Monoclonal Antibody-Superantigen Fusion Proteins: Tumor-Specific Agents for T-Cell-Based Tumor Therapy MIKAEL DOHLSTEN*T, LARS Abrahmstnt, PER BJ6RK*, PETER A

Total Page:16

File Type:pdf, Size:1020Kb

Monoclonal Antibody-Superantigen Fusion Proteins: Tumor-Specific Agents for T-Cell-Based Tumor Therapy MIKAEL DOHLSTEN*T, LARS Abrahmstnt, PER BJ6RK*, PETER A Proc. Natl. Acad. Sci. USA Vol. 91, pp. 8945-8949, September 1994 Immunology Monoclonal antibody-superantigen fusion proteins: Tumor-specific agents for T-cell-based tumor therapy MIKAEL DOHLSTEN*t, LARS ABRAHMStNt, PER BJ6RK*, PETER A. LANDO*, GUNNAR HEDLUND*t, GORAN FORSBERG*, THOMAS BRODIN*, NICK R. J. GASCOIGNE§, CECILIA F6RBERGt, PETER LINDf, AND TERJE KALLAND*t¶ *Pharmacia Oncology Immunology, Lund, Sweden; tThe Wallenberg Laboratory, Department of Tumor Immunology, University of Lund, Lund, Sweden; *Pharmacia Bioscience Center, Stockholm, Sweden; and §The Scripps Research Institute, Department of Immunology, La Jolla, CA 92037 Communicated by Hans J. Maller-Eberhard, April IS, 1994 (receivedfor review October 18, 1993) ABSTRACT The bacterial superantigen staphylococcal We recently demonstrated that chemical conjugation ofmAb enterotoxin A (SEA) is an extremely potent activator of T against colon carcinomas with the bacterial SAg staphylo- lymphocytes when presented on major histocompatibility com- coccal enterotoxin A (SEA) targeted T cells to lyse MHC plex (MHC) class H molecules. To develop a tumor-specific class II tumor cells in vitro (6) and induced tumor-suppressive superantigen for cancer therapy, we have made a recombinant lymphokines (7). We have now expressed in Escherichia coli fusion protein of SEA and the Fab region of the C215 mono- a recombinant fusion protein of SEA and a Fab fragment of donal antibody specific for human colon carcinoma cells. SEA a colon carcinoma-reactive mAb. Fab-SEA treatment of as part of a fusion protein showed a >10-fold reduction in mice carrying tumors expressing the relevant antigen resulted MHC class II binding compared to native SEA, and accord- in 85-99%o suppression of tumor growth in the absence of ingly, the affinity of the FabC215-SEA fusion protein for the overt systemic side effects. We believe that tumor-specific C215 tumor antigen was 100-fold stronger than to MHC class SAgs should be ofconsiderable interest as therapeutic agents 1I molecules. The FabC215-SEA fusion protein efficiently against cancer. targeted T cells to lyse C215+ MHC class II- human colon carcinoma cells, which demonstrates functional substitution of MATERIALS AND METHODS the MHC class fl-dependent presentation of SEA with tumor specificity. Treatment of mice carrying B16 melanoma cells Protein Reagents. Recombinant SEA was expressed in E. expressing a transfected C215 antigen resulted in 85-99% coli and purified to homogeneity. The hybridomas secreting inhibition of tumor growth and allowed long-term survival of the C215 (IgG2a) and C242 (IgGl) mAbs, reacting with animals. The therapeutic effect was dependent on antigen- human colon cancer, were produced at Pharmacia (8, 9). E. specific targeting of the FabC215-SEA fusion protein, since coli HB101 was used as host during the DNA construction native SEA and an antigen-irrelevant FabC242-SEA fusion work (10). protein did not influence tumor growth. The results suggest Cloning ofthe cDNAs Encoding the Murine Antibodies C215 that Fab-SEA fusion proteins convey superantigenicity on and C242. PCR was performed on C215 hybridoma cells by tumor cells, which evokes T cells to suppress tumor growth. using the total RNA isolation kit (Ginna/Biotecx Laborato- ries, Friendswood, TX). Reverse transcriptase (from Molo- The therapeutic use of naked monoclonal antibodies (mAbs) ney murine leukemia virus, Superscript, GIBCO/BRL) and in human epithelial cancer has met with limited success (1). random hexamer primers were used to make cDNA. The The amount of mAb found to accumulate in the tumor is primers Mhvp-7 (11) and Mushcp-21 (5'-CAATTTTCT- generally low, tumor cells display extensive heterogeneity in TGTCCACCTTGGTGCT) were used to isolate the heavy antigen expression, and the effector mechanisms responsible (H)-chain DNA. Similarly, the primers Mkvp-7 (11) and for tumor-cell destruction are insufficient (1, 2). Recent Muslcp-11 (5'-TGTGTCCCGGGATACAGTTGGTGCAG- progress in the molecular understanding ofT-cell recognition CATCAGCCC) were used to isolate the K-chain DNA. The has provided insight into the role of T cells in the response C242 hybridoma cell line was used as source material for against allogeneic and neoplastic tissues. The vigorous and cloning ofthe cDNAs encoding the immunoglobulin H chain destructive response against an allograft involves a high and the K chain. Polyadenylylated RNA was extracted from frequency of T lymphocytes that, via the T-cell receptor hybridoma cells, converted to double-stranded cDNA, and (TCR), recognizes peptide antigens presented in the context cloned into the phage A-based vector Uni-ZAP (Stratagene). of major histocompatibility complex (MHC) molecules. In Positive clones were used to prepare cDNA containing contrast, the frequency of T cells responding to a tumor is pBluescript SK(-) plasmids by phagemid excision and prop- generally low and insufficient to interfere with tumor growth. agation. The resulting cDNA-containing plasmids were char- This suggests that an attractive approach for immunotherapy acterized by restriction enzyme mapping, and the nucleotide would be to target a high frequency of T cells to the tumor sequences of inserts from selected plasmids were deter- area. We have therefore developed an antibody-based ther- mined. apy that provides tumor cells with superantigenicity. Super- Assembly of the Fab-SEA Gene Fusion Segment. The fol- antigens (SAgs) are a family of bacterial and viral proteins lowing pieces were used: (i) A Not I-Mlu I fragment con- that activate a high frequency of T cells to cytokine release taining the ribosomal binding site and most of the signal and cell-mediated cytotoxicity (3-5). Bacterial SAgs bind peptide encoding a portion of the gene from E. coli outer with high affinity to MHC class II molecules and subse- membrane protein A as translation and secretion signals for quently interact with T cells expressing particular sequences in the variable (V) region ofthe TCR ,/ chain (TCR V(3) (3-5). Abbreviations: SEA, staphylococcal enterotoxin A; SAg, superan- tigen; MHC, major histocompatibility complex; CTL, cytotoxic T lymphocyte; TCR, T-cell receptor; mAb, monoclonal antibody; V, The publication costs ofthis article were defrayed in part by page charge variable; C, constant; H, heavy; V8, (8 chain V region. payment. This article must therefore be hereby marked "advertisement" ITo whom reprint requests should be addressed at: Pharmacia in accordance with 18 U.S.C. §1734 solely to indicate this fact. Oncology Immunology, Scheelevagen 22, S-223 63 Lund, Sweden. 8945 Downloaded by guest on September 29, 2021 8946 Immunology: Dohlsten et al. Proc. Natl. Acad. Sci. USA 91 (1994) the H chain. The introduction of the Mlu I site changed the Cell Lines. The human B-cell lymphoma Raji, the human second to the last codon from CAG to AAC. (ii) The H chain colon carcinomas SW620 and Colo2O5, the murine B16 V region gene was modified by introducing flanking BssHII melanoma (ATCC, MD), and CHO cells were cultured as and Xma I sites by PCR. The last base of the former site is described (6, 16). SEA-reactive T-cell lines (>99%o CD3+) the first base ofthe gene and the latter spans codons 12-14 of were established from peripheral blood lymphocytes as de- the constant (C) portion. (iii) The gene encoding the first H scribed (6). chain C region was obtained from mAb C242 gene. The Transfection with cDNA Encoding the C215 Antigen. The encoded protein is identical to this portion of a consensus expression vector pKGE839 containing the GA733-2 cDNA murine IgG1 chain. A Xma I-Rsr II fragment was used from (encoding the C215 antigen) and the neomycin-resistance codon 12 including the first Cys codon of the hinge-encoding gene (14) was transfected into B16 melanoma cells or CHO region followed by the tripeptide spacer Gly-Gly-Pro. (iv) cells using Transfectam (Sepracor, IBF, Villeneuve-la- The SEA gene included on a Rsr II-Sfi I fragment was taken Garenne, France) and electroporation, respectively. G418- from the plasmid pKP554. The SEA gene contained on this resistent clones were harvested and subjected to repeated cell plasmid is identical to the sequence published by Betley and sorting. The sorted CHO-C215 cells were kept as a stable cell Mekalanos (12), except for two TAA stop codons. (v) The line, and high-expressing clones were selected from the translation and secretion signals for the K chain were derived B16-C215 cells. Establishment of CHO cells expressing from a Sfi I-Mlu I fragment. This contained 40 bp from the HLA-DR has been described (16). staphylococcal protein A gene (13), including the ribosomal Cytotoxicity Assay. Cytotoxicity was measured at various binding site and the first 10 bp ofthe signal peptide sequence, effector/target cell ratios in 4-h 51Cr release assays as de- followed by 59 bases to compose a synthetic signal peptide. scribed (6, 16). (vi) The K chain V region gene was modified by introducing Immunohistochenical Staining. Tumor tissues from colon flanking Dsa I and Kas I sites by PCR. The last base of the cancer patients were snap-frozen in liquid nitrogen-cooled former site is the first base of the gene and the latter spans isopentane and 4-pm frozen sections were prepared. After codons 4-6 of the C portion. (vii) The gene encoding the K drying and fixation in acetone, the sections were rehydrated chain C domain was obtained from the mAb C242 gene, with and stained with FabC215-SEA and SEA followed by anti- the same Kas I site as above introduced and ending with a mouse immunoglobulin-biotinylated antibody or rabbit anti- TAA stop codon followed by a Xba I site. SEA-biotinylated antibody and avidin-biotinylated peroxi- Expression and Purification ofFab-SEA Fusion Proteins. E. dase complex (17). The slides were developed in diaminoben- coli K-12 UL635 (ara-14, xyl-7, AompT, T4R) was used for zidine (Sigma) and mounted in DPX medium (Sigma).
Recommended publications
  • Elimination Pathways of Fusion Protein and Peptide Drugs
    Review Volume 11, Issue 2, 2021, 9139 - 9147 https://doi.org/10.33263/BRIAC112.91399147 Elimination Pathways of Fusion Protein and Peptide Drugs Ali Khodadoust 1 , Hossein Aghamollaei 2 , Ali Mohammad Latifi 1 , Kazem Hasanpour 3 , Mahdi Kamali 4 , Hamid Tebyanian 5 , Gholamreza Farnoosh 1,* 1 Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran 2 Chemical Injuries Research Center, Systems biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran 3 Sabzevar University of Medical Sciences, School of Medicine, Sabzevar, Iran 4 Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran 5 Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran * Correspondence: [email protected]; Scopus Author ID 55855454400 Received: 28.07.2020; Revised: 25.08.2020; Accepted: 27.08.2020; Published: 1.09.2020 Abstract: Fusion proteins have been known as an interesting subject for scientific researches in improving properties or making a new function by synergistically incorporating two protein domains into one complex. Fusion proteins are created by ligation of two or more protein domains in a single polypeptide with functional properties. Improvement of therapeutic function is one of the main goals for developing these products. Elimination from the body is one of the most important points that should be considered in the design and production of fusion proteins. This review describes some of the most important excretion/elimination pathways of fusion peptide and protein drugs as well as serious elimination challenges in the development and manufacturing of fusion proteins. Keywords: Fusion proteins; Therapy; Pharmacokinetics; Serum half-life; Peptide.
    [Show full text]
  • CUE-101, a Novel Fc Fusion Protein for Selective Targeting and Expansion of Anti-Tumor T Cells for Treatment of HPV-Driven Malignancies
    CUE-101, a novel Fc fusion protein for selective targeting and expansion of anti-tumor T cells for treatment of HPV-driven malignancies Natasha Girgis1, Steven N. Quayle1, Alyssa Nelson1, Dharma Raj Thapa1, Sandrine Hulot1, Lauren Kraemer1, Miguel Moreta1, Zohra Merazga1, Robert Ruidera1, Dominic Beal1, Mark Haydock1, Jonathan Soriano1, Luke Witt1, Jessica Ryabin1, Emily Spaulding1, John F. Ross1, Saso Cemerski1, Anish Suri1, Rodolfo Chaparro1, Ronald Seidel1, Kenneth J. Pienta2, Mary C. Simcox1 1Cue Biopharma, Cambridge, Massachusetts; 2The James Buchanan Brady Urological Institute and the Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland + + Background CUE-101 selectively expands E711-20-specific CD8 T mCUE-101 expands functional antigen-specific CD8 T • Human papilloma virus (HPV) is responsible for 72% of oropharyngeal, 90% of cervical, 90% cells from healthy human PBMCs cells in the tumor and the periphery of anal, and 71% of vulvar, vaginal, or penile cancers, causing significant morbidity and A. B. A. B. C. 9 mortality worldwide. Innovative therapies are urgently needed for these malignancies, Vehicle 100 nM CUE-101 107 15 Peripheral Blood particularly in the largely incurable metastatic setting. 106 10 5 • The E7 oncoprotein is constitutively expressed in HPV-associated cancers, is necessary for 105 ⍺PD-1 5 PE 4 Vehicle mCUE-101 Combo - 10 initiation and maintenance of malignant transformation, and is genetically conserved in 1 cancer (Mirabello 2017). 103 1 1 102 E7-Specific of CD8+ T cells % IFNg+,TNFa+
    [Show full text]
  • Interferon-Based Biopharmaceuticals: Overview on the Production, Purification, and Formulation
    Review Interferon-Based Biopharmaceuticals: Overview on the Production, Purification, and Formulation Leonor S. Castro 1,†, Guilherme S. Lobo 1,†, Patrícia Pereira 2 , Mara G. Freire 1 ,Márcia C. Neves 1,* and Augusto Q. Pedro 1,* 1 CICECO–Aveiro Institute of Materials, Chemistry Department, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; [email protected] (L.S.C.); [email protected] (G.S.L.); [email protected] (M.G.F.) 2 Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal; [email protected] * Correspondence: [email protected] (M.C.N.); [email protected] (A.Q.P.) † These authors contributed equally to this work. Abstract: The advent of biopharmaceuticals in modern medicine brought enormous benefits to the treatment of numerous human diseases and improved the well-being of many people worldwide. First introduced in the market in the early 1980s, the number of approved biopharmaceutical products has been steadily increasing, with therapeutic proteins, antibodies, and their derivatives accounting for most of the generated revenues. The success of pharmaceutical biotechnology is closely linked with remarkable developments in DNA recombinant technology, which has enabled the production of proteins with high specificity. Among promising biopharmaceuticals are interferons, first described by Isaacs and Lindenmann in 1957 and approved for clinical use in humans nearly thirty years later. Interferons are secreted autocrine and paracrine proteins, which by regulating several biochemical Citation: Castro, L.S.; Lobo, G.S.; pathways have a spectrum of clinical effectiveness against viral infections, malignant diseases, Pereira, P.; Freire, M.G.; Neves, M.C.; and multiple sclerosis.
    [Show full text]
  • A Novel Method for Targeted Protein Degradation
    bioRxiv preprint doi: https://doi.org/10.1101/2020.07.31.231787; this version posted August 2, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Predator: A novel method for targeted protein degradation Chuanyang Liu1, †, Jingyu Kuang1, †, Xinyuan Qiu1, †, Lu Min1, Wenying Li1, Jiaxin Ma1, Lingyun Zhu1,* 1 Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha 410073, China † These authors contributed equally to this work. ∗ Correspondence address. Tel: +86-731-87001812; Fax/Tel: +86-731-87001801; E-mail: [email protected] Graphic abstract bioRxiv preprint doi: https://doi.org/10.1101/2020.07.31.231787; this version posted August 2, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract Protein expression and degradation are fundamental to cell function and physiological status of organisms. Interfering with protein expression not only provides powerful strategies to analyze the function of proteins but also inspires effective treatment methods for diseases caused by protein dysfunction. Recently, harnessing the power of the ubiquitin-proteasome system for targeted protein degradation (TPD) has become the focus of researches. Over the past two decades, TPD technologies, such as E3 ligase modification, PROTACs, and the Trim-Away method, have successfully re-oriented the ubiquitin-proteasome pathway and thus degraded many pathogenic proteins and even "undruggable" targets.
    [Show full text]
  • Overview of Antibody Drug Delivery
    pharmaceutics Review Overview of Antibody Drug Delivery Sahar Awwad 1,2,* ID and Ukrit Angkawinitwong 1 1 UCL School of Pharmacy, London WC1N 1AX, UK; [email protected] 2 National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1 V9EL, UK * Correspondence: [email protected]; Tel.: +44-207-753-5802 Received: 27 March 2018; Accepted: 29 June 2018; Published: 4 July 2018 Abstract: Monoclonal antibodies (mAbs) are one of the most important classes of therapeutic proteins, which are used to treat a wide number of diseases (e.g., oncology, inflammation and autoimmune diseases). Monoclonal antibody technologies are continuing to evolve to develop medicines with increasingly improved safety profiles, with the identification of new drug targets being one key barrier for new antibody development. There are many opportunities for developing antibody formulations for better patient compliance, cost savings and lifecycle management, e.g., subcutaneous formulations. However, mAb-based medicines also have limitations that impact their clinical use; the most prominent challenges are their short pharmacokinetic properties and stability issues during manufacturing, transport and storage that can lead to aggregation and protein denaturation. The development of long acting protein formulations must maintain protein stability and be able to deliver a large enough dose over a prolonged period. Many strategies are being pursued to improve the formulation and dosage forms of antibodies to improve efficacy and to increase the range of applications for the clinical use of mAbs. Keywords: antibodies; protein; pharmacokinetics; drug delivery; stability 1.
    [Show full text]
  • COVID-19: Mechanisms of Vaccination and Immunity
    Review COVID-19: Mechanisms of Vaccination and Immunity Daniel E. Speiser 1,* and Martin F. Bachmann 2,3,4,* 1 Department of Oncology, University Hospital and University of Lausanne, 1066 Lausanne, Switzerland 2 International Immunology Centre, Anhui Agricultural University, Hefei 230036, China 3 Department of Rheumatology, Immunology and Allergology, Inselspital, University of Bern, 3010 Bern, Switzerland 4 Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland * Correspondence: [email protected] (D.E.S.); [email protected] (M.F.B.) Received: 2 July 2020; Accepted: 20 July 2020; Published: 22 July 2020 Abstract: Vaccines are needed to protect from SARS-CoV-2, the virus causing COVID-19. Vaccines that induce large quantities of high affinity virus-neutralizing antibodies may optimally prevent infection and avoid unfavorable effects. Vaccination trials require precise clinical management, complemented with detailed evaluation of safety and immune responses. Here, we review the pros and cons of available vaccine platforms and options to accelerate vaccine development towards the safe immunization of the world’s population against SARS-CoV-2. Favorable vaccines, used in well-designed vaccination strategies, may be critical for limiting harm and promoting trust and a long-term return to normal public life and economy. Keywords: SARS-CoV-2; COVID-19; nucleic acid tests; serology; vaccination; immunity 1. Introduction The COVID-19 pandemic holds great challenges for which the world is only partially prepared [1]. SARS-CoV-2 combines serious pathogenicity with high infectivity. The latter is enhanced by the fact that asymptomatic and pre-symptomatic individuals can transmit the virus, in contrast to SARS-CoV-1 and MERS-CoV, which were transmitted by symptomatic patients and could be contained more efficiently [2,3].
    [Show full text]
  • Incubation Period of Measles from Exposure to Prodrome ■ Exposure to Rash Onset Averages 11 to 12 Days
    Measles Paul Gastanaduy, MD; Penina Haber, MPH; Paul A. Rota, PhD; and Manisha Patel, MD, MS Measles is an acute, viral, infectious disease. References to measles can be found from as early as the 7th century. The Measles disease was described by the Persian physician Rhazes in the ● Acute viral infectious disease 10th century as “more to be dreaded than smallpox.” ● First described in 7th century In 1846, Peter Panum described the incubation period of ● Vaccines first licensed include measles and lifelong immunity after recovery from the disease. measles in 1963, MMR in 1971, John Enders and Thomas Chalmers Peebles isolated the virus in and MMRV in 2005 human and monkey kidney tissue culture in 1954. The first live, ● Infection nearly universal attenuated vaccine (Edmonston B strain) was licensed for use in during childhood in the United States in 1963. In 1971, a combined measles, mumps, prevaccine era and rubella (MMR) vaccine was licensed for use in the United ● Still common and often fatal States. In 2005, a combination measles, mumps, rubella, and in developing countries varicella (MMRV) vaccine was licensed. Before a vaccine was available, infection with measles virus was nearly universal during childhood, and more than 90% of persons were immune due to past infection by age 15 years. Measles is still a common and often fatal disease in developing countries. The World Health Organization estimates there were 142,300 deaths from measles globally in 2018. In the United 13 States, there have been recent outbreaks; the largest occurring in 2019 primarily among people who were not vaccinated.
    [Show full text]
  • The Strategy of Fusion Genes Construction Determines Efficient
    Vol. 61, No 3/2014 773–778 on-line at: www.actabp.pl Regular paper ­­The strategy of fusion genes construction determines efficient expression of introduced transcription factors Tomasz Adamus, Paweł Konieczny, Małgorzata Sekuła, Maciej Sułkowski and Marcin Majka* Department of Transplantation, Jagiellonian University Medical College, Kraków, Poland The main goal in gene therapy and biomedical research Introduction of transgene in the presence of reporter is an efficient transcription factors (TFs) delivery sys- gene often requires strategies that allow co-expression of tem. SNAIL, a zinc finger transcription factor, is strongly introduced genes, so several approaches have been em- involved in tumor, what makes its signaling pathways ployed. One of them is insertion of internal ribosomal an interesting research subject. The necessity of track- entry sites (IRESs) between genes, which is widely used ing activation of intracellular pathways has prompted (Mokrejs, 2006). The other strategy is the usage of dual fluorescent proteins usage as localization markers. Ad- or multiple promoter systems, but it can be associated vanced molecular cloning techniques allow to generate with differences in protein expression levels or silencing fusion proteins from fluorescent markers and transcrip- of some promoters (Radcliffe & Mitrophanous, 2004). tion factors. Depending on fusion strategy, the protein It is also possible to introduce genes applying multiple expression levels and nuclear transport ability are sig- vectors transfection, however uneven transcription effi- nificantly different. The P2A self-cleavage motif through ciency has to be considered as well as increased toxicity its cleavage ability allows two single proteins to be si- (Arbab et al., 2004). The fusion genes under control of multaneously expressed.
    [Show full text]
  • Forced Expression of the DEK-NUP214 Fusion Protein
    Sandén et al. BMC Cancer 2013, 13:440 http://www.biomedcentral.com/1471-2407/13/440 RESEARCH ARTICLE Open Access Forced expression of the DEK-NUP214 fusion protein promotes proliferation dependent on upregulation of mTOR Carl Sandén1*, Malin Ageberg1, Jessica Petersson1, Andreas Lennartsson2 and Urban Gullberg1 Abstract Background: The t(6;9)(p23;q34) chromosomal translocation is found in 1% of acute myeloid leukemia and encodes the fusion protein DEK-NUP214 (formerly DEK-CAN) with largely uncharacterized functions. Methods: We expressed DEK-NUP214 in the myeloid cell lines U937 and PL-21 and studied the effects on cellular functions. Results: In this study, we demonstrate that expression of DEK-NUP214 increases cellular proliferation. Western blot analysis revealed elevated levels of one of the key proteins regulating proliferation, the mechanistic target of rapamycin, mTOR. This conferred increased mTORC1 but not mTORC2 activity, as determined by the phosphorylation of their substrates, p70 S6 kinase and Akt. The functional importance of the mTOR upregulation was determined by assaying the downstream cellular processes; protein synthesis and glucose metabolism. A global translation assay revealed a substantial increase in the translation rate and a metabolic assay detected a shift from glycolysis to oxidative phosphorylation, as determined by a reduction in lactate production without a concomitant decrease in glucose consumption. Both these effects are in concordance with increased mTORC1 activity. Treatment with the mTORC1 inhibitor everolimus (RAD001) selectively reversed the DEK-NUP214-induced proliferation, demonstrating that the effect is mTOR-dependent. Conclusions: Our study shows that the DEK-NUP214 fusion gene increases proliferation by upregulation of mTOR, suggesting that patients with leukemias carrying DEK-NUP214 may benefit from treatment with mTOR inhibitors.
    [Show full text]
  • Studies to Prevent Degradation of Recombinant Fc-Fusion Protein Expressed in Mammalian Cell Line and Protein Characterization
    International Journal of Molecular Sciences Article Studies to Prevent Degradation of Recombinant Fc-Fusion Protein Expressed in Mammalian Cell Line and Protein Characterization Sanjukta Chakrabarti 1,†, Colin J. Barrow 2,†, Rupinder K. Kanwar 3, Venkata Ramana 1,*,† and Jagat R. Kanwar 3,* 1 Reliance Life Sciences, Dhirubhai Ambani Life Sciences Center, Navi Mumbai 400701, India; [email protected] 2 School of Life and Environmental Sciences, Deakin University, Waurn Ponds Campus, Geelong 3216, Australia; [email protected] 3 Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Centre for Molecular and Medical Research (C-MMR), Deakin University, Waurn Ponds Campus, Geelong 3216, Australia; [email protected] * Correspondence: [email protected] (V.R.); [email protected] (J.R.K.); Tel.: +91-22-6767-8402 (V.R.); +61-3-5227-1148 (J.R.K.) † These authors contributed equally to this work. Academic Editor: Salvador Ventura Received: 5 January 2016; Accepted: 27 May 2016; Published: 9 June 2016 Abstract: Clipping of recombinant proteins is a major issue in animal cell cultures. A recombinant Fc-fusion protein, VEGFR1(D1–D3)-Fc expressed in CHOK1SV GS-KO cells was observed to be undergoing clippings in lab scale cultures. Partial cleaving of expressed protein initiated early on in cell culture and was observed to increase over time in culture and also on storage. In this study, a few parameters were explored in a bid to inhibit clipping in the fusion protein The effects of culture temperature, duration of culture, the addition of an anti-clumping agent, ferric citrate and use of protease inhibitor cocktail on inhibition of proteolysis of the Fc fusion were studied.
    [Show full text]
  • Experimental Approaches in Delineating Mtor Signaling
    G C A T T A C G G C A T genes Review Experimental Approaches in Delineating mTOR Signaling Jiayi Qian 1,2, Siyuan Su 1,2 and Pengda Liu 1,2,* 1 Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; [email protected] (J.Q.); [email protected] (S.S.) 2 Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA * Correspondence: [email protected]; Tel.: +01-919-966-3522 Received: 11 June 2020; Accepted: 30 June 2020; Published: 2 July 2020 Abstract: The mTOR signaling controls essential biological functions including proliferation, growth, metabolism, autophagy, ageing, and others. Hyperactivation of mTOR signaling leads to a plethora of human disorders; thus, mTOR is an attractive drug target. The discovery of mTOR signaling started from isolation of rapamycin in 1975 and cloning of TOR genes in 1993. In the past 27 years, numerous research groups have contributed significantly to advancing our understanding of mTOR signaling and mTOR biology. Notably, a variety of experimental approaches have been employed in these studies to identify key mTOR pathway members that shape up the mTOR signaling we know today. Technique development drives mTOR research, while canonical biochemical and yeast genetics lay the foundation for mTOR studies. Here in this review, we summarize major experimental approaches used in the past in delineating mTOR signaling, including biochemical immunoprecipitation approaches, genetic approaches, immunofluorescence microscopic approaches, hypothesis-driven studies, protein sequence or motif search driven approaches, and bioinformatic approaches.
    [Show full text]
  • A Chimeric Fusion Protein Containing Transforming Growth Factor-Α
    Gene Therapy (1998) 5, 521–530 1998 Stockton Press All rights reserved 0969-7128/98 $12.00 http://www.stockton-press.co.uk/gt A chimeric fusion protein containing transforming growth factor-␣ mediates gene transfer via binding to the EGF receptor J Fominaya1,2, C Uherek1 and W Wels1 1Institute for Experimental Cancer Research, Tumor Biology Center, Breisacher Strasse 117, D-79106 Freiburg, Germany Fusion proteins engineered to incorporate distinct functions binding to EGF receptors. Complexes of the chimeric pro- which co-operate in mediating the cell-type specific uptake tein and plasmid DNA carrying a luciferase reporter gene, and intracellular delivery of DNA present an attractive after condensation with poly-L-lysine resulted in an up to approach for the development of self-assembling vector 150-fold increase in reporter gene expression in EGF systems for targeted gene transfer. Here we have chosen receptor expressing cells in comparison to poly-L-lysine– the EGF receptor overexpressed in many human tumors DNA complexes alone. While in COS-1 cells no of epithelial origin as a target for a novel modular fusion additional endosome escape activity was required, in protein. We have fused a cDNA fragment of the human A431 cells gene delivery was dependent on the simul- EGF receptor ligand TGF-␣ to sequences encoding the taneous presence of the endosome destabilizing translocation domain of Pseudomonas exotoxin A as an reagent chloroquine indicating that cell-type specific fac- endosome escape activity, and the DNA-binding domain of tors such as different intracellular routing of protein–DNA the yeast GAL4 transcription factor.
    [Show full text]