251
|
Cotter K, Capecci J, Sennoune S, Huss M, Maier M, Martinez-Zaguilan R, Forgac M. Activity of plasma membrane V-ATPases is critical for the invasion of MDA-MB231 breast cancer cells. J Biol Chem 2014; 290:3680-92. [PMID: 25505184 DOI: 10.1074/jbc.m114.611210] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The vacuolar (H(+))-ATPases (V-ATPases) are a family of ATP-driven proton pumps that couple ATP hydrolysis with translocation of protons across membranes. Previous studies have implicated V-ATPases in cancer cell invasion. It has been proposed that V-ATPases participate in invasion by localizing to the plasma membrane and causing acidification of the extracellular space. To test this hypothesis, we utilized two separate approaches to specifically inhibit plasma membrane V-ATPases. First, we stably transfected highly invasive MDA-MB231 cells with a V5-tagged construct of the membrane-embedded c subunit of the V-ATPase, allowing for extracellular expression of the V5 epitope. We evaluated the effect of addition of a monoclonal antibody directed against the V5 epitope on both V-ATPase-mediated proton translocation across the plasma membrane and invasion using an in vitro Matrigel assay. The addition of anti-V5 antibody resulted in acidification of the cytosol and a decrease in V-ATPase-dependent proton flux across the plasma membrane in transfected but not control (untransfected) cells. These results demonstrate that the anti-V5 antibody inhibits activity of plasma membrane V-ATPases in transfected cells. Addition of the anti-V5 antibody also inhibited in vitro invasion of transfected (but not untransfected) cells. Second, we utilized a biotin-conjugated form of the specific V-ATPase inhibitor bafilomycin. When bound to streptavidin, this compound cannot cross the plasma membrane. Addition of this compound to MDA-MB231 cells also inhibited in vitro invasion. These studies suggest that plasma membrane V-ATPases play an important role in invasion of breast cancer cells.
Collapse
Affiliation(s)
- Kristina Cotter
- From the Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Joseph Capecci
- From the Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Souad Sennoune
- the Department of Cell Physiology and Molecular Biophysics, Texas Tech University, Lubbock, Texas 79430
| | - Markus Huss
- the Department of Biology/Chemistry, Division of Animal Physiology, University of Osnabrück, 49069 Osnabrück, Germany, and
| | - Martin Maier
- the Institute of Organic Chemistry, University of Tuebingen, 72076 Tuebingen, Germany
| | - Raul Martinez-Zaguilan
- the Department of Cell Physiology and Molecular Biophysics, Texas Tech University, Lubbock, Texas 79430
| | - Michael Forgac
- From the Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111,
| |
Collapse
|
252
|
Gunawan ST, Liang K, Such GK, Johnston APR, Leung MKM, Cui J, Caruso F. Engineering enzyme-cleavable hybrid click capsules with a pH-sheddable coating for intracellular degradation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:4080-4086. [PMID: 25044500 DOI: 10.1002/smll.201400450] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Indexed: 06/03/2023]
Abstract
The engineering of layer-by-layer (LbL) hybrid click capsules that are responsive to biological stimuli is reported. The capsules comprise a pH-sheddable, non cross-linked outer coating that protects enzyme-cleavable inner layers. Upon cellular uptake, the outer coating is released and the capsules are enzymatically degraded. In vitro cell degradation results in rapid capsule degradation (10 min) upon cellular internalization.
Collapse
Affiliation(s)
- Sylvia T Gunawan
- Department of Chemical and Biomolecular Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | | | | | | | | | | |
Collapse
|
253
|
de la Torre C, Mondragón L, Coll C, Sancenón F, Marcos MD, Martínez-Máñez R, Amorós P, Pérez-Payá E, Orzáez M. Cathepsin-B Induced Controlled Release from Peptide-Capped Mesoporous Silica Nanoparticles. Chemistry 2014; 20:15309-14. [DOI: 10.1002/chem.201404382] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Indexed: 01/22/2023]
|
254
|
Fujii T, Kamiya M, Urano Y. In Vivo Imaging of Intraperitoneally Disseminated Tumors in Model Mice by Using Activatable Fluorescent Small-Molecular Probes for Activity of Cathepsins. Bioconjug Chem 2014; 25:1838-46. [DOI: 10.1021/bc5003289] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | - Yasuteru Urano
- Basic
Research Program, Japan Science and Technology Agency, K’s Gobancho, 7, Gobancho,
Chiyoda-ku, Tokyo 102-0076, Japan
| |
Collapse
|
255
|
Sun S, Kennedy RT. Droplet electrospray ionization mass spectrometry for high throughput screening for enzyme inhibitors. Anal Chem 2014; 86:9309-14. [PMID: 25137241 PMCID: PMC4165461 DOI: 10.1021/ac502542z] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 08/19/2014] [Indexed: 12/20/2022]
Abstract
High throughput screening (HTS) is important for identifying molecules with desired properties. Mass spectrometry (MS) is potentially powerful for label-free HTS due to its high sensitivity, speed, and resolution. Segmented flow, where samples are manipulated as droplets separated by an immiscible fluid, is an intriguing format for high throughput MS because it can be used to reliably and precisely manipulate nanoliter volumes and can be directly coupled to electrospray ionization (ESI) MS for rapid analysis. In this study, we describe a "MS Plate Reader" that couples standard multiwell plate HTS workflow to droplet ESI-MS. The MS plate reader can reformat 3072 samples from eight 384-well plates into nanoliter droplets segmented by an immiscible oil at 4.5 samples/s and sequentially analyze them by MS at 2 samples/s. Using the system, a label-free screen for cathepsin B modulators against 1280 chemicals was completed in 45 min with a high Z-factor (>0.72) and no false positives (24 of 24 hits confirmed). The assay revealed 11 structures not previously linked to cathepsin inhibition. For even larger scale screening, reformatting and analysis could be conducted simultaneously, which would enable more than 145,000 samples to be analyzed in 1 day.
Collapse
Affiliation(s)
- Shuwen Sun
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Robert T. Kennedy
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
256
|
Gurnev PA, Nestorovich EM. Channel-forming bacterial toxins in biosensing and macromolecule delivery. Toxins (Basel) 2014; 6:2483-540. [PMID: 25153255 PMCID: PMC4147595 DOI: 10.3390/toxins6082483] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/08/2014] [Accepted: 08/08/2014] [Indexed: 12/19/2022] Open
Abstract
To intoxicate cells, pore-forming bacterial toxins are evolved to allow for the transmembrane traffic of different substrates, ranging from small inorganic ions to cell-specific polypeptides. Recent developments in single-channel electrical recordings, X-ray crystallography, protein engineering, and computational methods have generated a large body of knowledge about the basic principles of channel-mediated molecular transport. These discoveries provide a robust framework for expansion of the described principles and methods toward use of biological nanopores in the growing field of nanobiotechnology. This article, written for a special volume on "Intracellular Traffic and Transport of Bacterial Protein Toxins", reviews the current state of applications of pore-forming bacterial toxins in small- and macromolecule-sensing, targeted cancer therapy, and drug delivery. We discuss the electrophysiological studies that explore molecular details of channel-facilitated protein and polymer transport across cellular membranes using both natural and foreign substrates. The review focuses on the structurally and functionally different bacterial toxins: gramicidin A of Bacillus brevis, α-hemolysin of Staphylococcus aureus, and binary toxin of Bacillus anthracis, which have found their "second life" in a variety of developing medical and technological applications.
Collapse
Affiliation(s)
- Philip A Gurnev
- Physics Department, University of Massachusetts, Amherst, MA 01003, USA.
| | | |
Collapse
|
257
|
Sun Y, Takada K, Nogi Y, Okada S, Matsunaga S. Lower homologues of ahpatinin, aspartic protease inhibitors, from a marine Streptomyces sp. JOURNAL OF NATURAL PRODUCTS 2014; 77:1749-1752. [PMID: 24960234 DOI: 10.1021/np500337m] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Two linear peptides, ahpatinin Ac (1) and ahpatinin Pr (2), were isolated together with the known ahpatinin (i)Bu, pepstatin Ac, pepstatin Pr, and pepsinostreptin from a Streptomyces sp. derived from a deep-sea sediment. The structure of ahpatinin Pr (2) was assigned by interpretation of NMR data and HPLC analysis of the hydrolysate after converting to the DNP-L-Val derivative. During the LCMS analysis of the acid hydrolysate, products arising from the retro-aldol cleavage of the statine and Ahppa units in 2 were observed and could facilitate the determination of the absolute configuration of the statine class of nonproteinogenic amino acids. Both ahpatinin Ac (1) and ahpatinin Pr (2) potently inhibited pepsin and moderately inhibited cathepsin B.
Collapse
Affiliation(s)
- Yi Sun
- Laboratory of Aquatic Natural Products Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo , Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | |
Collapse
|
258
|
Aggarwal N, Sloane BF. Cathepsin B: multiple roles in cancer. Proteomics Clin Appl 2014; 8:427-37. [PMID: 24677670 PMCID: PMC4205946 DOI: 10.1002/prca.201300105] [Citation(s) in RCA: 265] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/05/2013] [Accepted: 12/19/2013] [Indexed: 12/21/2022]
Abstract
Proteases, including intracellular proteases, play roles at many different stages of malignant progression. Our focus here is cathepsin B, a lysosomal cysteine cathepsin. High levels of cathepsin B are found in a wide variety of human cancers, levels that often induce secretion and association of cathepsin B with the tumor cell membrane. In experimental models, such as transgenic models of murine pancreatic and mammary carcinomas, causal roles for cathepsin B have been demonstrated in initiation, growth/tumor cell proliferation, angiogenesis, invasion, and metastasis. Tumor growth in transgenic models is promoted by cathepsin B in tumor-associated cells, for example, tumor-associated macrophages, as well as in tumor cells. In transgenic models, the absence of cathepsin B has been associated with enhanced apoptosis, yet cathepsin B also has been shown to contribute to apoptosis. Cathepsin B is part of a proteolytic pathway identified in xenograft models of human glioma; targeting only cathepsin B in these tumors is less effective than targeting cathepsin B in combination with other proteases or protease receptors. Understanding the mechanisms responsible for increased expression of cathepsin B in tumors and association of cathepsin B with tumor cell membranes is needed to determine whether targeting cathepsin B could be of therapeutic benefit.
Collapse
Affiliation(s)
- Neha Aggarwal
- Department of Physiology, Wayne State University School of Medicine, Detroit, Ml, USA
| | - Bonnie F. Sloane
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Ml, USA
| |
Collapse
|
259
|
Molecular Cloning and Characterization of Cystatin, a Cysteine Protease Inhibitor, fromBufo melanostictus. Biosci Biotechnol Biochem 2014; 77:2077-81. [DOI: 10.1271/bbb.130424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
260
|
Satsangi A, Roy SS, Satsangi RK, Vadlamudi RK, Ong JL. Design of a paclitaxel prodrug conjugate for active targeting of an enzyme upregulated in breast cancer cells. Mol Pharm 2014; 11:1906-18. [PMID: 24847940 DOI: 10.1021/mp500128k] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Breast cancer is the second most common cause of cancer-related deaths in women. Chemotherapy is an important treatment modality, and paclitaxel (PTX) is often the first-line therapy for its metastatic form. The two most notable limitations related to PTX-based treatment are the poor hydrophilicity of the drug and the systemic toxicity due to the drug's nonspecific and indiscriminate distribution among the tissues. The present work describes an approach to counter both challenges by designing a conjugate of PTX with a hydrophilic macromolecule that is coupled through a biocleavable linker, thereby allowing for active targeting to an enzyme significantly upregulated in cancer cells. The resultant strategy would allow for the release of the active ingredient preferentially at the site of action in related cancer cells and spare normal tissue. Thus, PTX was conjugated to the hydrophilic poly(amdioamine) [PAMAM] dendrimer through the cathepsin B-cleavable tetrapeptide Gly-Phe-Leu-Gly. The PTX prodrug conjugate (PGD) was compared to unbound PTX through in vitro evaluations against breast cancer cells and normal kidney cells as well as through in vivo evaluations using xenograft mice models. As compared to PTX, PGD demonstrated a higher cytotoxicity specific to cell lines with moderate-to-high cathepsin B activity; cells with comparatively lower cathepsin B activity demonstrated an inverse of this relationship. Regression analysis between the magnitude of PGD-induced cytotoxic increase over PTX and cathepsin B expression showed a strong, statistically significant correlation (r(2) = 0.652, p < 0.05). The PGD conjugate also demonstrated a markedly higher tumor reduction as compared to PTX treatment alone in MDA-MB-231 tumor xenograft models, with PGD-treated tumor volumes being 48% and 34% smaller than PTX-treated volumes at weeks 2 and 3 after treatment initiation.
Collapse
Affiliation(s)
- Arpan Satsangi
- Joint Graduate Program in Biomedical Engineering, University of Texas at San Antonio and the University of Texas Health Science Center at San Antonio , San Antonio, Texas 78249, United States
| | | | | | | | | |
Collapse
|
261
|
Pader I, Sengupta R, Cebula M, Xu J, Lundberg JO, Holmgren A, Johansson K, Arnér ESJ. Thioredoxin-related protein of 14 kDa is an efficient L-cystine reductase and S-denitrosylase. Proc Natl Acad Sci U S A 2014; 111:6964-9. [PMID: 24778250 PMCID: PMC4024855 DOI: 10.1073/pnas.1317320111] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Thioredoxin-related protein of 14 kDa (TRP14, also called TXNDC17 for thioredoxin domain containing 17, or TXNL5 for thioredoxin-like 5) is an evolutionarily well-conserved member of the thioredoxin (Trx)-fold protein family that lacks activity with classical Trx1 substrates. However, we discovered here that human TRP14 has a high enzymatic activity in reduction of l-cystine, where the catalytic efficiency (2,217 min(-1)⋅µM(-1)) coupled to Trx reductase 1 (TrxR1) using NADPH was fivefold higher compared with Trx1 (418 min(-1)⋅µM(-1)). Moreover, the l-cystine reduction with TRP14 was in contrast to that of Trx1 fully maintained in the presence of a protein disulfide substrate of Trx1 such as insulin, suggesting that TRP14 is a more dedicated l-cystine reductase compared with Trx1. We also found that TRP14 is an efficient S-denitrosylase with similar efficiency as Trx1 in catalyzing TrxR1-dependent denitrosylation of S-nitrosylated glutathione or of HEK293 cell-derived S-nitrosoproteins. Consequently, nitrosylated and thereby inactivated caspase 3 or cathepsin B could be reactivated through either Trx1- or TRP14-catalyzed denitrosylation reactions. TRP14 was also, in contrast to Trx1, completely resistant to inactivation by high concentrations of hydrogen peroxide. The oxidoreductase activities of TRP14 thereby complement those of Trx1 and must therefore be considered for the full understanding of enzymatic control of cellular thiols and nitrosothiols.
Collapse
Affiliation(s)
- Irina Pader
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics and
| | - Rajib Sengupta
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics and
| | - Marcus Cebula
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics and
| | - Jianqiang Xu
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics and
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Arne Holmgren
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics and
| | - Katarina Johansson
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics and
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics and
| |
Collapse
|
262
|
Gyparaki MT, Papavassiliou AG. Lysosome: the cell's ‘suicidal bag’ as a promising cancer target. Trends Mol Med 2014; 20:239-41. [DOI: 10.1016/j.molmed.2014.01.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 12/25/2022]
|
263
|
Lao Y, Wan G, Liu Z, Wang X, Ruan P, Xu W, Xu D, Xie W, Zhang Y, Xu H, Xu N. The natural compound oblongifolin C inhibits autophagic flux and enhances antitumor efficacy of nutrient deprivation. Autophagy 2014; 10:736-49. [PMID: 24642486 PMCID: PMC5119057 DOI: 10.4161/auto.28034] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Metabolic stress induces autophagy as an alternative source of energy and metabolites. Insufficient autophagy in nutrient-deprived cancer cells would be beneficial for cancer therapy. Here, we performed a functional screen in search of novel autophagy regulators from natural products. We showed that oblongifolin C (OC), a natural small molecule compound extracted from Garcinia yunnanensis Hu, is a potent autophagic flux inhibitor. Exposure to OC results in an increased number of autophagosomes and impaired degradation of SQSTM1/p62. Costaining of GFP-LC3B with LysoTracker Red or LAMP1 antibody demonstrates that autophagosome-lysosome fusion is blocked by OC treatment. Furthermore, OC inhibits lysosomal proteolytic activity by altering lysosomal acidification and downregulating the expression of lysosomal cathepsins. Importantly, OC can eliminate the tolerance of cancer cells to nutrient starvation. Starvation dramatically increases the susceptibility of cancer cells to OC-induced CASP3-dependent apoptosis in vitro. Subsequent studies in xenograft mouse model showed that OC has anticancer potency as revealed by increased staining of cleaved CASP3, LC3 puncta, and SQSTM1, as well as reduced expression of lysosomal cathepsins. Combined treatment with OC and caloric restriction potentiates anticancer efficacy of OC in vivo. Collectively, these data demonstrated that OC is a novel autophagic flux inhibitor and might be useful in anticancer therapy.
Collapse
Affiliation(s)
- Yuanzhi Lao
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Gang Wan
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China; School of Life Sciences; Tsinghua University; Beijing, China
| | - Zhenyan Liu
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Xiaoyu Wang
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Ping Ruan
- Department of Pathology; Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine; Nanning, China
| | - Wei Xu
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China; School of Life Sciences; Tsinghua University; Beijing, China
| | - Danqing Xu
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China; Department of Discovery Technologies; Roche R&D Center (China) Ltd; Shanghai, China
| | - Weidong Xie
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China
| | - Yaou Zhang
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China
| | - Hongxi Xu
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Naihan Xu
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China
| |
Collapse
|
264
|
Miller B, Friedman AJ, Choi H, Hogan J, McCammon JA, Hook V, Gerwick WH. The marine cyanobacterial metabolite gallinamide A is a potent and selective inhibitor of human cathepsin L. JOURNAL OF NATURAL PRODUCTS 2014; 77:92-9. [PMID: 24364476 PMCID: PMC3932306 DOI: 10.1021/np400727r] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
A number of marine natural products are potent inhibitors of proteases, an important drug target class in human diseases. Hence, marine cyanobacterial extracts were assessed for inhibitory activity to human cathepsin L. Herein, we have shown that gallinamide A potently and selectively inhibits the human cysteine protease cathepsin L. With 30 min of preincubation, gallinamide A displayed an IC50 of 5.0 nM, and kinetic analysis demonstrated an inhibition constant of ki = 9000 ± 260 M(-1) s(-1). Preincubation-dilution and activity-probe experiments revealed an irreversible mode of inhibition, and comparative IC50 values display a 28- to 320-fold greater selectivity toward cathepsin L than closely related human cysteine cathepsin V or B. Molecular docking and molecular dynamics simulations were used to determine the pose of gallinamide in the active site of cathepsin L. These data resulted in the identification of a pose characterized by high stability, a consistent hydrogen bond network, and the reactive Michael acceptor enamide of gallinamide A positioned near the active site cysteine of the protease, leading to a proposed mechanism of covalent inhibition. These data reveal and characterize the novel activity of gallinamide A as a potent inhibitor of human cathepsin L.
Collapse
Affiliation(s)
- Bailey Miller
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
| | - Aaron J Friedman
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 920393
| | - Hyukjae Choi
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
| | - James Hogan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA
| | - Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - William H. Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
- To whom correspondence should be addressed. Tel: (858) 534-0578. Fax: (858) 534-0529.
| |
Collapse
|
265
|
Kim J, Tung CH, Choi Y. Smart dual-functional warhead for folate receptor-specific activatable imaging and photodynamic therapy. Chem Commun (Camb) 2014; 50:10600-3. [DOI: 10.1039/c4cc04166f] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Folic acid as a bi-functional warhead to develop smart dual-targeted theranostic agents.
Collapse
Affiliation(s)
- Jisu Kim
- Molecular Imaging & Therapy Branch
- National Cancer Center
- Goyang-si, Korea
| | - Ching-Hsuan Tung
- Molecular Imaging Innovations Institute
- Department of Radiology
- Weil Cornell Medical College
- New York, USA
| | - Yongdoo Choi
- Molecular Imaging & Therapy Branch
- National Cancer Center
- Goyang-si, Korea
| |
Collapse
|
266
|
Fang BA, Kovačević Ž, Park KC, Kalinowski DS, Jansson PJ, Lane DJR, Sahni S, Richardson DR. Molecular functions of the iron-regulated metastasis suppressor, NDRG1, and its potential as a molecular target for cancer therapy. Biochim Biophys Acta Rev Cancer 2013; 1845:1-19. [PMID: 24269900 DOI: 10.1016/j.bbcan.2013.11.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/11/2013] [Accepted: 11/13/2013] [Indexed: 12/11/2022]
Abstract
N-myc down-regulated gene 1 (NDRG1) is a known metastasis suppressor in multiple cancers, being also involved in embryogenesis and development, cell growth and differentiation, lipid biosynthesis and myelination, stress responses and immunity. In addition to its primary role as a metastasis suppressor, NDRG1 can also influence other stages of carcinogenesis, namely angiogenesis and primary tumour growth. NDRG1 is regulated by multiple effectors in normal and neoplastic cells, including N-myc, histone acetylation, hypoxia, cellular iron levels and intracellular calcium. Further, studies have found that NDRG1 is up-regulated in neoplastic cells after treatment with novel iron chelators, which are a promising therapy for effective cancer management. Although the pathways by which NDRG1 exerts its functions in cancers have been documented, the relationship between the molecular structure of this protein and its functions remains unclear. In fact, recent studies suggest that, in certain cancers, NDRG1 is post-translationally modified, possibly by the activity of endogenous trypsins, leading to a subsequent alteration in its metastasis suppressor activity. This review describes the role of this important metastasis suppressor and discusses interesting unresolved issues regarding this protein.
Collapse
Affiliation(s)
- Bernard A Fang
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Blackburn Building (D06), The University of Sydney, Sydney, NSW 2006, Australia
| | - Žaklina Kovačević
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Blackburn Building (D06), The University of Sydney, Sydney, NSW 2006, Australia
| | - Kyung Chan Park
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Blackburn Building (D06), The University of Sydney, Sydney, NSW 2006, Australia
| | - Danuta S Kalinowski
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Blackburn Building (D06), The University of Sydney, Sydney, NSW 2006, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Blackburn Building (D06), The University of Sydney, Sydney, NSW 2006, Australia
| | - Darius J R Lane
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Blackburn Building (D06), The University of Sydney, Sydney, NSW 2006, Australia
| | - Sumit Sahni
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Blackburn Building (D06), The University of Sydney, Sydney, NSW 2006, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Blackburn Building (D06), The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
267
|
Capecci J, Forgac M. The function of vacuolar ATPase (V-ATPase) a subunit isoforms in invasiveness of MCF10a and MCF10CA1a human breast cancer cells. J Biol Chem 2013; 288:32731-32741. [PMID: 24072707 DOI: 10.1074/jbc.m113.503771] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The vacuolar H(+) ATPases (V-ATPases) are ATP-driven proton pumps that transport protons across both intracellular and plasma membranes. Previous studies have implicated V-ATPases in the invasiveness of various cancer cell lines. In this study, we evaluated the role of V-ATPases in the invasiveness of two closely matched human breast cancer lines. MCF10a cells are a non-invasive, immortalized breast epithelial cell line, and MCF10CA1a cells are a highly invasive, H-Ras-transformed derivative of MCF10a cells selected for their metastatic potential. Using an in vitro Matrigel assay, MCF10CA1a cells showed a much higher invasion than the parental MCF10a cells. Moreover, this increased invasion was completely sensitive to the specific V-ATPase inhibitor concanamycin. MCF10CA1a cells expressed much higher levels of both a1 and a3 subunit isoforms relative to the parental line. Isoforms of subunit a are responsible for subcellular localization of V-ATPases, with a3 and a4 targeting V-ATPases to the plasma membrane of specialized cells. Knockdown of either a3 alone or a3 and a4 together using isoform-specific siRNAs inhibited invasion by MCF10CA1a cells. Importantly, overexpression of a3 but not the other a subunit isoforms greatly increased the invasiveness of the parental MCF10a cells. Similarly, overexpression of a3 significantly increased expression of V-ATPases at the plasma membrane. These studies suggest that breast tumor cells employ particular a subunit isoforms to target V-ATPases to the plasma membrane, where they function in tumor cell invasion.
Collapse
Affiliation(s)
- Joseph Capecci
- From the Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Michael Forgac
- From the Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine and the Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111.
| |
Collapse
|
268
|
Shankar R, Samykutty A, Riggin C, Kannan S, Wenzel U, Kolhatkar R. Cathepsin B degradable star-shaped peptidic macromolecules for delivery of 2-methoxyestradiol. Mol Pharm 2013; 10:3776-88. [PMID: 23971990 DOI: 10.1021/mp400261h] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
2-Methoxyestradiol (2ME), a natural metabolite of estradiol, has antiproliferative and antiangiogenic activity. However, its clinical success is limited due to poor water solubility and poor pharmacokinetic parameters suggesting the need for a delivery vehicle. In this study we evaluated cathepsin B degradable star-shaped peptidic macromolecules (SPMs) that can potentially be used to create higher generation and high molecular weight peptidic polymer as delivery vehicle of 2ME. Two peptidic macromolecules having positively charged amine (ASPM) or negatively charged carboxyl surface groups (CSPM) were synthesized and evaluated for their degradation in the presence of cathepsin B and stability in the presence of neutral or acidic buffer and serum. Both ASPM and CSPM degraded rapidly in the presence of cathepsin B. Both were stable in neutral and acidic buffer whereas only CSPM exhibited substantial stability in the presence of serum. Both macromolecules were nontoxic toward breast cancer cells whereas 2ME-containing macromolecules exhibited antiproliferative activity in the micromolar range. Overall, results from the current study indicate that tetrapeptide GFLG can be used to create star-shaped macromolecules that are degraded in the presence of cathepsin B and have the potential to be developed as delivery vehicles of 2ME.
Collapse
Affiliation(s)
- Ravi Shankar
- Department of Biopharmaceutical Sciences, University of Illinois Chicago , Rockford, Illinois 61107, United States
| | | | | | | | | | | |
Collapse
|
269
|
HUANG ANCHENG, LIEN JINCHERNG, LIN MENGWEI, YANG JAISING, WU PINGPING, CHANG SHUJEN, LAI TUNGYUAN. Tetrandrine induces cell death in SAS human oral cancer cells through caspase activation-dependent apoptosis and LC3-I and LC3-II activation-dependent autophagy. Int J Oncol 2013; 43:485-94. [DOI: 10.3892/ijo.2013.1952] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 04/08/2013] [Indexed: 11/05/2022] Open
|