1
|
Costa MDN, Silva TA, Guimarães DSPSF, Ricci-Azevedo R, Teixeira FR, Silveira LR, Gomes MD, Faça VM, de Oliveira EB, Calado RT, Silva RN. The recombinant L-lysine α-oxidase from the fungus Trichoderma harzianum promotes apoptosis and necrosis of leukemia CD34 + hematopoietic cells. Microb Cell Fact 2024; 23:51. [PMID: 38355518 PMCID: PMC10865671 DOI: 10.1186/s12934-024-02315-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND In hematologic cancers, including leukemia, cells depend on amino acids for rapid growth. Anti-metabolites that prevent their synthesis or promote their degradation are considered potential cancer treatment agents. Amino acid deprivation triggers proliferation inhibition, autophagy, and programmed cell death. L-lysine, an essential amino acid, is required for tumor growth and has been investigated for its potential as a target for cancer treatment. L-lysine α-oxidase, a flavoenzyme that degrades L-lysine, has been studied for its ability to induce apoptosis and prevent cancer cell proliferation. In this study, we describe the use of L-lysine α-oxidase (LO) from the filamentous fungus Trichoderma harzianum for cancer treatment. RESULTS The study identified and characterized a novel LO from T. harzianum and demonstrated that the recombinant protein (rLO) has potent and selective cytotoxic effects on leukemic cells by triggering the apoptotic cascade through mitochondrial dysfunction. CONCLUSIONS The results support future translational studies using the recombinant LO as a potential drug for the treatment of leukemia.
Collapse
Affiliation(s)
- Mariana do Nascimento Costa
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Thiago Aparecido Silva
- Department of Cell Biology and Molecular and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Clinical Analysis, School of Pharmaceutical Sciences in Araraquara, Sao Paulo State University, Araraquara, SP, Brazil
| | | | - Rafael Ricci-Azevedo
- Department of Cell Biology and Molecular and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Felipe Roberti Teixeira
- Department of Genetics and Evolution, Center of Biological and Health Sciences, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Leonardo Reis Silveira
- Obesity and Comorbidities Research Center, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Marcelo Damário Gomes
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Vítor Marcel Faça
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Eduardo Brandt de Oliveira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rodrigo T Calado
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Roberto N Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
2
|
Lv Y, Yang X, Song Y, Yang D, Zheng K, Zhou S, Xie H, Guo R, Tang S. The Correlation Between Essential Amino Acid Tryptophan, Lysine, Phenylalanine and Chemotherapy of Breast Cancer. Technol Cancer Res Treat 2024; 23:15330338241286872. [PMID: 39435510 PMCID: PMC11497521 DOI: 10.1177/15330338241286872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/28/2024] [Accepted: 08/09/2024] [Indexed: 10/23/2024] Open
Abstract
To investigate the differences in serum tryptophan, lysine, and phenylalanine levels in breast cancer patients, the correlation between the three amino acids with the chemotherapy regimen, and their significance in the clinical diagnosis and treatment of breast cancer.Clinical data were collected from the Department of Breast Surgery at Yunnan Cancer Hospital, encompassing 216 cases from July to December 2020, including 91 healthy individuals, 38 with benign tumors, and 87 with cancer. Amino acid levels were measured using liquid chromatography-tandem mass spectrometry. Statistical analyses, such as the Kruskal-Wallis H-test and Wilcoxon test, were conducted to compare the levels of these amino acids across the healthy group, benign tumor group, and breast cancer group. The χ2 test and Fisher's exact probability method were employed to assess the relationship between amino acid levels and breast cancer stage, grade, and chemotherapy regimen.The results indicated that there were significant differences in serum lysine (H = 36.13, P < .001) and phenylalanine (H = 34.03, P < .001) levels among the three groups. However, tryptophan levels did not show statistically significant variances. Specifically, lysine and phenylalanine levels were significantly different when comparing the healthy group with the breast cancer group and the benign tumor group with the breast cancer group. These differences were not significant when comparing the healthy group with the benign tumor group. Furthermore, there were no statistically significant distinctions observed in lysine (F = 0.836, P > .05) and phenylalanine (F = 1.466, P > .05) levels across different conventional chemotherapy regimens among the breast cancer cases studied.Serum lysine and phenylalanine levels might serve as potential biomarkers for breast cancer, and the choice of chemotherapy regimen is unlikely to impact significant changes in these amino acid levels.
Collapse
Affiliation(s)
- Yafeng Lv
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Xuan Yang
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Ying Song
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Dechun Yang
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Kai Zheng
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Shaoqiang Zhou
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Hanhui Xie
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Rong Guo
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Shicong Tang
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| |
Collapse
|
3
|
Pokrovsky VS, Qoura LA, Demidova EA, Han Q, Hoffman RM. Targeting Methionine Addiction of Cancer Cells with Methioninase. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:944-952. [PMID: 37751865 DOI: 10.1134/s0006297923070076] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 09/28/2023]
Abstract
All types of cancer cells are addicted to methionine, which is known as the Hoffman effect. Restricting methionine inhibits the growth and proliferation of all tested types of cancer cells, leaving normal cells unaffected. Targeting methionine addiction with methioninase (METase), either alone or in combination with common cancer chemotherapy drugs, has been shown as an effective and safe therapy in various types of cancer cells and animal cancer models. About six years ago, recombinant METase (rMETase) was found to be able to be taken orally as a supplement, resulting in anecdotal positive results in patients with advanced cancer. Currently, there are 8 published clinical studies on METase, including two from the 1990s and six more recent ones. This review focuses on the results of clinical studies on METase-mediated methionine restriction, in particular, on the dosage of oral rMETase taken alone as a supplement or in combination with common chemotherapeutic agents in patients with advanced cancer.
Collapse
Affiliation(s)
- Vadim S Pokrovsky
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia.
- Research Institute of Molecular and Cellular Medicine, People's Friendship University of Russia (RUDN University), Moscow, 117198, Russia
- Department of Biotechnology, Sirius University of Science and Technology, Sochi, 354340, Russia
| | - Louay Abo Qoura
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia.
- Research Institute of Molecular and Cellular Medicine, People's Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Elena A Demidova
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | | | - Robert M Hoffman
- AntiCancer Inc., San Diego, CA 92111, USA.
- Department of Surgery, University of California, San Diego, La Jolla, CA 92037-7400, USA
| |
Collapse
|
4
|
Hababag EAC, Cauilan A, Quintero D, Bermudes D. Tryptophanase Expressed by Salmonella Halts Breast Cancer Cell Growth In Vitro and Inhibits Production of Immunosuppressive Kynurenine. Microorganisms 2023; 11:1355. [PMID: 37317329 DOI: 10.3390/microorganisms11051355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 06/16/2023] Open
Abstract
Tryptophan is an essential amino acid required for tumor cell growth and is also the precursor to kynurenine, an immunosuppressive molecule that plays a role in limiting anticancer immunity. Tryptophanase (TNase) is an enzyme expressed by different bacterial species that converts tryptophan into indole, pyruvate and ammonia, but is absent in the Salmonella strain VNP20009 that has been used as a therapeutic delivery vector. We cloned the Escherichia coli TNase operon tnaCAB into the VNP20009 (VNP20009-tnaCAB), and were able to detect linear production of indole over time, using Kovács reagent. In order to conduct further experiments using the whole bacteria, we added the antibiotic gentamicin to stop bacterial replication. Using a fixed number of bacteria, we found that there was no significant effect of gentamicin on stationary phase VNP20009-tnaCAB upon their ability to convert tryptophan to indole over time. We developed a procedure to extract indole from media while retaining tryptophan, and were able to measure tryptophan spectrophotometrically after exposure to gentamicin-inactivated whole bacterial cells. Using the tryptophan concentration equivalent to that present in DMEM cell culture media, a fixed number of bacteria were able to deplete 93.9% of the tryptophan in the culture media in 4 h. In VNP20009-tnaCAB depleted tissue culture media, MDA-MB-468 triple negative breast cancer cells were unable to divide, while those treated with media exposed only to VNP20009 continued cell division. Re-addition of tryptophan to conditioned culture media restored tumor cell growth. Treatment of tumor cells with molar equivalents of the TNase products indole, pyruvate and ammonia only caused a slight increase in tumor cell growth. Using an ELISA assay, we confirmed that TNase depletion of tryptophan also limits the production of immunosuppressive kynurenine in IFNγ-stimulated MDA-MB-468 cancer cells. Our results demonstrate that Salmonella VNP20009 expressing TNase has improved potential to stop tumor cell growth and reverse immunosuppression.
Collapse
Affiliation(s)
| | - Allea Cauilan
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
| | - David Quintero
- Los Angeles Medical Facility, Los Angeles, CA 90027, USA
| | - David Bermudes
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
| |
Collapse
|
5
|
Kaur N, Popli P, Tiwary N, Swami R. Small molecules as cancer targeting ligands: Shifting the paradigm. J Control Release 2023; 355:417-433. [PMID: 36754149 DOI: 10.1016/j.jconrel.2023.01.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 02/10/2023]
Abstract
Conventional chemotherapeutics exploration is hampered due to their nonspecific distribution leading to unintended serious toxicity. Toxicity is so severe that deciding to go for chemotherapy becomes a question of concern for many terminally ill cancer patients. However, with evolving times nanotechnology assisted in reducing the haywire distribution and channelizing the movement of drug-enclosing drug delivery systems to cancer cells to a greater extent, yet toxicity issues still could not be obliterated. Thus, active targeting appeared as a refuge, where ligands actively or specifically deliver linked chemotherapeutics and carriers to cancer cells. For a very long time, large molecule weight/macromolecular ligands (peptides and big polymers) were considered the first choice for ligand-directed active cancer targeting, due to their specificity towards overexpressed native cancer receptors. However, complex characterization, instability, and the expensive nature demanded to reconnoitre better alternatives for macromolecule ligands. The concept of small molecules as ligands emerged from the idea that few chemical molecules including chemotherapeutics have a higher affinity for cancer receptors, which are overexpressed on cell membranes, and may have the ability to assist in drug cellular uptake through endocytosis. But now the question is, can they assist the conjugated macro cargos to enter the cell or not? This present review will provide a holistic overview of the small molecule ligands explored till now.
Collapse
Affiliation(s)
- Navjeet Kaur
- Department of Physics, Mata Gujri College, Fatehgarh Sahib, Punjab, India
| | - Pankaj Popli
- Maharishi Markandeshwar College of Pharmacy, Maharishi Markandeshwar University, Mullana, India
| | - Neha Tiwary
- Maharishi Markandeshwar College of Pharmacy, Maharishi Markandeshwar University, Mullana, India
| | - Rajan Swami
- Chitkara College of Pharmacy, Chikara University, Punjab, India.
| |
Collapse
|
6
|
Abo Qoura L, Morozova E, Kulikova V, Karshieva S, Sokolova D, Koval V, Revtovich S, Demidkina T, Pokrovsky VS. Methionine γ-Lyase-Daidzein in Combination with S-Propyl-L-cysteine Sulfoxide as a Targeted Prodrug Enzyme System for Malignant Solid Tumor Xenografts. Int J Mol Sci 2022; 23:ijms231912048. [PMID: 36233347 PMCID: PMC9569779 DOI: 10.3390/ijms231912048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/10/2022] [Accepted: 10/06/2022] [Indexed: 12/01/2022] Open
Abstract
The purpose of this study was to determine the anticancer effect of dipropyl thiosulfinate produced in situ by the pharmacological pair: (1) conjugated with daidzein C115H methionine γ-lyase (EC 4.4.1.11, C115H MGL-Dz) and (2) the substrate, S-propyl-L-cysteine sulfoxide (propiin) against various solid tumor types in vitro and in vivo. The MTT test was used to calculate IC50 values for HT29, COLO205 and HCT116 (colon cancer); Panc1 and MIA-PaCa2 (pancreatic cancer); and 22Rv1, DU-145 and PC3 (prostate cancer). The most promising effect for colon cancer cells in vitro was observed in HT29 (IC50 = 6.9 µM). The IC50 values for MIA-PaCa2 and Panc1 were 3.4 and 3.8 µM, respectively. Among prostate cancer cells, 22Rv1 was the most sensitive (IC50 = 5.4 µM). In vivo antitumor activity of the pharmacological pair was studied in HT29, SW620, Panc1, MIA-PaCa2 and 22Rv1 subcutaneous xenografts in BALB/c nude mice. The application of C115H MGL-Dz /propiin demonstrated a significant reduction in the tumor volume of Panc1 (TGI 67%; p = 0.004), MIA-PaCa2 (TGI 50%; p = 0.011), HT29 (TGI 51%; p = 0.04) and 22Rv1 (TGI 70%; p = 0.043) xenografts. The results suggest that the combination of C115H MGL-Dz/propiin is able to suppress tumor growth in vitro and in vivo and the use of this pharmacological pair can be considered as a new strategy for the treatment of solid tumors.
Collapse
Affiliation(s)
- Louay Abo Qoura
- Department of Biochemistry, RUDN University, 117198 Moscow, Russia
| | - Elena Morozova
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
- Correspondence: (E.M.); (V.S.P.); Tel.: +7-915-143-03-91 (V.S.P.)
| | - Vitalia Kulikova
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Saida Karshieva
- Laboratory of Combined Treatment, N.N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, 115478 Moscow, Russia
| | - Darina Sokolova
- Department of Biochemistry, RUDN University, 117198 Moscow, Russia
- Laboratory of Combined Treatment, N.N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, 115478 Moscow, Russia
| | - Vasiliy Koval
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Svetlana Revtovich
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Tatyana Demidkina
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vadim S. Pokrovsky
- Department of Biochemistry, RUDN University, 117198 Moscow, Russia
- Laboratory of Combined Treatment, N.N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, 115478 Moscow, Russia
- Department of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia
- Correspondence: (E.M.); (V.S.P.); Tel.: +7-915-143-03-91 (V.S.P.)
| |
Collapse
|
7
|
Xia X, Lin Q, Zhao N, Zeng J, Yang J, Liu Z, Huang R. Anti-Colon Cancer Activity of Dietary Phytochemical Soyasaponin I and the Induction of Metabolic Shifts in HCT116. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27144382. [PMID: 35889255 PMCID: PMC9316303 DOI: 10.3390/molecules27144382] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 06/26/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022]
Abstract
Dietary phytochemicals play an important role in the prevention and treatment of colon cancer. It is reported that group B of soyasaponin, derived from dietary pulses, has anti-colonic effects on some colon cancer cell lines. However, it is uncertain which specific soybean saponins play a role. In our study, as one of the group B soyasaponin, the anti-colon cancer activity of soyasaponins I (SsI) was screened, and we found that it had the inhibitory effect of proliferation on colon cancer cell lines HCT116 (IC50 = 161.4 μM) and LoVo (IC50 = 180.5 μM), but no effect on HT29 between 0–200 μM. Then, nine potential targets of SsI on colon cancer were obtained by network pharmacology analysis. A total of 45 differential metabolites were identified by metabolomics analysis, and the KEGG pathway was mainly enriched in the pathways related to the absorption and metabolism of amino acids. Finally, molecular docking analysis predicted that SsI might dock with the protein of DNMT1, ERK1. The results indicated that the effect of SsI on HCT116 might be exerted by influencing amino acid metabolism and the estrogen signaling pathway. This study may provide the possibility for the application of SsI against colon cancer.
Collapse
Affiliation(s)
- Xuewei Xia
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (Q.L.); (J.Z.); (J.Y.); (Z.L.)
| | - Qianmin Lin
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (Q.L.); (J.Z.); (J.Y.); (Z.L.)
| | - Ning Zhao
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou 510006, China;
| | - Jinzi Zeng
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (Q.L.); (J.Z.); (J.Y.); (Z.L.)
| | - Jiajia Yang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (Q.L.); (J.Z.); (J.Y.); (Z.L.)
| | - Zhiyuan Liu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (Q.L.); (J.Z.); (J.Y.); (Z.L.)
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China; (X.X.); (Q.L.); (J.Z.); (J.Y.); (Z.L.)
- Correspondence:
| |
Collapse
|
8
|
Alexandrova SS, Gladilina YA, Pokrovskaya MV, Sokolov NN, Zhdanov DD. [Mechanisms of development of side effects and drug resistance to asparaginase and ways to overcome them]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:104-116. [PMID: 35485484 DOI: 10.18097/pbmc20226802104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Asparaginase is one of the most important chemotherapeutic agents against acute lymphoblastic leukemia, the most common form of blood cancer. To date, both asparaginases from E. coli and Dickeya dadantii (formerly known as Erwinia chrysanthemi), used in hematology, induce chemoresistance in cancer cells and side effects in the form of hypersensitivity of immune reactions. Leukemic cells may be resistant to asparaginase due to the increased activity of asparagine synthetase and other mechanisms associated with resistance to asparaginase. Therefore, the search for new sources of L-asparaginases with improved pharmacological properties remains a promising and prospective study. This article discusses the mechanisms of development of resistance and drug resistance to L-asparaginase, as well as possible ways to overcome them.
Collapse
Affiliation(s)
| | | | | | - N N Sokolov
- Institute of Biomedical Chemistry, Moscow, Russia
| | - D D Zhdanov
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
9
|
Pokrovskaya MV, Pokrovsky VS, Aleksandrova SS, Sokolov NN, Zhdanov DD. Molecular Analysis of L-Asparaginases for Clarification of the Mechanism of Action and Optimization of Pharmacological Functions. Pharmaceutics 2022; 14:pharmaceutics14030599. [PMID: 35335974 PMCID: PMC8948990 DOI: 10.3390/pharmaceutics14030599] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 12/19/2022] Open
Abstract
L-asparaginases (EC 3.5.1.1) are a family of enzymes that catalyze the hydrolysis of L-asparagine to L-aspartic acid and ammonia. These proteins with different biochemical, physicochemical and pharmacological properties are found in many organisms, including bacteria, fungi, algae, plants and mammals. To date, asparaginases from E. coli and Dickeya dadantii (formerly known as Erwinia chrysanthemi) are widely used in hematology for the treatment of lymphoblastic leukemias. However, their medical use is limited by side effects associated with the ability of these enzymes to hydrolyze L-glutamine, as well as the development of immune reactions. To solve these issues, gene-editing methods to introduce amino-acid substitutions of the enzyme are implemented. In this review, we focused on molecular analysis of the mechanism of enzyme action and to optimize the antitumor activity.
Collapse
Affiliation(s)
- Marina V. Pokrovskaya
- Institute of Biomedical Chemistry, Pogodinskaya Str. 10/8, 119121 Moscow, Russia; (M.V.P.); (S.S.A.); (N.N.S.)
| | - Vadim S. Pokrovsky
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya Str. 6, 117198 Moscow, Russia;
- Laboratory of Combined Treatment, N.N. Blokhin Cancer Research Center, Kashirskoe Shosse 24, 115478 Moscow, Russia
- Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Olimpiisky Prospect 1, 354340 Sochi, Russia
| | - Svetlana S. Aleksandrova
- Institute of Biomedical Chemistry, Pogodinskaya Str. 10/8, 119121 Moscow, Russia; (M.V.P.); (S.S.A.); (N.N.S.)
| | - Nikolay N. Sokolov
- Institute of Biomedical Chemistry, Pogodinskaya Str. 10/8, 119121 Moscow, Russia; (M.V.P.); (S.S.A.); (N.N.S.)
| | - Dmitry D. Zhdanov
- Institute of Biomedical Chemistry, Pogodinskaya Str. 10/8, 119121 Moscow, Russia; (M.V.P.); (S.S.A.); (N.N.S.)
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya Str. 6, 117198 Moscow, Russia;
- Correspondence:
| |
Collapse
|
10
|
Fan K, Liu Z, Gao M, Tu K, Xu Q, Zhang Y. Targeting Nutrient Dependency in Cancer Treatment. Front Oncol 2022; 12:820173. [PMID: 35178349 PMCID: PMC8846368 DOI: 10.3389/fonc.2022.820173] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Metabolic reprogramming is one of the hallmarks of tumor. Growing evidence suggests metabolic changes that support oncogenic progression may cause selective vulnerabilities that can be exploited for cancer treatment. Increasing demands for certain nutrients under genetic determination or environmental challenge enhance dependency of tumor cells on specific nutrient, which could be therapeutically developed through targeting such nutrient dependency. Various nutrients including several amino acids and glucose have been found to induce dependency in genetic alteration- or context-dependent manners. In this review, we discuss the extensively studied nutrient dependency and the biological mechanisms behind such vulnerabilities. Besides, existing applications and strategies to target nutrient dependency in different cancer types, accompanied with remaining challenges to further exploit these metabolic vulnerabilities to improve cancer therapies, are reviewed.
Collapse
Affiliation(s)
- Kexin Fan
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zhan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Second Medical College, Karamay, China
| | - Min Gao
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China.,Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
| | - Yilei Zhang
- The Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
11
|
Lukasheva EV, Babayeva G, Karshieva SS, Zhdanov DD, Pokrovsky VS. L-Lysine α-Oxidase: Enzyme with Anticancer Properties. Pharmaceuticals (Basel) 2021; 14:1070. [PMID: 34832852 PMCID: PMC8618108 DOI: 10.3390/ph14111070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/19/2022] Open
Abstract
L-lysine α-oxidase (LO), one of L-amino acid oxidases, deaminates L-lysine with the yield of H2O2, ammonia, and α-keto-ε-aminocaproate. Multiple in vitro and in vivo studies have reported cytotoxic, antitumor, antimetastatic, and antitumor activity of LO. Unlike asparaginase, LO has a dual mechanism of action: depletion of L-lysine and formation of H2O2, both targeting tumor growth. Prominent results were obtained on murine and human tumor models, including human colon cancer xenografts HCT 116, LS174T, and T47D with maximum T/C 12, 37, and 36%, respectively. The data obtained from human cancer xenografts in immunodeficient mice confirm the potential of LO as an agent for colon cancer treatment. In this review, we discuss recently discovered molecular mechanisms of biological action and the potential of LO as anticancer enzyme.
Collapse
Affiliation(s)
- Elena V. Lukasheva
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), Miklukho—Maklaya Street 6, 117198 Moscow, Russia; (E.V.L.); (G.B.)
| | - Gulalek Babayeva
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), Miklukho—Maklaya Street 6, 117198 Moscow, Russia; (E.V.L.); (G.B.)
- Laboratory of Combined Treatment, N.N. Blokhin Cancer Research Center, Kashirskoe Shosse 24, 115478 Moscow, Russia;
| | - Saida Sh. Karshieva
- Laboratory of Combined Treatment, N.N. Blokhin Cancer Research Center, Kashirskoe Shosse 24, 115478 Moscow, Russia;
| | - Dmitry D. Zhdanov
- Institute of Biomedical Chemistry, Pogodinskaya Street 10/8, 119121 Moscow, Russia;
| | - Vadim S. Pokrovsky
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), Miklukho—Maklaya Street 6, 117198 Moscow, Russia; (E.V.L.); (G.B.)
- Laboratory of Combined Treatment, N.N. Blokhin Cancer Research Center, Kashirskoe Shosse 24, 115478 Moscow, Russia;
- Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, 1 Olimpiisky Prospect, 354340 Sochi, Russia
| |
Collapse
|
12
|
Cao W, Jin M, Yang K, Chen B, Xiong M, Li X, Cao G. Fenton/Fenton-like metal-based nanomaterials combine with oxidase for synergistic tumor therapy. J Nanobiotechnology 2021; 19:325. [PMID: 34656118 PMCID: PMC8520258 DOI: 10.1186/s12951-021-01074-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023] Open
Abstract
Chemodynamic therapy (CDT) catalyzed by transition metal and starvation therapy catalyzed by intracellular metabolite oxidases are both classic tumor treatments based on nanocatalysts. CDT monotherapy has limitations including low catalytic efficiency of metal ions and insufficient endogenous hydrogen peroxide (H2O2). Also, single starvation therapy shows limited ability on resisting tumors. The “metal-oxidase” cascade catalytic system is to introduce intracellular metabolite oxidases into the metal-based nanoplatform, which perfectly solves the shortcomings of the above-mentioned monotherapiesIn this system, oxidases can not only consume tumor nutrients to produce a “starvation effect”, but also provide CDT with sufficient H2O2 and a suitable acidic environment, which further promote synergy between CDT and starvation therapy, leading to enhanced antitumor effects. More importantly, the “metal-oxidase” system can be combined with other antitumor therapies (such as photothermal therapy, hypoxia-activated drug therapy, chemotherapy, and immunotherapy) to maximize their antitumor effects. In addition, both metal-based nanoparticles and oxidases can activate tumor immunity through multiple pathways, so the combination of the “metal-oxidase” system with immunotherapy has a powerful synergistic effect. This article firstly introduced the metals which induce CDT and the oxidases which induce starvation therapy and then described the “metal-oxidase” cascade catalytic system in detail. Moreover, we highlight the application of the “metal-oxidase” system in combination with numerous antitumor therapies, especially in combination with immunotherapy, expecting to provide new ideas for tumor treatment.
Collapse
Affiliation(s)
- Wei Cao
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Mengyao Jin
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Kang Yang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Bo Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Maoming Xiong
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Xiang Li
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China.
| | - Guodong Cao
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| |
Collapse
|
13
|
Cioni P, Gabellieri E, Campanini B, Bettati S, Raboni S. Use of Exogenous Enzymes in Human Therapy: Approved Drugs and Potential Applications. Curr Med Chem 2021; 29:411-452. [PMID: 34259137 DOI: 10.2174/0929867328666210713094722] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 11/22/2022]
Abstract
The development of safe and efficacious enzyme-based human therapies has increased greatly in the last decades, thanks to remarkable advances in the understanding of the molecular mechanisms responsible for different diseases, and the characterization of the catalytic activity of relevant exogenous enzymes that may play a remedial effect in the treatment of such pathologies. Several enzyme-based biotherapeutics have been approved by FDA (the U.S. Food and Drug Administration) and EMA (the European Medicines Agency) and many are undergoing clinical trials. Apart from enzyme replacement therapy in human genetic diseases, which is not discussed in this review, approved enzymes for human therapy find applications in several fields, from cancer therapy to thrombolysis and the treatment, e.g., of clotting disorders, cystic fibrosis, lactose intolerance and collagen-based disorders. The majority of therapeutic enzymes are of microbial origin, the most convenient source due to fast, simple and cost-effective production and manipulation. The use of microbial recombinant enzymes has broadened prospects for human therapy but some hurdles such as high immunogenicity, protein instability, short half-life and low substrate affinity, still need to be tackled. Alternative sources of enzymes, with reduced side effects and improved activity, as well as genetic modification of the enzymes and novel delivery systems are constantly searched. Chemical modification strategies, targeted- and/or nanocarrier-mediated delivery, directed evolution and site-specific mutagenesis, fusion proteins generated by genetic manipulation are the most explored tools to reduce toxicity and improve bioavailability and cellular targeting. This review provides a description of exogenous enzymes that are presently employed for the therapeutic management of human diseases with their current FDA/EMA-approved status, along with those already experimented at the clinical level and potential promising candidates.
Collapse
Affiliation(s)
- Patrizia Cioni
- Institute of Biophysics, National Research Council, Via Moruzzi 1, 56124 Pisa. Italy
| | - Edi Gabellieri
- Institute of Biophysics, National Research Council, Via Moruzzi 1, 56124 Pisa. Italy
| | - Barbara Campanini
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 23/A, 43124 Parma. Italy
| | - Stefano Bettati
- Institute of Biophysics, National Research Council, Via Moruzzi 1, 56124 Pisa. Italy
| | - Samanta Raboni
- Institute of Biophysics, National Research Council, Via Moruzzi 1, 56124 Pisa. Italy
| |
Collapse
|
14
|
Wang Z, Xie Q, Zhou H, Zhang M, Shen J, Ju D. Amino Acid Degrading Enzymes and Autophagy in Cancer Therapy. Front Pharmacol 2021; 11:582587. [PMID: 33510635 PMCID: PMC7836011 DOI: 10.3389/fphar.2020.582587] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/20/2020] [Indexed: 12/27/2022] Open
Abstract
Recently, there has been renewed interest in metabolic therapy for cancer, particularly in amino acid deprivation by enzymes. L-asparaginase was approved for the treatment of acute lymphoblastic leukemia by the U.S. Food and Drug Administration. Arginine deiminase and recombinant human arginase have been developed into clinical trials as potential cancer therapeutic agents for the treatment of arginine-auxotrophic tumors. Moreover, other novel amino acid degrading enzymes, such as glutaminase, methionase, lysine oxidase, phenylalanine ammonia lyase, have been developed for the treatment of malignant cancers. One of the greatest obstacles faced by anticancer drugs is the development of drug resistance, which is reported to be associated with autophagy. Autophagy is an evolutionarily conserved catabolic process that is responsible for the degradation of dysfunctional proteins and organelles. There is a growing body of literature revealing that, in response to metabolism stress, autophagy could be induced by amino acid deprivation. The manipulation of autophagy in combination with amino acid degrading enzymes is actively being investigated as a potential therapeutic approach in preclinical studies. Importantly, shedding light on how autophagy fuels tumor metabolism during amino acid deprivation will enable more potential combinational therapeutic strategies. This study summarizes recent advances, discussing several potential anticancer enzymes, and highlighting the promising combined therapeutic strategy of amino acid degrading enzymes and autophagy modulators in tumors
Collapse
Affiliation(s)
- Ziyu Wang
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Qinghong Xie
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China
| | - Haifeng Zhou
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, China
| | - Min Zhang
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, China
| | - Jie Shen
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China
| |
Collapse
|
15
|
Vachher M, Sen A, Kapila R, Nigam A. Microbial therapeutic enzymes: A promising area of biopharmaceuticals. CURRENT RESEARCH IN BIOTECHNOLOGY 2021. [DOI: 10.1016/j.crbiot.2021.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
16
|
Plyasova AA, Pokrovskaya MV, Lisitsyna OM, Pokrovsky VS, Alexandrova SS, Hilal A, Sokolov NN, Zhdanov DD. Penetration into Cancer Cells via Clathrin-Dependent Mechanism Allows L-Asparaginase from Rhodospirillum rubrum to Inhibit Telomerase. Pharmaceuticals (Basel) 2020; 13:E286. [PMID: 33008089 PMCID: PMC7650658 DOI: 10.3390/ph13100286] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 01/19/2023] Open
Abstract
The anticancer effect of L-asparaginases (L-ASNases) is attributable to their ability to hydrolyze L-asparagine in the bloodstream and cancer cell microenvironment. Rhodospirillum rubrum (RrA) has dual mechanism of action and plays a role in the suppression of telomerase activity. The aim of this work was to investigate the possible mechanism of RrA penetration into human cancer cells. Labeling of widely used L-ASNases by fluorescein isothiocyanate followed by flow cytometry and fluorescent microscopy demonstrated that only RrA can interact with cell membranes. The screening of inhibitors of receptor-mediated endocytosis demonstrated the involvement of clathrin receptors in RrA penetration into cells. Confocal microscopy confirmed the cytoplasmic and nuclear localization of RrA in human breast cancer SKBR3 cells. Two predicted nuclear localization motifs allow RrA to penetrate into the cell nucleus and inhibit telomerase. Chromatin relaxation promoted by different agents can increase the ability of RrA to suppress the expression of telomerase main catalytic subunit. Our study demonstrated for the first time the ability of RrA to penetrate into human cancer cells and the involvement of clathrin receptors in this process.
Collapse
Affiliation(s)
- Anna A. Plyasova
- Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia; (A.A.P.); (M.V.P.); (S.S.A.); (A.H.); (N.N.S.)
| | - Marina V. Pokrovskaya
- Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia; (A.A.P.); (M.V.P.); (S.S.A.); (A.H.); (N.N.S.)
| | - Olga M. Lisitsyna
- International Biotechnology Center “Generium” LLC, Vladimirskaya st. 14, 601125 Volginsky, Russia;
| | - Vadim S. Pokrovsky
- N.N. Blokhin Cancer Research Center, Kashirskoe Shosse 24, 115478 Moscow, Russia;
- Department of Biochemistry, Рeoples Friendship University of Russia (RUDN University), Miklukho-Maklaya st. 6, 117198 Moscow, Russia
| | - Svetlana S. Alexandrova
- Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia; (A.A.P.); (M.V.P.); (S.S.A.); (A.H.); (N.N.S.)
| | - Abdullah Hilal
- Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia; (A.A.P.); (M.V.P.); (S.S.A.); (A.H.); (N.N.S.)
| | - Nikolay N. Sokolov
- Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia; (A.A.P.); (M.V.P.); (S.S.A.); (A.H.); (N.N.S.)
| | - Dmitry D. Zhdanov
- Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia; (A.A.P.); (M.V.P.); (S.S.A.); (A.H.); (N.N.S.)
- Department of Biochemistry, Рeoples Friendship University of Russia (RUDN University), Miklukho-Maklaya st. 6, 117198 Moscow, Russia
| |
Collapse
|
17
|
Biomedical applications of microbial phenylalanine ammonia lyase: Current status and future prospects. Biochimie 2020; 177:142-152. [PMID: 32828824 DOI: 10.1016/j.biochi.2020.08.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/01/2020] [Accepted: 08/18/2020] [Indexed: 12/18/2022]
Abstract
Phenylalanine ammonia lyase (PAL) has recently emerged as an important therapeutic enzyme with several biomedical applications. The enzyme catabolizes l-phenylalanine to trans-cinnamate and ammonia. PAL is widely distributed in higher plants, some algae, ferns, and microorganisms, but absent in animals. Although microbial PAL has been extensively exploited in the past for producing industrially important metabolites, its high substrate specificity and catalytic efficacy lately spurred interest in its biomedical applications. PEG-PAL drug named Palynziq™, isolated from Anabaena variabilis has been recently approved for the treatment of adult phenylketonuria (PKU) patients. Further, it has exhibited high potency in regressing tumors and treating tyrosine related metabolic abnormalities like tyrosinemia. Several therapeutically valuable metabolites have been biosynthesized via its catalytic action including dietary supplements, antimicrobial peptides, aspartame, amino-acids, and their derivatives. This review focuses on all the prospective biomedical applications of PAL. It also provides an overview of the structure, production parameters, and various strategies to improve the therapeutic potential of this enzyme. Engineered PAL with improved pharmacodynamic and pharmacokinetic properties will further establish this enzyme as a highly efficient biological drug.
Collapse
|
18
|
Chepikova OE, Malin D, Strekalova E, Lukasheva EV, Zamyatnin AA, Cryns VL. Lysine oxidase exposes a dependency on the thioredoxin antioxidant pathway in triple-negative breast cancer cells. Breast Cancer Res Treat 2020; 183:549-564. [PMID: 32696316 DOI: 10.1007/s10549-020-05801-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/11/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Transformed cells are vulnerable to depletion of certain amino acids. Lysine oxidase (LO) catalyzes the oxidative deamination of lysine, resulting in lysine depletion and hydrogen peroxide production. Although LO has broad antitumor activity in preclinical models, the cytotoxic mechanisms of LO are poorly understood. METHODS Triple (ER/PR/HER2)-negative breast cancer (TNBC) cells were treated with control media, lysine-free media or control media supplemented with LO and examined for cell viability, caspase activation, induction of reactive oxygen species (ROS) and antioxidant signaling. To determine the role of nuclear factor erythroid 2-related factor 2 (NRF2) and thioredoxin reductase-1 (TXNRD1) in LO-induced cell death, NRF2 and TXNRD1 were individually silenced by RNAi. Additionally, the pan-TXNRD inhibitor auranofin was used in combination with LO. RESULTS LO activates caspase-independent cell death that is suppressed by necroptosis and ferroptosis inhibitors, which are inactive against lysine depletion, pointing to fundamental differences between LO and lysine depletion. LO rapidly induces ROS with a return to baseline levels within 24 h that coincides temporally with induction of TXNRD activity, the rate-limiting enzyme in the thioredoxin antioxidant pathway. ROS induction is required for LO-mediated cell death and NRF2-dependent induction of TXNRD1. Silencing NRF2 or TXNRD1 enhances the cytotoxicity of LO. The pan-TXNRD inhibitor auranofin is synergistic with LO against transformed breast epithelial cells, but not untransformed cells, underscoring the tumor-selectivity of this strategy. CONCLUSIONS LO exposes a redox vulnerability of TNBC cells to TXNRD inhibition by rendering tumor cells dependent on the thioredoxin antioxidant pathway for survival.
Collapse
Affiliation(s)
- Olga E Chepikova
- Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, MFCB 4144, 1685 Highland Avenue, Madison, WI, 53705, USA.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Dmitry Malin
- Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, MFCB 4144, 1685 Highland Avenue, Madison, WI, 53705, USA
| | - Elena Strekalova
- Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, MFCB 4144, 1685 Highland Avenue, Madison, WI, 53705, USA
| | - Elena V Lukasheva
- Peoples' Friendship, University of Russia (RUDN University), Moscow, Russia
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vincent L Cryns
- Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, MFCB 4144, 1685 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
19
|
Zarei M, Rahbar MR, Negahdaripour M, Morowvat MH, Nezafat N, Ghasemi Y. Cell Penetrating Peptide: Sequence-Based Computational Prediction for Intercellular Delivery of Arginine Deiminase. CURR PROTEOMICS 2020. [DOI: 10.2174/1570164616666190701120351] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:Cell-Penetrating Peptides (CPPs), a family of short peptides, are broadly used as the carrier in the delivery of drugs and different therapeutic agents. Thanks to the existence of valuable databases, computational screening of the experimentally validated CPPs can help the researchers to select more effective CPPs for the intercellular delivery of therapeutic proteins. Arginine deiminase of Mycoplasma hominis, an arginine-degrading enzyme, is currently in the clinical trial for treating several arginine auxotrophic cancers. However, some tumor cells have developed resistance to ADI treatment. The ADI resistance arises from the over-expression of argininosuccinate synthetase 1 enzyme, which is involved in arginine synthesis. Intracellular delivery of ADI into tumor cells is suggested as an efficient approach to overcome the aforesaid drawback.Objective:In this study, in-silico tools were used for evaluating the experimentally validated CPPs to select the best CPP candidates for the intracellular delivery of ADI.Results:In this regard, 150 CPPs of protein cargo available at CPPsite were retrieved and evaluated by the CellPPD server. The best CPP candidates for the intracellular delivery of ADI were selected based on stability and antigenicity of the ADI-CPP fusion form. The conjugated forms of ADI with each of the three CPPs including EGFP-hcT (9-32), EGFP-ppTG20, and F(SG)4TP10 were stable and nonantigenic; thus, these sequences were introduced as the best CPP candidates for the intracellular delivery of ADI. In addition, the proposed CPPs had appropriate positive charge and lengths for an efficient cellular uptake.Conclusion:These three introduced CPPs not only are appropriate for the intracellular delivery of ADI, but also can overcome the limitation of its therapeutic application, including short half-life and antigenicity.
Collapse
Affiliation(s)
- Mahboubeh Zarei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Rahbar
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
20
|
Zamyatnin AA. Proteins in Pharmacology: Restrictions, Challenges and Opportunities. Curr Med Chem 2019; 26:362-364. [DOI: 10.2174/092986732603190326151415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russian Federation
| |
Collapse
|
21
|
Du F, Chen J, Liu H, Cai Y, Cao T, Han W, Yi X, Qian M, Tian D, Nie Y, Wu K, Fan D, Xia L. SOX12 promotes colorectal cancer cell proliferation and metastasis by regulating asparagine synthesis. Cell Death Dis 2019; 10:239. [PMID: 30858360 PMCID: PMC6412063 DOI: 10.1038/s41419-019-1481-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 12/11/2022]
Abstract
The sex-determining region Y (SRY)-box (SOX) family has a crucial role in carcinogenesis and cancer progression. However, the role of SOX12 and the mechanism by which it is dysregulated in colorectal cancer (CRC) remain unclear. Here we analyzed SOX12 expression patterns in two independent CRC cohorts (cohort I, n = 390; cohort II, n = 363) and found that SOX12 was significantly upregulated in CRC, indicating a poor prognosis in CRC patients. Overexpression of SOX12 promoted CRC cell proliferation and metastasis, whereas downregulation of SOX12 hampered CRC aggressiveness. Mechanistically, SOX12 facilitated asparagine synthesis by transactivating glutaminase (GLS), glutamic oxaloacetic transaminase 2 (GOT2), and asparagine synthetase (ASNS). Downregulation of GLS, GOT2, and ASNS blocked SOX12-mediated CRC cell proliferation and metastasis, whereas ectopic expression of GLS, GOT2, and ASNS attenuated the SOX12 knockdown-induced suppression of CRC progression. In addition, serial deletion, site-directed mutagenesis, luciferase reporter, and chromatin immunoprecipitation (ChIP) assays indicated that hypoxia-inducible factor 1α (HIF-1α) directly binds to the SOX12 promoter and induces SOX12 expression. Administration of l-asparaginase decreased SOX12-mediated tumor growth and metastasis. In human CRC samples, SOX12 expression positively correlated with GLS, GOT2, ASNS, and HIF-1α expression. Based on these results, SOX12 may serve as a prognostic biomarker and l-asparaginase represents a potential novel therapeutic agent for CRC.
Collapse
Affiliation(s)
- Feng Du
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Jie Chen
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Hao Liu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Yanhui Cai
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Tianyu Cao
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Weili Han
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Xiaofang Yi
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Meirui Qian
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Limin Xia
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, China. .,Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
22
|
Yan S, Yang L, Lu L, Guo Q, Hu X, Yuan Y, Li Y, Wu M, Zhang J. Improved pharmacokinetic characteristics and bioactive effects of anticancer enzyme delivery systems. Expert Opin Drug Metab Toxicol 2018; 14:951-960. [DOI: 10.1080/17425255.2018.1505863] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Shenglei Yan
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Lan Yang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Luyang Lu
- College of Pharmacy, Southwest University for Nationalities, Chengdu, China
| | - Qi Guo
- Center for Certification and Evaluation, Chongqing Food and Drug Administration, Chongqing, China
| | - Xueyuan Hu
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Yuming Yuan
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Yao Li
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Mingjun Wu
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|