51
|
Dentis JL, Schreiber NB, Gilliam JN, Schutz LF, Spicer LJ. Changes in brain ribonuclease (BRB) messenger RNA in granulosa cells (GCs) of dominant vs subordinate ovarian follicles of cattle and the regulation of BRB gene expression in bovine GCs. Domest Anim Endocrinol 2016; 55:32-40. [PMID: 26773365 PMCID: PMC4779677 DOI: 10.1016/j.domaniend.2015.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 11/15/2022]
Abstract
Brain ribonuclease (BRB) is a member of the ribonuclease A superfamily that is constitutively expressed in a range of tissues and is the functional homolog of human ribonuclease 1. This study was designed to characterize BRB gene expression in granulosa cells (GCs) during development of bovine dominant ovarian follicles and to determine the hormonal regulation of BRB in GCs. Estrous cycles of Holstein cows (n = 18) were synchronized, and cows were ovariectomized on either day 3 to 4 or day 5 to 6 after ovulation during dominant follicle growth and selection. Ovaries were collected, follicular fluid (FFL) was aspirated, and GCs were collected for RNA isolation and quantitative polymerase chain reaction. Follicles were categorized as small (1-5 mm; pooled per ovary), medium (5-8 mm; individually collected), or large (8.1-17 mm; individually collected) based on surface diameter. Estradiol (E2) and progesterone (P4) levels were measured by radioimmunoassay (RIA) in FFL. Abundance of BRB messenger RNA (mRNA) in GCs was 8.6- to 11.8-fold greater (P < 0.05) in small (n = 31), medium (n = 66), and large (n = 33) subordinate E2-inactive (FFL E2 < P4) follicles than in large (n = 16) dominant E2-active (FFL E2 > P4) follicles. In the largest 4 follicles, GCs BRB mRNA abundance was negatively correlated (P < 0.01) with FFL E2 (r = -0.65) and E2:P4 ratio (r = -0.46). In experiment 2, GCs from large (8-22 mm diameter) and small (1-5 mm diameter) follicles were treated with insulin-like growth factor 1 (IGF1; 0 or 30 ng/mL) and/or tumor necrosis factor alpha (0 or 30 ng/mL); IGF1 increased (P < 0.05) BRB mRNA abundance, and tumor necrosis factor alpha decreased (P < 0.001) the IGF1-induced BRB mRNA abundance in large-follicle GCs. In experiment 3 to 6, E2, follicle-stimulating hormone, fibroblast growth factor 9, cortisol, wingless 3A, or sonic hedgehog did not affect (P > 0.10) abundance of BRB mRNA in GCs; thyroxine and luteinizing hormone increased (P < 0.05), whereas prostaglandin E2 (PGE2) decreased (P < 0.05) BRB mRNA abundance in small-follicle GCs. Treatment of small-follicle GCs with recombinant human RNase1 increased (P < 0.05) GCs numbers and E2 production. In conclusion, BRB is a hormonally and developmentally regulated gene in bovine GCs and may regulate E2 production during follicular growth in cattle.
Collapse
Affiliation(s)
- J L Dentis
- Department of Animal Science, Oklahoma State University, Stillwater, OK, 74078, USA
| | - N B Schreiber
- Department of Animal Science, Oklahoma State University, Stillwater, OK, 74078, USA
| | - J N Gilliam
- Department of Veterinary Clinical Sciences, Oklahoma State University, Stillwater, OK, 74078, USA
| | - L F Schutz
- Department of Animal Science, Oklahoma State University, Stillwater, OK, 74078, USA
| | - L J Spicer
- Department of Animal Science, Oklahoma State University, Stillwater, OK, 74078, USA.
| |
Collapse
|
52
|
Burnysheva KM, Petrushanko IY, Spirin PV, Prassolov VS, Makarov AA, Mitkevich VA. Ribonuclease binase induces death in T-cell acute lymphoblastic leukemia cells by apoptosis. Mol Biol 2016. [DOI: 10.1134/s0026893316020035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
53
|
Wang Z, Lin F, Liu J, Qiu F. A Novel Ribonuclease from Rana Chensinensis and Its Potential for the Treatment of Human Breast Cancer. Cancer Biother Radiopharm 2015; 30:380-5. [PMID: 26502078 DOI: 10.1089/cbr.2015.1891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Onconase, a member of the pancreatic RNAase A superfamily of ribonucleases, is a chemotherapeutic agent, which has demonstrated selective antitumor activity in a variety of human malignancies. However, little is known about the mechanisms of it's action on human breast cancer cells. To investigate a novel Onconase from the frog of Rana chensinensis changbaishanensis on human breast cancer cells and the underlying mechanisms, a novel Onconase named Rdchonc from Rana chensinensis changbaishanensis was cloned by polymerase chain reaction. SDS-PAGE revealed that the Rdchonc had a high heterologous expression in Escherichia coli BL21(DE3). The MTT assay indicated that purified Rdchonc was cytotoxic to human breast cancer MCF-7 and MD-MB-231 cells. Treatment with 20 μg/mL Rdchonc protein significantly reduced the invasive capacities of MCF-7 and MD-MB-231 cells. Interestingly, the authors found that such inhibitory effort on tumor cell growth induced by Rdchonc treatment may be explained by the regulation of proapoptotic Bcl-2 family proteins and inhibition of MEK/ERK phosphorylation.
Collapse
Affiliation(s)
- Zuozhao Wang
- 1 College of Chemistry, Jilin University , Changchun, People's Republic of China .,2 Laboratory of Nutrition and Functional Food, Jilin University , Changchun, People's Republic of China
| | - Feng Lin
- 1 College of Chemistry, Jilin University , Changchun, People's Republic of China
| | - Jingbo Liu
- 2 Laboratory of Nutrition and Functional Food, Jilin University , Changchun, People's Republic of China
| | - Fangping Qiu
- 1 College of Chemistry, Jilin University , Changchun, People's Republic of China
| |
Collapse
|
54
|
Nesiel-Nuttman L, Schwartz B, Shoseyov O. Human recombinant truncated RNASET2, devoid of RNase activity; A potential cancer therapeutic agent. Oncotarget 2015; 5:11464-78. [PMID: 25426551 PMCID: PMC4294338 DOI: 10.18632/oncotarget.2562] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 10/01/2014] [Indexed: 01/16/2023] Open
Abstract
Human RNASET2 has been implicated in antitumorigenic and antiangiogenic activities, independent of its ribonuclease capacities. We constructed a truncated version of human RNASET2, starting at E50 (trT2-50) and devoid of ribonuclease activity. trT2-50 maintained its ability to bind actin and to inhibit angiogenesis and tumorigenesis. trT2-50 binds to cell surface actin and formed a complex with actin in vitro. The antiangiogenic effect of this protein was demonstrated in human umbilical vein endothelial cells (HUVECs) by its ability to arrest tube formation on Matrigel, induced by angiogenic factors. Immunofluorescence staining of HUVECs showed nuclear and cytosolic RNASET2 protein that was no longer detectable inside the cell following trT2-50 treatment. This effect was associated with disruption of the intracellular actin network. trT2-50 co-localized with angiogenin, suggesting that both molecules bind (or compete) for similar cellular epitopes. Moreover, trT2-50 led to a significant inhibition of tumor development. Histological analysis demonstrated abundant necrotic tissue and a substantial loss of endothelial structure in trT2-50-treated tumors. Collectively, the present results indicate that trT2-50, a molecule engineered to be deficient of its catalytic activity, still maintained its actin binding and anticancer-related biological activities. We therefore suggest that trT2-50 may serve as a potential cancer therapeutic agent.
Collapse
Affiliation(s)
- Liron Nesiel-Nuttman
- The Robert H. Smith Institute of Plant Science and Genetics in Agriculture, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, ISRAEL
| | - Betty Schwartz
- School of Nutritional Sciences Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, ISRAEL
| | - Oded Shoseyov
- The Robert H. Smith Institute of Plant Science and Genetics in Agriculture, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, ISRAEL
| |
Collapse
|
55
|
Glinka EM. Killing of cancer cells through the use of eukaryotic expression vectors harbouring genes encoding nucleases and ribonuclease inhibitor. Tumour Biol 2015; 36:3147-57. [PMID: 25874497 DOI: 10.1007/s13277-015-3360-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/19/2015] [Indexed: 11/29/2022] Open
Abstract
Cancer gene therapy vectors are promising tools for killing cancer cells with the purpose of eradicating malignant tumours entirely. Different delivery methods of vectors into the cancer cells, including both non-viral and viral, as well as promoters for the targeted expression of genes encoding anticancer proteins were developed for effective and selective killing of cancer cells without harming healthy cells. Many vectors have been created to kill cancer cells, and some vectors suppress malignant tumours with high efficiency. This review is focused on vectors bearing genes for nucleases such as deoxyribonucleases (caspase-activated DNase, deoxyribonuclease I-like 3, endonuclease G) and ribonucleases (human polynucleotide phosphorylase, ribonuclease L, α-sarcin, barnase), as well as vectors harbouring gene encoding ribonuclease inhibitor. The data concerning the functionality and the efficacy of such vectors are presented.
Collapse
|
56
|
Fiorini C, Cordani M, Gotte G, Picone D, Donadelli M. Onconase induces autophagy sensitizing pancreatic cancer cells to gemcitabine and activates Akt/mTOR pathway in a ROS-dependent manner. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:549-60. [PMID: 25533084 DOI: 10.1016/j.bbamcr.2014.12.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/05/2014] [Accepted: 12/11/2014] [Indexed: 12/28/2022]
Abstract
Onconase® (ONC) is a member of the RNase super-family that is secreted in oocytes and early embryos of Rana pipiens. Over the last years, research interest about this small and basic frog RNase, also called ranpirnase, constantly increased because of its high cytotoxicity and anticancer properties. Onconase is currently used in clinical trials for cancer therapy; however, the precise mechanisms determining cytotoxicity in cancer cells have not yet been fully investigated. In the present manuscript, we evaluate the antitumoral property of onconase in pancreatic adenocarcinoma cells and in non-tumorigenic cells as a control. We demonstrate that ONC stimulates a strong antiproliferative and proapoptotic effect in cancer cells by reporting for the first time that ONC triggers Beclin1-mediated autophagic cancer cell death. In addition, ONC inhibits the expression of mitochondrial uncoupling protein 2 (UCP2) and of manganese-dependent superoxide dismutase (MnSOD) triggering mitochondrial superoxide ion production. ONC-induced reactive oxygen species (ROS) are responsible for Akt/mTOR pathway stimulation determining the sensitivity of cancer cells to mTOR inhibitors and lessening autophagic stimulation. This indicates ROS/Akt/mTOR axis as a strategy adopted by cancer cells to reduce ONC-mediated cytotoxic autophagy stimulation. In addition, we demonstrate that ONC can sensitize pancreatic cancer cells to the standard chemotherapeutic agent gemcitabine allowing a reduction of drug concentration when used in combination settings, thus suggesting a lowering of chemotherapy-related side effects. Altogether, our results shed more light on the mechanisms lying at the basis of ONC antiproliferative effect in cancer cells and support its potential use to develop new anticancer strategies.
Collapse
Affiliation(s)
- Claudia Fiorini
- Department of Life and Reproduction Sciences, Biochemistry Section, University of Verona, Verona, Italy
| | - Marco Cordani
- Department of Life and Reproduction Sciences, Biochemistry Section, University of Verona, Verona, Italy
| | - Giovanni Gotte
- Department of Life and Reproduction Sciences, Biochemistry Section, University of Verona, Verona, Italy
| | - Delia Picone
- Department of Chemical Sciences, University of Naples "Federico II", Naples, Italy
| | - Massimo Donadelli
- Department of Life and Reproduction Sciences, Biochemistry Section, University of Verona, Verona, Italy.
| |
Collapse
|
57
|
Fernández-Ulibarri I, Hammer K, Arndt MAE, Kaufmann JK, Dorer D, Engelhardt S, Kontermann RE, Hess J, Allgayer H, Krauss J, Nettelbeck DM. Genetic delivery of an immunoRNase by an oncolytic adenovirus enhances anticancer activity. Int J Cancer 2014; 136:2228-40. [PMID: 25303768 DOI: 10.1002/ijc.29258] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 09/22/2014] [Indexed: 01/27/2023]
Abstract
Antibody therapy of solid cancers is well established, but suffers from unsatisfactory tumor penetration of large immunoglobulins or from low serum retention of antibody fragments. Oncolytic viruses are in advanced clinical development showing excellent safety, but suboptimal potency due to limited virus spread within tumors. Here, by developing an immunoRNase-encoding oncolytic adenovirus, we combine viral oncolysis with intratumoral genetic delivery of a small antibody-fusion protein for targeted bystander killing of tumor cells (viro-antibody therapy). Specifically, we explore genetic delivery of a small immunoRNase consisting of an EGFR-binding scFv antibody fragment fused to the RNase Onconase (ONC(EGFR)) that induces tumor cell death by RNA degradation after cellular internalization. Onconase is a frog RNase that combines lack of immunogenicity and excellent safety in patients with high tumor killing potency due to its resistance to the human cytosolic RNase inhibitor. We show that ONC(EGFR) expression by oncolytic adenoviruses is feasible with an optimized, replication-dependent gene expression strategy. Virus-encoded ONC(EGFR) induces potent and EGFR-dependent bystander killing of tumor cells. Importantly, the ONC(EGFR)-encoding oncolytic adenovirus showed dramatically increased cytotoxicity specifically to EGFR-positive tumor cells in vitro and significantly enhanced therapeutic activity in a mouse xenograft tumor model. The latter demonstrates that ONC(EGFR) is expressed at levels sufficient to trigger tumor cell killing in vivo. The established ONC(EGFR)-encoding oncolytic adenovirus represents a novel agent for treatment of EGFR-positive tumors. This viro-antibody therapy platform can be further developed for targeted/personalized cancer therapy by exploiting antibody diversity to target further established or emerging tumor markers or combinations thereof.
Collapse
Affiliation(s)
- Inés Fernández-Ulibarri
- Oncolytic Adenovirus Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Mitkevich VA, Burnysheva KM, Ilinskaya ON, Pace CN, Makarov AA. Cytotoxicity of RNase Sa to the acute myeloid leukemia Kasumi-1 cells depends on the net charge. Oncoscience 2014; 1:738-44. [PMID: 25594000 PMCID: PMC4278273 DOI: 10.18632/oncoscience.97] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 11/10/2014] [Indexed: 01/21/2023] Open
Abstract
The majority of known cytotoxic RNases are basic proteins which destroy intracellular RNA. Cationization of RNases is considered to be an effective strategy for strengthening their antitumor properties. We constructed a set of RNase Sa variants consisting of charge reversal mutants, charge neutralization mutants, and variants with positively charged cluster at the N-terminus. All constructs retain a high level of catalytic activity and differ in net charge. Using acute myeloid leukemia cells Kasumi-1 we have shown that (i) cytotoxicity of RNase Sa mutants is linearly enhanced by cationization, (ii) the ability of cytotoxic mutants to induce cell death is caused by induction of apoptosis and (iii) localization of positive charge on N-terminus does not contribute to RNase Sa cytotoxicity. Capacity to induce apoptosis in malignant cells and the absence of necrotic effects make the RNase Sa mutants with high positive charge a suitable anti-cancer agent.
Collapse
Affiliation(s)
- Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ksenia M Burnysheva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Olga N Ilinskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia ; Department of Microbiology, Kazan Federal (Volga-Region) University, Kazan, Russia
| | - C Nick Pace
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA ; Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas, USA
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
59
|
Arnold U. Stability and folding of amphibian ribonuclease A superfamily members in comparison with mammalian homologues. FEBS J 2014; 281:3559-75. [PMID: 24966023 DOI: 10.1111/febs.12891] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 06/18/2014] [Indexed: 01/05/2023]
Abstract
Comparative studies on homologous proteins can provide knowledge on how limited changes in the primary structure find their expression in large effects on catalytic activity, stability or the folding behavior. For more than half a century, members of the ribonuclease A superfamily have been the subject of a myriad of studies on protein folding and stability. Both the unfolding and refolding kinetics as well as the structure of several folding intermediates of ribonuclease A have been characterized in detail. Moreover, the RNA-degrading activity of these enzymes provides a basis for their cytotoxicity, which renders them potential tumor therapeutics. Because amphibian ribonuclease A homologues evade the human ribonuclease inhibitor, they emerged as particularly promising candidates. Interestingly, the amphibian ribonuclease A homologues investigated to date are more stable than the mammalian homologues. Nevertheless, despite the generation of numerous genetically engineered variants, knowledge of the folding of amphibian ribonuclease A homologues remains rather limited. An exception is onconase, a ribonuclease A homologue from Rana pipiens, which has been characterized in detail. This review summarizes the data on the unfolding and refolding kinetics and pathways, as well on the stability of amphibian ribonuclease A homologues compared with those of ribonuclease A, the best known member of this superfamily.
Collapse
Affiliation(s)
- Ulrich Arnold
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Germany
| |
Collapse
|
60
|
Site-specific RNase A activity was dramatically reduced in serum from multiple types of cancer patients. PLoS One 2014; 9:e96490. [PMID: 24805924 PMCID: PMC4013009 DOI: 10.1371/journal.pone.0096490] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 04/08/2014] [Indexed: 01/13/2023] Open
Abstract
Potent RNase activities were found in the serum of mammals but the physiological function of the RNases was never well illustrated, largely due to the caveats in methods of RNase activity measurement. None of the existing methods can distinguish between RNases with different target specificities. A systematic study was recently carried out in our lab to investigate the site-specificity of serum RNases on double-stranded RNA substrates, and found that serum RNases cleave double-stranded RNAs predominantly at 5′-U/A-3′ and 5′-C/A-3′ dinucleotide sites, in a manner closely resembling RNase A. Based on this finding, a FRET assay was developed in the current study to measure this site-specific serum RNase activity in human samples using a double stranded RNA substrate. We demonstrated that the method has a dynamic range of 10−5 mg/ml- 10−1 mg/ml using serial dilution of RNase A. The sera of 303 cancer patients were subjected to comparison with 128 healthy controls, and it was found that serum RNase activities visualized with this site-specific double stranded probe were found to be significantly reduced in patients with gastric cancer, liver cancer, pancreatic cancer, esophageal cancer, ovary cancer, cervical cancer, bladder cancer, kidney cancer and lung cancer, while only minor changes were found in breast and colon cancer patients. This is the first report using double stranded RNA as probe to quantify site-specific activities of RNase A in a serum. The results illustrated that RNase A might be further evaluated to determine if it can serve as a new class of biomarkers for certain cancer types.
Collapse
|
61
|
|
62
|
SMOLEWSKI PIOTR, WITKOWSKA MAGDALENA, ZWOLINSKA MALGORZATA, CEBULA-OBRZUT BARBARA, MAJCHRZAK AGATA, JESKE ALEKSANDRA, DARZYNKIEWICZ ZBIGNIEW, ARDELT WOJCIECH, ARDELT BARBARA, ROBAK TADEUSZ. Cytotoxic activity of the amphibian ribonucleases onconase and r-amphinase on tumor cells from B cell lymphoproliferative disorders. Int J Oncol 2014; 45:419-25. [DOI: 10.3892/ijo.2014.2405] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 01/03/2014] [Indexed: 11/05/2022] Open
|
63
|
Leczyme: a new candidate drug for cancer therapy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:421415. [PMID: 24864241 PMCID: PMC4017849 DOI: 10.1155/2014/421415] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/27/2014] [Indexed: 11/18/2022]
Abstract
Sialic acid-binding lectin (SBL), isolated from oocytes of Rana catesbeiana, is leczyme and has both lectin and ribonuclease (RNase) activities. A remarkable antitumor effect of SBL has also been reported. SBL agglutinates various kinds of tumor cells but not normal cells. SBL agglutination activity is not affected by mono- or oligosaccharides. However, SBL-induced agglutination and antitumor effects are inhibited by sialomucin but not asialomucin. In addition, SBL has very little effect on sialidase-treated cells. SBL causes cancer-selective induction of apoptosis by multiple signaling pathways, which target RNA. Synergistic antitumor effects with other molecules, such as tumor necrosis factor-related apoptosis ligand (TRAIL) and interferon-γ (IFN-γ), have been reported. Thus, SBL may be a novel candidate molecule for anticancer drug development. Sialoglycoconjugates on the tumor cell surface may be associated with lectin activity and antitumor effects of SBL. We review the properties of SBL, particularly its lectin, RNase, and antitumor activities, and comprehensively examine the potential application of SBL for clinical purposes.
Collapse
|
64
|
Shih YP, Chou CC, Chen YL, Huang KF, Wang AHJ. Linked production of pyroglutamate-modified proteins via self-cleavage of fusion tags with TEV protease and autonomous N-terminal cyclization with glutaminyl cyclase in vivo. PLoS One 2014; 9:e94812. [PMID: 24733552 PMCID: PMC3986218 DOI: 10.1371/journal.pone.0094812] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 03/19/2014] [Indexed: 12/28/2022] Open
Abstract
Overproduction of N-terminal pyroglutamate (pGlu)-modified proteins utilizing Escherichia coli or eukaryotic cells is a challenging work owing to the fact that the recombinant proteins need to be recovered by proteolytic removal of fusion tags to expose the N-terminal glutaminyl or glutamyl residue, which is then converted into pGlu catalyzed by the enzyme glutaminyl cyclase. Herein we describe a new method for production of N-terminal pGlu-containing proteins in vivo via intracellular self-cleavage of fusion tags by tobacco etch virus (TEV) protease and then immediate N-terminal cyclization of passenger target proteins by a bacterial glutaminyl cyclase. To combine with the sticky-end PCR cloning strategy, this design allows the gene of target proteins to be efficiently inserted into the expression vector using two unique cloning sites (i.e., SnaB I and Xho I), and the soluble and N-terminal pGlu-containing proteins are then produced in vivo. Our method has been successfully applied to the production of pGlu-modified enhanced green fluorescence protein and monocyte chemoattractant proteins. This design will facilitate the production of protein drugs and drug target proteins that possess an N-terminal pGlu residue required for their physiological activities.
Collapse
Affiliation(s)
- Yan-Ping Shih
- Institute of Biological Chemistry and Core Facilities for Protein Structural Analysis, Academia Sinica, Taipei, Taiwan
| | - Chi-Chi Chou
- Institute of Biological Chemistry and Core Facilities for Protein Structural Analysis, Academia Sinica, Taipei, Taiwan
| | - Yi-Ling Chen
- Institute of Biological Chemistry and Core Facilities for Protein Structural Analysis, Academia Sinica, Taipei, Taiwan
| | - Kai-Fa Huang
- Institute of Biological Chemistry and Core Facilities for Protein Structural Analysis, Academia Sinica, Taipei, Taiwan
- * E-mail: (AHJW); (KFH)
| | - Andrew H.- J. Wang
- Institute of Biological Chemistry and Core Facilities for Protein Structural Analysis, Academia Sinica, Taipei, Taiwan
- * E-mail: (AHJW); (KFH)
| |
Collapse
|
65
|
Structure, mechanism, and specificity of a eukaryal tRNA restriction enzyme involved in self-nonself discrimination. Cell Rep 2014; 7:339-347. [PMID: 24726365 DOI: 10.1016/j.celrep.2014.03.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/26/2014] [Accepted: 03/11/2014] [Indexed: 11/22/2022] Open
Abstract
tRNA restriction by anticodon nucleases underlies cellular stress responses and self-nonself discrimination in a wide range of taxa. Anticodon breakage inhibits protein synthesis, which, in turn, results in growth arrest or cell death. The eukaryal ribotoxin PaT secreted by Pichia acaciae inhibits growth of Saccharomyces cerevisiae via cleavage of tRNA(Gln(UUG)). We find that recombinant PaT incises a synthetic tRNA(Gln(UUG)) stem-loop RNA by transesterification at a single site 3' of the wobble uridine, yielding 2',3'-cyclic phosphate and 5'-OH ends. Incision is suppressed by replacement of the wobble nucleobase with adenine or guanine. The crystal structure of PaT reveals a distinctive fold and active site, essential components of which are demonstrated by mutagenesis. Pichia acaciae evades self-toxicity via a distinctive intracellular immunity protein, ImmPaT, which binds PaT and blocks nuclease activity. Our results highlight the evolutionary diversity of tRNA restriction and immunity systems.
Collapse
|
66
|
Yiang GT, Tsai HF, Chen JR, Chou PL, Wu TK, Liu HC, Chang WJ, Liu LC, Tseng HH, Yu YL. RC-6 ribonuclease induces caspase activation, cellular senescence and neuron-like morphology in NT2 embryonal carcinoma cells. Oncol Rep 2014; 31:1738-44. [PMID: 24535104 DOI: 10.3892/or.2014.3023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 11/25/2013] [Indexed: 11/05/2022] Open
Abstract
Frog ribonucleases have been demonstrated to have anticancer activities. However, whether RC-6 ribonuclease exerts anticancer activity on human embryonal carcinoma cells remains unclear. In the present study, RC-6 induced cytotoxicity in NT2 cells (a human embryonal carcinoma cell line) and our studies showed that RC-6 can exert anticancer effects and induce caspase-9 and -3 activities. Moreover, to date, there is no evidence that frog ribonuclease-induced cytotoxicity effects are related to cellular senescence. Therefore, our studies showed that RC-6 can increase p16 and p21 protein levels and induce cellular senescence in NT2 cells. Notably, similar to retinoic acid-differentiated NT2 cells, neuron-like morphology was found on some remaining live cells after RC-6 treatment. In conclusion, our study is the first to demonstrate that RC-6 can induce cytotoxic effects, caspase-9/-3 activities, cellular senescence and neuron-like morphology in NT2 cells.
Collapse
Affiliation(s)
- Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan, R.O.C
| | - Hsiu-Feng Tsai
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan, R.O.C
| | - Jer-Rong Chen
- Department of Surgery, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| | - Pei-Lun Chou
- Division of Allergy-Immunology-Rheumatology, Department of Internal Medicine, Saint Mary's Hospital Luodong, Yilan 265, Taiwan, R.O.C
| | - Tsai-Kun Wu
- Division of Renal Medicine, Tungs' Taichung Metroharbor Hospital, Taichung 435, Taiwan, R.O.C
| | - Hsiao-Chun Liu
- Department of Nursing, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan, R.O.C
| | - Wei-Jung Chang
- Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Liang-Chih Liu
- Department of Surgery, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| | - Hsu-Hung Tseng
- Division of General Surgery, Taichung Hospital, Ministry of Health and Welfare, Taichung 403, Taiwan, R.O.C
| | - Yung-Luen Yu
- Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
| |
Collapse
|
67
|
Structural and functional relationships of natural and artificial dimeric bovine ribonucleases: new scaffolds for potential antitumor drugs. FEBS Lett 2013; 587:3601-8. [PMID: 24113657 DOI: 10.1016/j.febslet.2013.09.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/23/2013] [Accepted: 09/24/2013] [Indexed: 11/20/2022]
Abstract
Protein aggregation via 3D domain swapping is a complex mechanism which can lead to the acquisition of new biological, benign or also malignant functions, such as amyloid deposits. In this context, RNase A represents a fascinating model system, since by dislocating different polypeptide chain regions, it forms many diverse oligomers. No other protein displays such a large number of different quaternary structures. Here we report a comparative structural analysis between natural and artificial RNase A dimers and bovine seminal ribonuclease, a natively dimeric RNase with antitumor activity, with the aim to design RNase A derivatives with improved pharmacological potential.
Collapse
|
68
|
Shah Mahmud R, Ilinskaya O. Antiviral Activity of Binase against the Pandemic Influenza A (H1N1) Virus. Acta Naturae 2013; 5:44-51. [PMID: 24455182 PMCID: PMC3890988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The lack of effective antiviral drugs restricts the control of the dangerous RNA-containing influenza A (H1N1) virus. Extracellular ribonuclease of Bacilli (binase) was shown to manifest antiviral activity during single- and multi-cycle viral replication in the range of concentrations non-toxic to epithelial cells and 0.01-0.1 multiplicity of infection. During antiviral treatment for 15-30 min, the concentration of 1 μg/ml binase reduced the amount of focus-forming units of viruses by a factor of 3-10 and suppressed the virus-induced cytopathic effect in A549 human lung cells. The possible mechanisms of interaction between the virus and enzyme are discussed. Positive charges in both binase and viral hemagglutinin cause electrostatic interaction with negatively charged sialic acid on the host cell's surface followed by its penetration into the cell. Capsid elimination and release of viral RNA from endosome to the cytoplasm allows catalytic RNA cleavage by internalized binase. The data obtained confirm that binase is an effective antiviral agent against the pandemic influenza A (H1N1) virus. Certain progress in this field is associated with clarifying the detailed mechanism underlying the antiviral action of binase and development of the most effective way for its practical use.
Collapse
Affiliation(s)
- R. Shah Mahmud
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kremlyovskaya Str., 18, Kazan, Russia, 420008
| | - O.N. Ilinskaya
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kremlyovskaya Str., 18, Kazan, Russia, 420008
| |
Collapse
|
69
|
Sundlass NK, Eller CH, Cui Q, Raines RT. Contribution of electrostatics to the binding of pancreatic-type ribonucleases to membranes. Biochemistry 2013; 52:6304-12. [PMID: 23947917 DOI: 10.1021/bi400619m] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Pancreatic-type ribonucleases show clinical promise as chemotherapeutic agents but are limited in efficacy by the inefficiency of their uptake by human cells. Cellular uptake can be increased by the addition of positive charges to the surface of ribonucleases, either by site-directed mutagenesis or by chemical modification. This observation has led to the hypothesis that ribonuclease uptake by cells depends on electrostatics. Here, we use a combination of experimental and computational methods to ascertain the contribution of electrostatics to the cellular uptake of ribonucleases. We focus on three homologous ribonucleases: Onconase (frog), ribonuclease A (cow), and ribonuclease 1 (human). Our results support the hypothesis that electrostatics are necessary for the cellular uptake of Onconase. In contrast, specific interactions with cell-surface components likely contribute more to the cellular uptake of ribonuclease A and ribonuclease 1 than do electrostatics. These findings provide insight for the design of new cytotoxic ribonucleases.
Collapse
Affiliation(s)
- Nadia K Sundlass
- Medical Scientist Training Program and Graduate Program in Biophysics, ‡Department of Biochemistry, and §Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | | | | | | |
Collapse
|
70
|
Mironova NL, Petrushanko IY, Patutina OA, Sen'kova AV, Simonenko OV, Mitkevich VA, Markov OV, Zenkova MA, Makarov AA. Ribonuclease binase inhibits primary tumor growth and metastases via apoptosis induction in tumor cells. Cell Cycle 2013; 12:2120-31. [PMID: 23759588 DOI: 10.4161/cc.25164] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Exogenous ribonucleases are known to inhibit tumor growth via apoptosis induction in tumor cells, allowing to consider them as promising anticancer drugs for clinical application. In this work the antitumor potential of binase was evaluated in vivo and the mechanism of cytotoxic effect of binase on tumor cells was comprehensively studied in vitro. We investigated tumoricidal activity of binase using three murine tumor models of Lewis lung carcinoma (LLC), lymphosarcoma RLS 40 and melanoma B-16. We show for the first time that intraperitoneal injection of binase at a dose range 0.1-5 mg/kg results in retardation of primary tumor growth up to 45% in LLC and RLS 40 and inhibits metastasis up to 50% in LLC and RLS 40 and up to 70% in B-16 melanoma. Binase does not exhibit overall toxic effect and displays a general systemic and immunomodulatory effects. Treatment of RLS 40-bearing animals with binase together with polychemotherapy revealed that binase decreases the hepatotoxicity of polychemotherapy while maintaining its antitumor effect. It was demonstrated that the cytotoxic effect of binase is realized via the induction of the intrinsic and extrinsic apoptotic pathways. Activation of intrinsic apoptotic pathway is manifested by a drop of mitochondrial potential, increase in calcium concentration and inhibition of respiratory activity. Subsequent synthesis of TNF-α in the cells under the action of binase triggers extrinsic apoptotic pathway through the binding of TNF with cell-death receptors and activation of caspase 8. Thus binase is a potential anticancer therapeutics inducing apoptosis in cancer cells.
Collapse
Affiliation(s)
- Nadezhda L Mironova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Shirshikov FV, Cherepnev GV, Ilinskaya ON, Kalacheva NV. A hydrophobic segment of some cytotoxic ribonucleases. Med Hypotheses 2013; 81:328-34. [PMID: 23679997 DOI: 10.1016/j.mehy.2013.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 03/23/2013] [Accepted: 04/04/2013] [Indexed: 12/20/2022]
Abstract
The exact mechanism by which cytotoxic ribonucleases reach the cytosol of tumor cells remains unclear. The interaction of ribonucleases with a lipid bilayer is involved in the translocation of ribonucleases across the endosomal membrane. Here, we aimed to study the hydropathy character of toxic antitumor ribonucleases (bovine seminal ribonuclease and binase) and two non-toxic ribonucleases (bovine pancreatic ribonuclease and human pancreatic ribonuclease) by sliding-window hydrophobicity analysis. Comparative hydropathy plot analysis of the non-toxic pancreatic ribonucleases and their toxic variants was also performed. The data obtained indicate that some cytotoxic ribonucleases have a hydrophobic segment, which is sterically available for the hydrophobic interaction with a tumor cell membrane and endosomal membrane. After dissociation, subunits of dimeric ribonucleases are probably capable of thermodynamically favorable interaction with the interfacial region of a lipid bilayer. Remarkably the hydrophobic segment is not identified in the amino acid sequences of non-toxic ribonucleases. The paper describes the hydrophobic properties of toxic RNases that are essential for both the model of a lipid-protein interaction and the cytotoxicity mechanism unraveling.
Collapse
Affiliation(s)
- Fedor V Shirshikov
- Department of Microbiology, Kazan Volga Region Federal University, Kazan, Tatarstan, Russia.
| | | | | | | |
Collapse
|
72
|
Cabrera-Fuentes HA, Aslam M, Saffarzadeh M, Kolpakov A, Zelenikhin P, Preissner KT, Ilinskaya ON. Internalization of Bacillus intermedius ribonuclease (BINASE) induces human alveolar adenocarcinoma cell death. Toxicon 2013; 69:219-26. [PMID: 23567038 DOI: 10.1016/j.toxicon.2013.03.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 02/01/2013] [Accepted: 03/22/2013] [Indexed: 11/18/2022]
Abstract
Ribonuclease (RNase) treatment represents a novel mechanism based approach to anticancer therapy as an alternative to the DNA damaging drugs commonly used in clinical practice. Apart from their ribonucleolytic activity, cytotoxic effects have attracted a considerable attention to RNases because of their potential as selective agents for treatment of certain malignancies. Among these enzymes, Binase, an RNase from Bacillus intermedius, has shown promising results. Here, we have found that binase selectively attacked human A549 alveolar adenocarcinoma cells to trigger an apoptotic response, whereas normal lung epithelial cells LEK were not affected by the ribonuclease. The tumor transformation led to the modification of certain cellular characteristics causing cell sensitivity to binase. Although a general mode for RNases cytotoxicity includes their penetration into the cell, translocation to the cytosol and degradation of ribonucleic acid, many aspects of this process have not been fully elucidated. Our data revealed the following time-dependent changes induced by binase in A549 cells: (a) fast permanent internalization of the enzyme during the first hours of treatment; (b) temporary increase in cellular permeability for macromolecules during the 4-6 h of treatment; (c) apoptotic alterations in population after 24 h and (d) DNA fragmentation and cell death after 72 h of treatment with binase. Elucidation of these molecular strategies used by this promising toxin provides us essential information for the development of new anticancer drugs.
Collapse
Affiliation(s)
- Hector Alejandro Cabrera-Fuentes
- Department of Microbiology, Kazan Federal University, Kremlevskaya Str. 18, 420008 Kazan, Republic of Tatarstan, Russian Federation.
| | | | | | | | | | | | | |
Collapse
|
73
|
Mitkevich VA, Kretova OV, Petrushanko IY, Burnysheva KM, Sosin DV, Simonenko OV, Ilinskaya ON, Tchurikov NA, Makarov AA. Ribonuclease binase apoptotic signature in leukemic Kasumi-1 cells. Biochimie 2013; 95:1344-9. [PMID: 23499289 DOI: 10.1016/j.biochi.2013.02.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 02/26/2013] [Indexed: 10/27/2022]
Abstract
Cytotoxic exogenous RNases triggering apoptotic response in malignant cells have potential as anticancer drugs; surprisingly, detailed characterization of the RNase-induced apoptosis has not been conducted so far. Here we show that a cytotoxic RNase from Bacillus intermedius (binase) induces extrinsic and intrinsic apoptotic pathways in leukemic Kasumi-1 cells. The experiments were performed using TaqMan Array Human Apoptosis 96-well Plate for gene expression analysis, and flow cytometry. Cytometric studies demonstrated dissipation of the mitochondrial membrane potential, opening of mitochondrial permeability transition pores, activation of caspases, increase of intracellular Ca(2+) and decrease of reactive oxygen species levels. We found that expression of 62 apoptotic genes is up-regulated, including 16 genes that are highly up-regulated, and only one gene was found to be down-regulated. The highest, 16 fold increase of the expression level was observed for TNF gene. Highly up-regulated genes also include the non-canonical NF-κB signaling pathway and inflammatory caspases 1,4. The obtained results suggest that binase induces evolutionary acquired cellular response to a microbial agent and triggers unusual apoptosis pathway.
Collapse
Affiliation(s)
- Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova Str 32, 119991 Moscow, Russia
| | | | | | | | | | | | | | | | | |
Collapse
|
74
|
García-Mayoral MF, Canales Á, Díaz D, López-Prados J, Moussaoui M, de Paz JL, Angulo J, Nieto PM, Jiménez-Barbero J, Boix E, Bruix M. Insights into the glycosaminoglycan-mediated cytotoxic mechanism of eosinophil cationic protein revealed by NMR. ACS Chem Biol 2013; 8:144-51. [PMID: 23025322 DOI: 10.1021/cb300386v] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Protein-glycosaminoglycan interactions are essential in many biological processes and human diseases, yet how their recognition occurs is poorly understood. Eosinophil cationic protein (ECP) is a cytotoxic ribonuclease that interacts with glycosaminoglycans at the cell surface; this promotes the destabilization of the cellular membrane and triggers ECP's toxic activity. To understand this membrane destabilization event and the differences in the toxicity of ECP and its homologues, the high resolution solution structure of the complex between full length folded ECP and a heparin-derived trisaccharide (O-iPr-α-D-GlcNS6S-α(1-4)-L-IdoA2S-α(1-4)-D-GlcNS6S) has been solved by NMR methods and molecular dynamics simulations. The bound protein retains the tertiary structure of the free protein. The (2)S(0) conformation of the IdoA ring is preferably recognized by the protein. We have identified the precise location of the heparin binding site, dissected the specific interactions responsible for molecular recognition, and defined the structural requirements for this interaction. The structure reveals the contribution of Arg7, Gln14, and His15 in helix α1, Gln40 in strand β1, His64 in loop 4, and His128 in strand β6 in the recognition event and corroborates the previously reported participation of residues Arg34-Asn39. The participation of the catalytic triad (His15, Lys38, His128) in recognizing the heparin mimetic reveals, at atomic resolution, the mechanism of heparin's inhibition of ECP's ribonucleolytic activity. We have integrated all the available data to propose a molecular model for the membrane interaction process. The solved NMR complex provides the structural model necessary to design inhibitors to block ECP's toxicity implicated in eosinophil pathologies.
Collapse
Affiliation(s)
- M. Flor García-Mayoral
- Departamento
de Química
Física Biológica, Instituto de Química Física Rocasolano, CSIC, Madrid, Spain
| | - Ángeles Canales
- Departamento de Química
Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - Dolores Díaz
- Departamento de Biología
Físico Química, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Javier López-Prados
- Departamento de Química
Orgánica y Biológica, Instituto de Investigaciones Químicas, CSIC-Universidad de Sevilla, Sevilla,
Spain
| | - Mohammed Moussaoui
- Departamento de Bioquímica
y Biología Molecular, Facultad de Biociencias, Universidad Autónoma de Barcelona, Barcelona,
Spain
| | - José L. de Paz
- Departamento de Química
Orgánica y Biológica, Instituto de Investigaciones Químicas, CSIC-Universidad de Sevilla, Sevilla,
Spain
| | - Jesús Angulo
- Departamento de Química
Orgánica y Biológica, Instituto de Investigaciones Químicas, CSIC-Universidad de Sevilla, Sevilla,
Spain
| | - Pedro M. Nieto
- Departamento de Química
Orgánica y Biológica, Instituto de Investigaciones Químicas, CSIC-Universidad de Sevilla, Sevilla,
Spain
| | - Jesús Jiménez-Barbero
- Departamento de Biología
Físico Química, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Ester Boix
- Departamento de Bioquímica
y Biología Molecular, Facultad de Biociencias, Universidad Autónoma de Barcelona, Barcelona,
Spain
| | - Marta Bruix
- Departamento
de Química
Física Biológica, Instituto de Química Física Rocasolano, CSIC, Madrid, Spain
| |
Collapse
|
75
|
Nucleotide binding architecture for secreted cytotoxic endoribonucleases. Biochimie 2012; 95:1087-97. [PMID: 23274129 DOI: 10.1016/j.biochi.2012.12.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 12/13/2012] [Indexed: 12/20/2022]
Abstract
Vertebrate secreted RNases are small cationic protein endowed with an endoribonuclease activity that belong to the RNase A superfamily and display diverse cytotoxic activities. In an effort to unravel their mechanism of action, we have analysed their nucleotide binding recognition patterns. General shared features with other nucleotide binding proteins were deduced from overall statistics on the available structure complexes at the Protein Data Bank and compared with the particularities of selected representative endoribonuclease families. Results were compared with other endoribonuclease representative families and with the overall protein-nucleotide interaction features. Preferred amino acids and atom types involved in pair bonding interactions were identified, defining the spatial motives for phosphate, base and ribose building blocks. Together with the conserved catalytic triad at the active site, variability was observed for secondary binding subsites that may contribute to the proper substrate alignment and could explain the distinct substrate preference patterns. Highly conserved binding patterns were identified for the pyrimidine and purine subsites at the main and secondary base subsites. Particular substitution could be ascribed to specific adenine or guanine specificities. Distribution of evolutionary conserved residues were compared to search for the structure determinants that underlie their diverse catalytic efficiency and those that may account for putative physiological substrate targets or other non-catalytic biological activities that contribute to the antipathogen role of the RNases involved in the host defence system. A side by side comparison with another endoribonuclease superfamily of secreted cytotoxic proteins, the microbial RNases, was carried on to analyse the common features and peculiarities that rule their substrate recognition. The data provides the structural basis for the development of applied therapies targeting cellular nucleotide polymers.
Collapse
|
76
|
Cui L, Peng H, Zhang R, Chen Y, Zhao L, Tang K. Recombinant hHscFv-RC-RNase protein derived from transgenic tobacco acts as a bifunctional molecular complex against hepatocellular carcinoma. Biotechnol Appl Biochem 2012; 59:323-9. [PMID: 23586908 DOI: 10.1002/bab.1039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 08/09/2012] [Indexed: 01/03/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common clinical primary malignant tumor; however, efficient drugs for the treatment of HCC are still lacking at the present time. To develop a new approach for liver cancer therapy, we designed a chimeric gene (his-HR) encoding a single-chain variable fragment of human HAb25 (hHscFv) fused to a cytotoxic ribonuclease from Rana catesbeiana (RC-RNase) and expressed the corresponding fusion protein in transgenic tobacco (Nicotiana tabacum). Eleven positive transgenic plant lines were identified from 204 regenerated tobacco plants by PCR and Southern blot analysis, and the immunocompetence of the recombinant his-HR protein was confirmed by Western blotting. The expression levels of his-HR protein ranged from 0.75 to 1.99 µg/g in the fresh tobacco leaves. To characterize the bifunction of the expressed his-HR protein in tobacco, binding specificity and cell toxicity to several cell lines were examined by the indirect immunocytochemical streptavidin-biotin complex method and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide assay. Data indicated that the his-HR protein had stronger specific binding affinity to HepG2 (human liver HCC cell line) than to the other tumor cell lines and normal liver cell line, and the capacity to kill the HCC cell lines SMMC7721 and HepG2 with an half maximal inhibiting concentration of 2.0 and 2.4 nM, respectively. The results suggest that recombinant bifunctional his-HR protein derived from transgenic plants may provide a novel strategy to treat HCC in the future.
Collapse
Affiliation(s)
- Lijie Cui
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | | | | | | | | | | |
Collapse
|
77
|
Tomé-Amat J, Menéndez-Méndez A, García-Ortega L, Batt CA, Oñaderra M, Martínez-del-Pozo A, Gavilanes JG, Lacadena J. Production and characterization of scFvA33T1, an immunoRNase targeting colon cancer cells. FEBS J 2012; 279:3022-32. [PMID: 22748038 DOI: 10.1111/j.1742-4658.2012.08683.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Within the last 10 years, the use of different RNases as therapeutic agents for various diseases has been pursued. Furthermore, the advancements of recombinant technology have allowed the assembly of proteins with different functions. In this regard, immunoribonucleases (immunoRNases) stand out as some of the most promising therapeutic candidates given their enzymatic and non-mutagenic character. Accordingly, the work reported here describes fusing RNase T1, one of the most studied members of the microbial RNase family, to the single-chain variable fragment (scFv) of a monoclonal antibody that targets the glycoprotein A33 antigen (GPA33) from human colon cancer cells. A heterologous production system, which employs the yeast Pichia pastoris, has been optimized to produce this immunoRNase (scFvA33T1) with yields of ∼ 5-10 mg · L(-1). The purified protein appears to be correctly folded as it retains its antigen specificity and ribonucleolytic activity. Finally, it also shows specific binding to, internalization into and toxicity against GPA33-positive cell lines compared with the control, GPA33-negative cells. Overall, it can be concluded that scFvA33T1 is a promising therapeutic fusion protein with the additional advantage that presumably it can be produced and purified in large amounts using an easily scalable yeast-based system.
Collapse
Affiliation(s)
- Jaime Tomé-Amat
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Gutschner T, Diederichs S. The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol 2012. [PMID: 22664915 DOI: 10.4161/rna.20481395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
With the advent of next generation sequencing methods and progress in transcriptome analysis, it became obvious that the human genome contains much more than just protein-coding genes. In fact, up to 70% of our genome is transcribed into RNA that does not serve as templates for proteins. In this review, we focus on the emerging roles of these long non-coding RNAs (lncRNAs) in the field of tumor biology. Long ncRNAs were found to be deregulated in several human cancers and show tissue-specific expression. Functional studies revealed a broad spectrum of mechanisms applied by lncRNAs such as HOTAIR, MALAT1, ANRIL or lincRNA-p21 to fulfill their functions. Here, we link the cellular processes influenced by long ncRNAs to the hallmarks of cancer and therefore provide an ncRNA point-of-view on tumor biology. This should stimulate new research directions and therapeutic options considering long ncRNAs as novel prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Tony Gutschner
- Helmholtz-University-Group "Molecular RNA Biology & Cancer", German Cancer Research Center DKFZ, Heidelberg, Germany
| | | |
Collapse
|
79
|
Abstract
With the advent of next generation sequencing methods and progress in transcriptome analysis, it became obvious that the human genome contains much more than just protein-coding genes. In fact, up to 70% of our genome is transcribed into RNA that does not serve as templates for proteins. In this review, we focus on the emerging roles of these long non-coding RNAs (lncRNAs) in the field of tumor biology. Long ncRNAs were found to be deregulated in several human cancers and show tissue-specific expression. Functional studies revealed a broad spectrum of mechanisms applied by lncRNAs such as HOTAIR, MALAT1, ANRIL or lincRNA-p21 to fulfill their functions. Here, we link the cellular processes influenced by long ncRNAs to the hallmarks of cancer and therefore provide an ncRNA point-of-view on tumor biology. This should stimulate new research directions and therapeutic options considering long ncRNAs as novel prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Tony Gutschner
- Helmholtz-University-Group "Molecular RNA Biology & Cancer", German Cancer Research Center DKFZ, Heidelberg, Germany
| | | |
Collapse
|
80
|
Abstract
Mammalian pancreatic-type ribonucleases (ptRNases) comprise an enzyme family that is remarkably well suited for therapeutic exploitation. ptRNases are robust and prodigious catalysts of RNA cleavage that can naturally access the cytosol. Instilling cytotoxic activity requires endowing them with the ability to evade a cytosolic inhibitor protein while retaining other key attributes. These efforts have informed our understanding of ptRNase-based cytotoxins, as well as the action of protein-based drugs with cytosolic targets. Here, we address the most pressing problems encountered in the design of cytotoxic ptRNases, along with potential solutions. In addition, we describe assays that can be used to evaluate a successful design in vitro, in cellulo, and in vivo. The emerging information validates the continuing development of ptRNases as chemotherapeutic agents.
Collapse
Affiliation(s)
- Jo E Lomax
- Graduate Program in Cellular & Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | |
Collapse
|
81
|
Vidalino L, Monti L, Haase A, Moro A, Acquati F, Taramelli R, Macchi P. Intracellular trafficking of RNASET2, a novel component of P-bodies. Biol Cell 2011; 104:13-21. [DOI: 10.1111/boc.201100092] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 10/25/2011] [Indexed: 12/01/2022]
|
82
|
Sundlass NK, Raines RT. Arginine residues are more effective than lysine residues in eliciting the cellular uptake of onconase. Biochemistry 2011; 50:10293-9. [PMID: 21980976 DOI: 10.1021/bi200979k] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Onconase is an amphibian member of the pancreatic ribonuclease family of enzymes that is in clinical trials for the treatment of cancer. Onconase, which has an abundance of lysine residues, is internalized by cancer cells through endocytosis in a mechanism similar to that of cell-penetrating peptides. Here, we compare the effect of lysine versus arginine residues on the biochemical attributes necessary for Onconase to elicit its cytotoxic activity. In the variant R-Onconase, 10 of the 12 lysine residues in Onconase are replaced with arginine, leaving only the two active-site lysines intact. Cytometric assays quantifying internalization showed a 3-fold increase in the internalization of R-Onconase compared with Onconase. R-Onconase also showed greater affinity for heparin and a 2-fold increase in ribonucleolytic activity. Nonetheless, arginine substitution endowed only a slight increase in toxicity toward human cancer cells. Analysis of denaturation induced with guanidine-HCl showed that R-Onconase has less conformational stability than does the wild-type enzyme; moreover, R-Onconase is more susceptible to proteolytic degradation. These data indicate that arginine residues are more effective than lysine in eliciting cellular internalization but can compromise other aspects of protein structure and function.
Collapse
Affiliation(s)
- Nadia K Sundlass
- Medical Scientist Training Program and Biophysics Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | | |
Collapse
|
83
|
Holloway DE, Singh UP, Shogen K, Acharya KR. Crystal structure of Onconase at 1.1 Å resolution--insights into substrate binding and collective motion. FEBS J 2011; 278:4136-49. [PMID: 21895975 PMCID: PMC3397563 DOI: 10.1111/j.1742-4658.2011.08320.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Onconase® (ONC) is an amphibian member of the pancreatic ribonuclease superfamily that is selectively toxic to tumor cells. It is a much less efficient enzyme than the archetypal ribonuclease A and, in an attempt to gain further insight, we report the first atomic resolution crystal structure of ONC, determined in complex with sulfate ions at 100 K. The electron density map is of a quality sufficient to reveal significant nonplanarity in several peptide bonds. The majority of active site residues are very well defined, with the exceptions being Lys31 from the catalytic triad and Lys33 from the B1 subsite, which are relatively mobile but rigidify upon nucleotide binding. Cryocooling causes a compaction of the unit cell and the protein contained within. This is principally the result of an inward movement of one of the lobes of the enzyme (lobe 2), which also narrows the active site cleft. Binding a nucleotide in place of sulfate is associated with an approximately perpendicular movement of lobe 2 and has little further effect on the cleft width. Aspects of this deformation are present in the principal axes of anisotropy extracted from Cα atomic displacement parameters, indicating its intrinsic nature. The three lowest-frequency modes of ONC motion predicted by an anisotropic network model are compaction/expansion variations in which lobe 2 is the prime mover. Two of these have high similarity to the cryocooling response and imply that the essential ‘breathing’ motion of ribonuclease A is conserved in ONC. Instead, shifts in conformational equilibria may contribute to the reduced ribonucleolytic activity of ONC.
Collapse
Affiliation(s)
- Daniel E Holloway
- Department of Biology and Biochemistry, University of Bath, Bath, UK
| | | | | | | |
Collapse
|
84
|
Abstract
RNases are enzymes that cleave RNAs, resulting in remarkably diverse biological consequences. Many RNases are cytotoxic. In some cases, they attack selectively malignant cells triggering an apoptotic response. A number of eukaryotic and bacterial RNase-based strategies are being developed for use in anticancer and antiviral therapy. However, the physiological functions of these RNases are often poorly understood. This review focuses on the properties of the extracellular RNases from Bacillus amyloliquefaciens (barnase) and Bacillus intermedius (binase), the characteristics of their biosynthesis regulation and their physiological role, with an emphasis on the similarities and differences. Barnase and binase can be regarded as molecular twins according to their highly similar structure, physical-chemical and catalytic properties. Nevertheless, the 'life paths' of these enzymes are not the same, as their expression in bacteria is controlled by diverse signals. Binase is predominantly synthesized under phosphate starvation, whereas barnase production is strictly dependent on the multifunctional Spo0A regulator controlling sporulation, biofilm formation and cannibalism. Barnase and binase also have some distinctions in practical applications. Barnase was initially suggested to be useful in research and biotechnology as a tool for studying protein-protein interactions, for RNA elimination from biological samples, for affinity purification of RNase fusion proteins, for the development of cloning vectors and for sterility acquisition by transgenic plants. Binase, as later barnase, was tested for antiviral, antitumour and immunogenic effects. Both RNases have found their own niche in cancer research as a result of success in targeted delivery and selectivity towards tumour cells.
Collapse
Affiliation(s)
- Vera Ulyanova
- Department of Microbiology, Kazan (Volga Region) Federal University, Kazan, Russia
| | | | | |
Collapse
|
85
|
Lawal A, Jejelowo O, Chopra AK, Rosenzweig JA. Ribonucleases and bacterial virulence. Microb Biotechnol 2011; 4:558-71. [PMID: 21375713 PMCID: PMC3123680 DOI: 10.1111/j.1751-7915.2010.00212.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Accepted: 08/12/2010] [Indexed: 12/26/2022] Open
Abstract
Bacterial stress responses provide them the opportunity to survive hostile environments, proliferate and potentially cause diseases in humans and animals. The way in which pathogenic bacteria interact with host immune cells triggers a complicated series of events that include rapid genetic re-programming in response to the various host conditions encountered. Viewed in this light, the bacterial host-cell induced stress response (HCISR) is similar to any other well-characterized environmental stress to which bacteria must respond by upregulating a group of specific stress-responsive genes. Post stress, bacteria must resume their pre-stress genetic program, and, as a consequence, must degrade unnecessary stress responsive transcripts through RNA decay mechanisms. Further, there is a well-established role for several ribonucleases in the cold shock response whereby they modulate the changing transcript landscape in response to the stress, and during acclimation and subsequent genetic re-programming post stress. Recently, ribonucleases have been implicated as virulence-associated factors in several notable Gram-negative pathogens including, the yersiniae, the salmonellae, Helicobacter pylori, Shigella flexneri and Aeromonas hydrophila. This review will focus on the roles played by ribonucleases in bacterial virulence, other bacterial stress responses, and on their novel therapeutic applications.
Collapse
Affiliation(s)
- Abidat Lawal
- Department of Biology, Center for Bionanotechnology and Environmental Research (CBER), Texas Southern University, 3100 Cleburne Street, Houston, TX, USA
| | - Olufisayo Jejelowo
- Department of Biology, Center for Bionanotechnology and Environmental Research (CBER), Texas Southern University, 3100 Cleburne Street, Houston, TX, USA
| | - Ashok K. Chopra
- Department of Microbiology and Immunology, Sealy Center for Vaccine Development, and the Institute of Human Infections and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
| | - Jason A. Rosenzweig
- Department of Biology, Center for Bionanotechnology and Environmental Research (CBER), Texas Southern University, 3100 Cleburne Street, Houston, TX, USA
| |
Collapse
|
86
|
|
87
|
Andrady C, Sharma SK, Chester KA. Antibody-enzyme fusion proteins for cancer therapy. Immunotherapy 2011; 3:193-211. [PMID: 21322759 DOI: 10.2217/imt.10.90] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Advances in biomolecular technology have allowed the development of genetically fused antibody-enzymes. Antibody-enzyme fusion proteins have been used to target tumors for cancer therapy in two ways. In one system, an antibody-enzyme is pretargeted to the tumor followed by administration of an inactive prodrug that is converted to its active form by the pretargeted enzyme. This system has been described as antibody-directed enzyme prodrug therapy. The other system uses antibody-enzyme fusion proteins as direct therapeutics, where the enzyme is toxic in its own right. The key feature in this approach is that the antibody is used to internalize the toxic enzyme into the tumor cell, which activates cell-death processes. This antibody-enzyme system has been largely applied to deliver ribonucleases. This article addresses these two antibody-enzyme targeting strategies for cancer therapy from concept to (pre)clinical trials.
Collapse
Affiliation(s)
- Carima Andrady
- Cancer Research UK Targeting & Imaging Group, Department of Oncology, UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street, London WC1E6BT, UK.
| | | | | |
Collapse
|
88
|
Antosiewicz JM, Shugar D. Poisson–Boltzmann continuum-solvation models: applications to pH-dependent properties of biomolecules. MOLECULAR BIOSYSTEMS 2011; 7:2923-49. [DOI: 10.1039/c1mb05170a] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
89
|
Patutina O, Mironova N, Ryabchikova E, Popova N, Nikolin V, Kaledin V, Vlassov V, Zenkova M. Inhibition of metastasis development by daily administration of ultralow doses of RNase A and DNase I. Biochimie 2010; 93:689-96. [PMID: 21194552 DOI: 10.1016/j.biochi.2010.12.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 12/16/2010] [Indexed: 12/18/2022]
Abstract
Recent data on the involvement of miRNA and circulating tumor-derived DNA in regulation of tumorigenesis showed a great prospect for these molecules as a novel class of therapeutic targets and gave a new start for the study of enzymes cleaving nucleic acids as potential antitumor and antimetastatic agents. In the present paper using two murine tumor models with pulmonary or liver metastases we studied the antimetastatic potential of RNase A and DNase I and performed a search for possible molecular targets of the enzymes. Herein, we show for the first time that daily administration of ultralow doses of RNase A (0.5-50 μg/kg) and DNase I (0.02-2.3 mg/kg) inhibits the development of metastasis to 60-90% and RNase A exerts 30% retardation of tumor growth. Remarkably, the increase in RNase A dose from 50 μg/kg to 10mg/kg leads to a disappearance of antitumor and antimetastatic effects. Simultaneous treatment of tumor-bearing animals with RNase A and DNase I leads to an additive effect and results in almost total absence of metastases. The use of RNase A as an adjuvant in conjunction with conventional cytostatic cyclophosphamide results in a reliable enhancement of antitumor and antimetastatic effect of the therapy compared with the use of these agents individually. The search for possible molecular mechanism of antimetastatic effect of nucleases showed that daily administration of the enzymes reduced the pathologically increased level of extracellular nucleic acids and increased nuclease activity of the blood plasma of tumor-bearing mice back to the level of healthy animals. Thus, we unequivocally show that the proposed protocol of treatment of tumor-bearing animals with RNase A and DNase I has a general systemic and immunomodulatory effect, leads to a drastic suppression of metastasis development, and in perspective may become an effective component of intensive complex therapy of cancer.
Collapse
Affiliation(s)
- Olga Patutina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russian Federation
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Meineke B, Schwer B, Schaffrath R, Shuman S. Determinants of eukaryal cell killing by the bacterial ribotoxin PrrC. Nucleic Acids Res 2010; 39:687-700. [PMID: 20855293 PMCID: PMC3025547 DOI: 10.1093/nar/gkq831] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
tRNA damage inflicted by the Escherichia coli anticodon nuclease PrrC (EcoPrrC) underlies an antiviral response to phage T4 infection. PrrC homologs are present in many bacterial proteomes, though their biological activities are uncharted. PrrCs consist of two domains: an N-terminal NTPase module related to the ABC family and a distinctive C-terminal ribonuclease module. In this article, we report that the expression of EcoPrrC in budding yeast is fungicidal, signifying that PrrC is toxic in a eukaryon in the absence of other bacterial or viral proteins. Whereas Streptococcus PrrC is also toxic in yeast, Neisseria and Xanthomonas PrrCs are not. Via analysis of the effects of 118 mutations on EcoPrrC toxicity in yeast, we identified 22 essential residues in the NTPase domain and 11 in the nuclease domain. Overexpressing PrrCs with mutations in the NTPase active site ameliorated the toxicity of wild-type EcoPrrC. Our findings support a model in which EcoPrrC toxicity is contingent on head-to-tail dimerization of the NTPase domains to form two composite NTP phosphohydrolase sites. Comparisons of EcoPrrC activity in a variety of yeast genetic backgrounds, and the rescuing effects of tRNA overexpression, implicate tRNALys(UUU) as a target of EcoPrrC toxicity in yeast.
Collapse
Affiliation(s)
- Birthe Meineke
- Molecular Biology Program, Sloan-Kettering Institute, Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065 USA and Department of Genetics, University of Leicester, Leicester, LE1 7RH, UK
| | - Beate Schwer
- Molecular Biology Program, Sloan-Kettering Institute, Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065 USA and Department of Genetics, University of Leicester, Leicester, LE1 7RH, UK
| | - Raffael Schaffrath
- Molecular Biology Program, Sloan-Kettering Institute, Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065 USA and Department of Genetics, University of Leicester, Leicester, LE1 7RH, UK
| | - Stewart Shuman
- Molecular Biology Program, Sloan-Kettering Institute, Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065 USA and Department of Genetics, University of Leicester, Leicester, LE1 7RH, UK
- *To whom correspondence should be addressed. Tel: +1 212 639 7145; Fax: +1 212 717 3623;
| |
Collapse
|
91
|
Fang EF, Ng TB. Ribonucleases of different origins with a wide spectrum of medicinal applications. Biochim Biophys Acta Rev Cancer 2010; 1815:65-74. [PMID: 20843477 DOI: 10.1016/j.bbcan.2010.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Revised: 09/03/2010] [Accepted: 09/03/2010] [Indexed: 12/18/2022]
Abstract
Ribonucleases (RNases) are a type of nucleases that catalyze the degradation of RNA into smaller components. They exist in a wide range of life forms from prokaryotes to eukaryotes. RNase-controlled RNA degradation is a determining factor in the control of gene expression, maturation and turnover, which are further associated with the progression of cancers and infectious diseases. Over the years, RNases purified from multiple origins have drawn increasing attention from medical scientists due to their remarkable antitumor properties. In this review, we present a brief summary of the representative RNases of fungal, bacterial, plant, and animal origins and outline their potential medicinal value in the treatment of tumor and AIDS. Among them, the most clinically promising RNases are mushroom RNases, Binase and Barnase from bacteria, ginseng RNases, and Onconase from frog (Rana pipiens). Fast developing protein engineering of RNases, which display more potent cytotoxic activity on and greater selectivity for malignant cells, has also aroused the interest of researchers. The multiple anti-cancer mechanisms of RNases are also included. To sum up, these inspiring studies unveil a new perspective for RNases as potential therapeutic agents.
Collapse
Affiliation(s)
- Evandro Fei Fang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | |
Collapse
|
92
|
Los M. New, exciting developments in experimental therapies in the early 21st century. Eur J Pharmacol 2009; 625:1-5. [DOI: 10.1016/j.ejphar.2009.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 10/08/2009] [Accepted: 10/08/2009] [Indexed: 12/15/2022]
|