resistant to the highly substrate-selective cotransin analogue CT8, was more resistant than wild-type HER3 but only at low coibamide A (3 nM) concentrations; HER3 (G11L/S15L) expression was inhibited by higher concentrations of either natural product. Time- and concentration-dependent decreases in HER protein expression induced a commensurate reduction in AKT/MAPK signaling in breast and lung cancer cell types and loss in cell viability. Coibamide A potentiated the cytotoxic efficacy of small molecule kinase inhibitors lapatinib and erlotinib in breast and lung cancer cell types, respectively. These data indicate that natural product modulators of Sec61 function have value as chemical probes to interrogate HER/ErbB signaling in treatment-resistant human cancers.


Subject(s)
Depsipeptides/administration & dosage , Drug Delivery Systems/methods , SEC Translocation Channels/antagonists & inhibitors , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , HEK293 Cells , Humans , MCF-7 Cells , SEC Translocation Channels/metabolism
5.
ACS Chem Biol ; 15(8): 2125-2136, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32608972

ABSTRACT

Coibamide A (CbA) is a marine natural product with potent antiproliferative activity against human cancer cells and a unique selectivity profile. Despite promising antitumor activity, the mechanism of cytotoxicity and specific cellular target of CbA remain unknown. Here, we develop an optimized synthetic CbA photoaffinity probe (photo-CbA) and use it to demonstrate that CbA directly targets the Sec61α subunit of the Sec61 protein translocon. CbA binding to Sec61 results in broad substrate-nonselective inhibition of ER protein import and potent cytotoxicity against specific cancer cell lines. CbA targets a lumenal cavity of Sec61 that is partially shared with known Sec61 inhibitors, yet profiling against resistance conferring Sec61α mutations identified from human HCT116 cells suggests a distinct binding mode for CbA. Specifically, despite conferring strong resistance to all previously known Sec61 inhibitors, the Sec61α mutant R66I remains sensitive to CbA. A further unbiased screen for Sec61α resistance mutations identified the CbA-resistant mutation S71P, which confirms nonidentical binding sites for CbA and apratoxin A and supports the susceptibility of the Sec61 plug region for channel inhibition. Remarkably, CbA, apratoxin A, and ipomoeassin F do not display comparable patterns of potency and selectivity in the NCI60 panel of human cancer cell lines. Our work connecting CbA activity with selective prevention of secretory and membrane protein biogenesis by inhibition of Sec61 opens up possibilities for developing new Sec61 inhibitors with improved drug-like properties that are based on the coibamide pharmacophore.


Subject(s)
Depsipeptides/pharmacology , Membrane Proteins/antagonists & inhibitors , SEC Translocation Channels/drug effects , Binding Sites , Cells, Cultured , Depsipeptides/metabolism , Humans , Membrane Proteins/biosynthesis , Photoaffinity Labels/chemistry , SEC Translocation Channels/metabolism
6.
Methods Mol Biol ; 2155: 193-200, 2020.
Article in English | MEDLINE | ID: mdl-32474878

ABSTRACT

The pancreas is composed of different cellular populations, organized into distinct functional units, including acinar clusters, islets of Langerhans, and the ductal system. As a result of research into diabetes, several optical techniques have been developed for the three-dimensional visualization of islet populations, so as to better understand their anatomical characteristics. These approaches are largely reliant on three-dimensional whole-mount immunofluorescence staining. In this chapter, we review a revised whole mount immunofluorescence staining method for studying adult pancreatic islet morphology. This method uses smaller samples and combines the blocking and permeabilization steps. This reduces the time needed, relative to existing protocols; the method is compatible with regular confocal microscopy as well.


Subject(s)
Fluorescent Antibody Technique , Microscopy, Confocal , Pancreas/cytology , Pancreas/metabolism , Animals , Dissection , Fluorescent Antibody Technique/methods , Imaging, Three-Dimensional , Mice , Microscopy, Confocal/methods
7.
Sci Rep ; 8(1): 13451, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30194315

ABSTRACT

Cadherin-mediated cell-cell adhesion plays an important role in organ development and changes in cadherin expression are often linked to morphogenetic and pathogenic events. Cadherins interact with other intracellular components to form adherens junctions (AJs) and provide mechanical attachments between adjacent cells. E-cadherin (Cdh1) represents an integral component of these intercellular junctions. To elucidate the function of E-cadherin in the developing pancreas, we generated and studied pancreas-specific Cdh1-knockout (Cdh1ΔPan/ΔPan) mice. Cdh1ΔPan/ΔPan mice exhibit normal body size at birth, but fail to gain weight and become hypoglycemic soon afterward. We found that E-cadherin is not required for the establishment of apical-basal polarity or pancreatic exocrine cell identity at birth. However, four days after birth, the pancreata of Cdh1ΔPan/ΔPan mutants display progressive deterioration of exocrine architecture and dysregulation of Wnt and YAP signaling. At this time point, the acinar cells of Cdh1ΔPan/ΔPan mutants begin to exhibit ductal phenotypes, suggesting acinar-to-ductal metaplasia (ADM) in the E-cadherin-deficient pancreas. Our findings demonstrate that E-cadherin plays an integral role in the maintenance of exocrine architecture and regulation of homeostatic signaling. The present study provides insights into the involvement of cadherin-mediated cell-cell adhesion in pathogenic conditions such as pancreatitis or pancreatic cancer.


Subject(s)
Cadherins/metabolism , Pancreas, Exocrine/growth & development , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cadherins/genetics , Cell Adhesion/genetics , Cell Cycle Proteins , Mice , Mice, Knockout , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatitis/genetics , Pancreatitis/metabolism , Pancreatitis/pathology , Phosphoproteins/genetics , Phosphoproteins/metabolism , YAP-Signaling Proteins
8.
Mar Drugs ; 16(3)2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29494533

ABSTRACT

Our understanding of autophagy and lysosomal function has been greatly enhanced by the discovery of natural product structures that can serve as chemical probes to reveal new patterns of signal transduction in cells. Coibamide A is a cytotoxic marine natural product that induces mTOR-independent autophagy as an adaptive stress response that precedes cell death. Autophagy-related (ATG) protein 5 (ATG5) is required for coibamide-induced autophagy but not required for coibamide-induced apoptosis. Using wild-type and autophagy-deficient mouse embryonic fibroblasts (MEFs) we demonstrate that coibamide-induced toxicity is delayed in ATG5-/- cells relative to ATG5+/+ cells. Time-dependent changes in annexin V staining, membrane integrity, metabolic capacity and caspase activation indicated that MEFs with a functional autophagy pathway are more sensitive to coibamide A. This pattern could be distinguished from autophagy modulators that induce acute ER stress (thapsigargin, tunicamycin), ATP depletion (oligomycin A) or mTORC1 inhibition (rapamycin), but was shared with the Sec61 inhibitor apratoxin A. Coibamide- or apratoxin-induced cell stress was further distinguished from the action of thapsigargin by a pattern of early LC3-II accumulation in the absence of CHOP or BiP expression. Time-dependent changes in ATG5-ATG12, PARP1 and caspase-3 expression patterns were consistent with the conversion of ATG5 to a pro-death signal in response to both compounds.


Subject(s)
Apoptosis/drug effects , Autophagy-Related Protein 5/metabolism , Autophagy/drug effects , Depsipeptides/toxicity , Animals , Autophagy-Related Protein 5/genetics , Cell Line , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Fibroblasts , Gene Knockout Techniques , Marine Toxins/toxicity , Mice , Signal Transduction/drug effects , Thapsigargin/toxicity
9.
J Med Chem ; 60(18): 7850-7862, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28841379

ABSTRACT

Mandelalides A-D (1-4) are macrocyclic polyketides known to have an unusual bioactivity profile influenced by compound glycosylation and growth phase of cultured cells. The isolation and characterization of additional natural congeners, mandelalides E-L (5-12), and the supply of synthetic compounds 1 and 12, as well as seco-mandelalide A methyl ester (13), have now facilitated mechanism of action and structure-activity relationship studies. Glycosylated mandelalides are effective inhibitors of aerobic respiration in living cells. Macrolides 1 and 2 inhibit mitochondrial function similar to oligomycin A and apoptolidin A, selective inhibitors of the mammalian ATP synthase (complex V). 1 inhibits ATP synthase activity from isolated mitochondria and triggers caspase-dependent apoptosis in HeLa cells, which are more sensitive to inhibition by 1 in the presence of the glycolysis inhibitor 2-deoxyglucose. Thus, mandelalide cytotoxicity depends on basal metabolic phenotype; cells with an oxidative phenotype are most likely to be inhibited by the mandelalides.


Subject(s)
Apoptosis/drug effects , Macrolides/chemistry , Macrolides/pharmacology , Mitochondria/drug effects , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Cell Survival/drug effects , Glycosylation , HEK293 Cells , HeLa Cells , Humans , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism
10.
Chembiochem ; 17(17): 1585-8, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27305101

ABSTRACT

Pactamycin is a bacteria-derived aminocyclitol antibiotic with a wide-range of biological activity. Its chemical structure and potent biological activities have made it an interesting lead compound for drug discovery and development. Despite its unusual chemical structure, many aspects of its formation in nature remain elusive. Using a combination of genetic inactivation and metabolic analysis, we investigated the tailoring processes of pactamycin biosynthesis in Streptomyces pactum. The results provide insights into the sequence of events during the tailoring steps of pactamycin biosynthesis and explain the unusual production of various pactamycin analogues by S. pactum mutants. We also identified two new pactamycin analogues that have better selectivity indexes than pactamycin against malarial parasites.


Subject(s)
Antibiotics, Antineoplastic/biosynthesis , Pactamycin/analogs & derivatives , Pactamycin/biosynthesis , Streptomyces/metabolism , Antibiotics, Antineoplastic/chemistry , Molecular Conformation , Pactamycin/chemistry , Streptomyces/genetics
11.
Org Lett ; 18(6): 1374-7, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26914981

ABSTRACT

Recollection of the tunicate source of the mandelalides has provided new and known analogues that have facilitated expanded analyses of the disputed cancer cytotoxicity of mandelalide A following a number of recent reported total syntheses. Using newly characterized mandelalide E, reisolated natural mandelalides B and C, and synthetic mandelalide A, the cytotoxicity of the mandelalides is demonstrated to be strongly influenced by compound glycosylation and assay cell density. Glycosylated mandelalides reduced the viability of human cancer cells cultured at a high starting density with a rank order of potency A > B ≫ E, yet display dramatically reduced cytotoxic efficacy against low density cultures.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Macrolides/chemical synthesis , Macrolides/pharmacology , Animals , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Glycosylation , HCT116 Cells , HeLa Cells , Humans , Macrolides/chemistry , Molecular Structure , Neoplasms , Stereoisomerism , Urochordata/chemistry
12.
J Am Chem Soc ; 138(3): 770-3, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26759923

ABSTRACT

The total synthesis of mandelalide A and its ring-expanded macrolide isomer isomandelalide A has been achieved. Unexpected high levels of cytotoxicity were observed with the ring-expanded isomandelalide A with a rank order of potency: mandelalide A > isomandelalide A > mandelalide B. Key aspects of the synthesis include Ag-catalyzed cyclizations (AgCC's) to construct both the THF and THP rings present in the macrocycle, diastereoselective Sharpless dihydroylation of a cis-enyne, and lithium acetylide coupling with a chiral epoxide.


Subject(s)
Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/pharmacology , Drug Discovery , Macrolides/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Catalysis , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclization , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Macrolides/chemistry , Macrolides/pharmacology , Molecular Conformation , Silver/chemistry , Stereoisomerism
13.
Invest New Drugs ; 34(1): 24-40, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26563191

ABSTRACT

Coibamide A is a cytotoxic lariat depsipeptide isolated from a rare cyanobacterium found within the marine reserve of Coiba National Park, Panama. Earlier testing of coibamide A in the National Cancer Institute in vitro 60 human tumor cell line panel (NCI-60) revealed potent anti-proliferative activity and a unique selectivity profile, potentially reflecting a new target or mechanism of action. In the present study we evaluated the antitumor activity of coibamide A in several functional cell-based assays and in vivo. U87-MG and SF-295 glioblastoma cells showed reduced migratory and invasive capacity and underwent G1 cell cycle arrest as, likely indirect, consequences of treatment. Coibamide A inhibited extracellular VEGFA secreted from U87-MG glioblastoma and MDA-MB-231 breast cancer cells with low nM potency, attenuated proliferation and migration of normal human umbilical vein endothelial cells (HUVECs) and selectively decreased expression of vascular endothelial growth factor receptor 2 (VEGFR2). We report that coibamide A retains potent antitumor properties in a nude mouse xenograft model of glioblastoma; established subcutaneous U87-MG tumors failed to grow for up to 28 days in response to 0.3 mg/Kg doses of coibamide A. However, the natural product was also associated with varied patterns of weight loss and thus targeted delivery and/or medicinal chemistry approaches will almost certainly be required to improve the toxicity profile of this unusual macrocycle. Finally, similarities between coibamide A- and apratoxin A-induced changes in cell morphology, decreases in VEGFR2 expression and macroautophagy signaling in HUVECs raise the possibility that both cyanobacterial natural products share a common mechanism of action.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Depsipeptides/pharmacology , Glioblastoma/drug therapy , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Glioblastoma/pathology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Nude , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Xenograft Model Antitumor Assays
14.
Org Lett ; 17(10): 2526-9, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25945812

ABSTRACT

Two new apoptolidins, 2'-O-succinyl-apoptolidin A (11) and 3'-O-succinyl-apoptolidin A (12), were isolated from the culture broth of an Indonesian Amycolatopsis sp. ICBB 8242. These compounds inhibit the proliferation and viability of human H292 and HeLa cells. However, in contrast to apoptolidin A (1), they do not inhibit cellular respiration in H292 cells. It is proposed that apoptolidins are produced and secreted in their succinylated forms and 1 is the hydrolysis product of 11 and 12.


Subject(s)
Actinomycetales/chemistry , Macrolides/chemical synthesis , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Indonesia , Macrolides/chemistry , Molecular Structure
15.
J Nat Prod ; 78(3): 413-20, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25562664

ABSTRACT

Two new cyclic depsipeptides, companeramides A (1) and B (2), have been isolated from the phylogenetically characterized cyanobacterial collection that yielded the previously reported cancer cell toxin coibamide A (collected from Coiba Island, Panama). The planar structures of the companeramides, which contain 3-amino-2-methyl-7-octynoic acid (Amoya), hydroxy isovaleric acid (Hiva), and eight α-amino acid units, were established by NMR spectroscopy and mass spectrometry. The absolute configuration of each companeramide was assigned using a combination of Marfey's methodology and chiral-phase HPLC analysis of complete and partial hydrolysis products compared to commercial and synthesized standards. Companeramides A (1) and B (2) showed high nanomolar in vitro antiplasmodial activity but were not overtly cytotoxic to four human cancer cell lines at the doses tested.


Subject(s)
Antineoplastic Agents/isolation & purification , Cyanobacteria/chemistry , Depsipeptides/isolation & purification , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Chromatography, High Pressure Liquid , Depsipeptides/chemistry , Depsipeptides/pharmacology , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Panama
16.
Biochem Pharmacol ; 93(3): 251-65, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25511868

ABSTRACT

Apoptolidin A was first isolated as a secondary metabolite of a Nocardiopsis sp. and is the founding member of a family of potential selective cancer cell toxins. We now report the isolation, production and pharmacological characterization of apoptolidins A and C from an alternate actinomycete producer, an Amycolatopsis sp. from soil samples collected in Indonesia. We investigated the action of apoptolidins A and C in representative human glioblastoma cells, lung cancer cells and mouse embryonic fibroblasts (MEFs) to better understand the mechanism of action of the known apoptolidins. Shifts in cellular metabolism in intact cells and the status of the AMP-activated protein kinase (AMPK) stress pathway in response to apoptolidin A were entirely consistent with the actions of an ATP synthase inhibitor. We find the metabolic phenotype of the cell to be a critical determinant of apoptolidin sensitivity and the likely basis for cancer cell selectivity. The apoptolidins induce indirect activation of AMPK and trigger autophagy in sensitive cell types without significant inhibition of mTORC1. Human U87-MG glioblastoma cells and wild type MEFs showed increased phosphorylation of AMPK (Thr172), ACC (Ser79) and ULK1 (Ser555), whereas AMPKα-null MEFs and more glycolytic SF-295 glioblastoma cells lacked this response. Although both are reported to be selective inhibitors of mitochondrial ATP synthase, differences between apoptolidin- and oligomycin A-induced responses in cells indicate that the action of these macrolides is not identical.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Survival/physiology , Macrolides/pharmacology , Oligomycins/pharmacology , Pyrones/pharmacology , Soil Microbiology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Humans , Macrolides/isolation & purification , Mice , Mice, Knockout , Oligomycins/isolation & purification , Pyrones/isolation & purification
17.
PLoS One ; 8(6): e65250, 2013.
Article in English | MEDLINE | ID: mdl-23762328

ABSTRACT

Coibamide A is an N-methyl-stabilized depsipeptide that was isolated from a marine cyanobacterium as part of an International Cooperative Biodiversity Groups (ICBG) program based in Panama. Previous testing of coibamide A in the NCI in vitro 60 cancer cell line panel revealed a potent anti-proliferative response and "COMPARE-negative" profile indicative of a unique mechanism of action. We report that coibamide A is a more potent and efficacious cytotoxin than was previously appreciated, inducing concentration- and time-dependent cytotoxicity (EC50<100 nM) in human U87-MG and SF-295 glioblastoma cells and mouse embryonic fibroblasts (MEFs). This activity was lost upon linearization of the molecule, highlighting the importance of the cyclized structure for both anti-proliferative and cytotoxic responses. We show that coibamide A induces autophagosome accumulation in human glioblastoma cell types and MEFs via an mTOR-independent mechanism; no change was observed in the phosphorylation state of ULK1 (Ser-757), p70 S6K1 (Thr-389), S6 ribosomal protein (Ser-235/236) and 4EBP-1 (Thr-37/46). Coibamide A also induces morphologically and biochemically distinct forms of cell death according to cell type. SF-295 glioblastoma cells showed caspase-3 activation and evidence of apoptotic cell death in a pattern that was also seen in wild-type and autophagy-deficient (ATG5-null) MEFs. In contrast, cell death in U87-MG glioblastoma cells was characterized by extensive cytoplasmic vacuolization and lacked clear apoptotic features. Cell death was attenuated, but still triggered, in Apaf-1-null MEFs lacking a functional mitochondria-mediated apoptotic pathway. From the study of ATG5-null MEFs we conclude that a conventional autophagy response is not required for coibamide A-induced cell death, but likely occurs in dying cells in response to treatment. Coibamide A represents a natural product scaffold with potential for the study of mTOR-independent signaling and cell death mechanisms in apoptotic-resistant cancer cells.


Subject(s)
Autophagy/drug effects , Cytotoxins/pharmacology , Depsipeptides/pharmacology , Fibroblasts/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/drug effects , Apoptotic Protease-Activating Factor 1/deficiency , Apoptotic Protease-Activating Factor 1/genetics , Autophagy-Related Protein 5 , Autophagy-Related Protein-1 Homolog , Caspase 3/genetics , Caspase 3/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Organ Specificity , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL