1
|
Srivastava A, Pati S, Kaushik H, Singh S, Garg LC. Toxin-antitoxin systems and their medical applications: current status and future perspective. Appl Microbiol Biotechnol 2021; 105:1803-1821. [PMID: 33582835 DOI: 10.1007/s00253-021-11134-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/11/2022]
Abstract
Almost all bacteria synthesize two types of toxins-one for its survival by regulating different cellular processes and another as a strategy to interact with host cells for pathogenesis. Usually, "bacterial toxins" are contemplated as virulence factors that harm the host organism. However, toxins produced by bacteria, as a survival strategy against the host, also hamper its cellular processes. To overcome this, the bacteria have evolved with the production of a molecule, referred to as antitoxin, to negate the deleterious effect of the toxin against itself. The toxin and antitoxins are encoded by a two-component toxin-antitoxin (TA) system. The antitoxin, a protein or RNA, sequesters the toxins of the TA system for neutralization within the bacterial cell. In this review, we have described different TA systems of bacteria and their potential medical and biotechnological applications. It is of interest to note that while bacterial toxin-antitoxin systems have been well studied, the TA system in unicellular eukaryotes, though predicted by the investigators, have never been paid the desired attention. In the present review, we have also touched upon the TA system of eukaryotes identified to date. KEY POINTS: Bacterial toxins harm the host and also affect the bacterial cellular processes. The antitoxin produced by bacteria protect it from the toxin's harmful effects. The toxin-antitoxin systems can be targeted for various medical applications.
Collapse
Affiliation(s)
- Akriti Srivastava
- Department of Life Sciences, Shiv Nadar University, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh, 201314, India
| | - Soumya Pati
- Department of Life Sciences, Shiv Nadar University, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh, 201314, India
| | - Himani Kaushik
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Lalit C Garg
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, 110067, India.
| |
Collapse
|
2
|
Jacobson JM, Jadlowsky JK, Lacey SF, Fraietta JA, Plesa G, Chono H, Lee DH, Kulikovskaya I, Bartoszek C, Chen F, Tian L, Dimitri A, Levine BL, Veloso EA, Hwang WT, June CH. Autologous CD4 T Lymphocytes Modified with a Tat-Dependent, Virus-Specific Endoribonuclease Gene in HIV-Infected Individuals. Mol Ther 2021; 29:626-635. [PMID: 33186691 PMCID: PMC7854306 DOI: 10.1016/j.ymthe.2020.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/28/2020] [Accepted: 11/05/2020] [Indexed: 11/24/2022] Open
Abstract
MazF is an Escherichia coli-derived endoribonuclease that selectively cleaves ACA sequences of mRNA prevalent in HIV. We administered a single infusion of autologous CD4 T lymphocytes modified to express a Tat-dependent MazF transgene to 10 HIV-infected individuals (six remaining on antiretroviral therapy [ART]; four undergoing treatment interruption post-infusion) in order to provide a population of HIV-resistant immune cells. In participants who remained on ART, increases in CD4 and CD8 T cell counts of ~200 cells/mm3 each occurred within 2 weeks of infusion and persisted for at least 6 months. Modified cells were detectable for several months in the blood and trafficked to gastrointestinal lymph tissue. HIV-1 Tat introduced ex vivo to the modified CD4+ T cells induced MazF expression in both pre- and post-infusion samples, and MazF expression was detected in vivo post-viral-rebound during ATI. One participant experienced mild cytokine release syndrome. In sum, this study of a single infusion of MazF-modified CD4 T lymphocytes demonstrated safety of these cells, distribution to lymph tissue and maintenance of Tat-inducible MazF endoribonuclease activity, as well as sustained elevation of blood CD4 and CD8 T cell counts. Future studies to assess effects on viremia and latent proviral reservoir are warranted.
Collapse
Affiliation(s)
- Jeffrey M Jacobson
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Julie K Jadlowsky
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Simon F Lacey
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Dong H Lee
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Irina Kulikovskaya
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chelsie Bartoszek
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fang Chen
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lifeng Tian
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander Dimitri
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth A Veloso
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei-Ting Hwang
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
3
|
Targeting Type II Toxin-Antitoxin Systems as Antibacterial Strategies. Toxins (Basel) 2020; 12:toxins12090568. [PMID: 32899634 PMCID: PMC7551001 DOI: 10.3390/toxins12090568] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 08/31/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
The identification of novel targets for antimicrobial agents is crucial for combating infectious diseases caused by evolving bacterial pathogens. Components of bacterial toxin–antitoxin (TA) systems have been recognized as promising therapeutic targets. These widespread genetic modules are usually composed of two genes that encode a toxic protein targeting an essential cellular process and an antitoxin that counteracts the activity of the toxin. Uncontrolled toxin expression may elicit a bactericidal effect, so they may be considered “intracellular molecular bombs” that can lead to elimination of their host cells. Based on the molecular nature of antitoxins and their mode of interaction with toxins, TA systems have been classified into six groups. The most prevalent are type II TA systems. Due to their ubiquity among clinical isolates of pathogenic bacteria and the essential processes targeted, they are promising candidates for the development of novel antimicrobial strategies. In this review, we describe the distribution of type II TA systems in clinically relevant human pathogens, examine how these systems could be developed as the targets for novel antibacterials, and discuss possible undesirable effects of such therapeutic intervention, such as the induction of persister cells, biofilm formation and toxicity to eukaryotic cells.
Collapse
|
4
|
Okamoto M, Chono H, Hidaka A, Toyama M, Mineno J, Baba M. Induction of E. coli-derived endonuclease MazF suppresses HIV-1 production and causes apoptosis in latently infected cells. Biochem Biophys Res Commun 2020; 530:597-602. [PMID: 32747090 DOI: 10.1016/j.bbrc.2020.07.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 07/22/2020] [Indexed: 10/23/2022]
Abstract
The current antiretroviral therapy cannot cure the patients infected with human immunodeficiency virus type 1 (HIV-1) due to the existence of latently infected cells capable of virus production from harboring proviral DNA. MazF is an ACA nucleotide sequence-specific endoribonuclease derived from Escherichia coli. The conditional expression of MazF by binding of HIV-1 Tat to the promoter region of a MazF-expression vector has previously been shown to selectively inhibit HIV-1 replication in acutely infected cells. The expression of MazF significantly suppressed tumor necrosis factor (TNF)-α-induced HIV-1 production and viral RNA expression in the HIV-1 latently infected cell line OM-10.1 transduced with the MazF-expression vector (OM-10.1/MFR). Moreover, the viability of OM-10.1/MFR cells decreased with increasing concentrations of TNF-α, whereas such decrease was not observed for HL-60 cells transduced with the MazF-expression vector (HL-60/MFR), the uninfected parental cell line of OM-10.1. TNF-α increased the expression of cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase in OM-10.1/MFR cells, indicating that the cell death was caused by the induction of apoptosis. TNF-α-induced expression of MazF mRNA was detected in OM-10.1/MFR but not HL-60/MFR cells, suggesting that TNF-α-induced apoptosis of latently infected cells was due to the expression of MazF. Thus, the anti-HIV-1 gene therapy using the MazF-expression vector may have potential for the cure of HIV-1 infection in combination with suitable latency reversing agents through reducing the size of latently infected cells without viral reactivation.
Collapse
Affiliation(s)
- Mika Okamoto
- Division of Antiviral Chemotherapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, 890-8544, Japan
| | | | - Akemi Hidaka
- Division of Antiviral Chemotherapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, 890-8544, Japan
| | - Masaaki Toyama
- Division of Antiviral Chemotherapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, 890-8544, Japan
| | | | - Masanori Baba
- Division of Antiviral Chemotherapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, 890-8544, Japan.
| |
Collapse
|
5
|
Jahanshahi S, Li Y. An Effective Method for Quantifying RNA Expression of IbsC-SibC, a Type I Toxin-Antitoxin System in Escherichia coli. Chembiochem 2020; 21:3120-3130. [PMID: 32516493 DOI: 10.1002/cbic.202000280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/04/2020] [Indexed: 01/28/2023]
Abstract
Toxin and antitoxin (TA) systems are small genetic modules consisting of a toxin protein and an RNA or protein antitoxin. It is difficult to study their functions in a large part due to the lack of effective methods to study toxin RNAs, which usually exist at exceptionally low levels. Herein, we describe a sensitive reverse transcription quantitative PCR (RT-qPCR) method that is able to quantitate such RNA species. The method was directed at detection of the toxin mRNA of the ibsC-sibC TA pair, and its high specificity was validated by sequencing. The approach was used to determine relative expression of the IbsC and SibC RNAs at different cell-growth phases; this revealed an expression pattern that cannot be explained by the prevailing notion of growth stasis by the toxin and rescue by the antitoxin. The usefulness of the method was further showcased by the determination of average cellular copy numbers of the IbsC-SibC RNAs in wild-type E. coli cells and RNA abundance in E. coli cells engineered with extra copies of the ibsC-sibC genes. With a robust method to quantitate cellular small RNAs at very low concentrations, we are now equipped to study the expression of TA systems under different conditions to gain useful insights about their functions.
Collapse
Affiliation(s)
- Shahrzad Jahanshahi
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, DeGroote School of Medicine, School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4 K1, Canada
| | - Yingfu Li
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, DeGroote School of Medicine, School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4 K1, Canada
| |
Collapse
|
6
|
Gao CE, Song Q, Zhang M, Li J, Miao Y, Li Z, Dong J. Generation, ex vivo expansion and safety of engineered PD1-knockout primary T cells from cynomolgus macaques. Mol Immunol 2020; 124:100-108. [PMID: 32554100 DOI: 10.1016/j.molimm.2020.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/22/2020] [Accepted: 05/07/2020] [Indexed: 10/24/2022]
Abstract
Programmed cell death protein 1 (PD1) is a cell-surface receptor that plays a vital regulatory role in suppressing inflammatory T cell activity; therefore, it is an ideal target for T cell-redirecting therapies. Here, we describe a cynomolgus macaque model for studying the transfer of PD1-modified T cells. We developed the first T cell engager targeting the disruption of PD1 by electroporation of plasmids encoding sgRNA and Cas9. There were no significant differences between mock T cells and PD1-knockout (PD1-KO) T cells in terms of cell viability, T cell signature marker expression, cell apoptosis, or cell cycling during prolonged in vitro culture. However, in a mixed lymphocyte reaction, PD1-KO T cells exhibited increased proliferation for both CD4+ and CD8+T cells and enhanced IFNγ release. We adoptively transferred autologous PD1-KO T cells into three cynomolgus monkeys. The PD1-KO T cells did not cause overt toxicity as measured by evaluating body weight, hematological parameters, and blood chemistry parameters. Histopathological analyses of tissues showed no lesions related to the infused PD1-KO T cells. Our findings demonstrate the utility of cynomolgus monkeys in expanding PD1-KO T cells and evaluating the safety of this immunotherapy and provide a new strategy for T cell-based adoptive cell therapies.
Collapse
Affiliation(s)
- Chang-E Gao
- Department of Medical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China; Department of Medical Oncology, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Qian Song
- Department of Medical Oncology, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Ming Zhang
- Department of Radiotherapy, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Jian Li
- Kunming Biological Diversity Regional Center of Instruments, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
| | - Yi Miao
- Department of Medical Oncology, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Zhenhui Li
- Department of Radiotherapy, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Jian Dong
- Department of Medical Oncology, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China.
| |
Collapse
|
7
|
Yamamoto S, Matsumoto SI, Shimizu H, Hirabayashi H. Quantitative application of flow cytometry for the analysis of circulating human T cells: A preclinical pharmacokinetic study. Drug Metab Pharmacokinet 2020; 35:207-213. [DOI: 10.1016/j.dmpk.2019.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/09/2019] [Accepted: 11/10/2019] [Indexed: 02/06/2023]
|
8
|
Bravo A, Ruiz-Cruz S, Alkorta I, Espinosa M. When Humans Met Superbugs: Strategies to Tackle Bacterial Resistances to Antibiotics. Biomol Concepts 2018; 9:216-226. [PMID: 30811343 DOI: 10.1515/bmc-2018-0021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 12/17/2018] [Indexed: 12/25/2022] Open
Abstract
Bacterial resistance to antibiotics poses enormous health and economic burdens to our society, and it is of the essence to explore old and new ways to deal with these problems. Here we review the current status of multi-resistance genes and how they spread among bacteria. We discuss strategies to deal with resistant bacteria, namely the search for new targets and the use of inhibitors of protein-protein interactions, fragment-based methods, or modified antisense RNAs. Finally, we discuss integrated approaches that consider bacterial populations and their niches, as well as the role of global regulators that activate and/or repress the expression of multiple genes in fluctuating environments and, therefore, enable resistant bacteria to colonize new niches. Understanding how the global regulatory circuits work is, probably, the best way to tackle bacterial resistance.
Collapse
Affiliation(s)
- Alicia Bravo
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Sofia Ruiz-Cruz
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Itziar Alkorta
- Instituto BIOFISIKA (CSIC, UPV/EHU), Departamento de Bioquímica y Biología Molecular, Universidad del Pais Vasco, P.O. Box 644, 48080 Bilbao, Spain
| | - Manuel Espinosa
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, 9, 28040 Madrid, Spain
| |
Collapse
|
9
|
Gao C, Song Q, Zhang M, Li J, Yi M, Dong J. A method for expansion of T cells from cynomolgus monkey (Macaca fascicularis). In Vitro Cell Dev Biol Anim 2018; 54:549-554. [PMID: 30112696 DOI: 10.1007/s11626-018-0278-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/06/2018] [Indexed: 10/28/2022]
Abstract
T cells have been successfully applied to cancer immunotherapy. However, a challenge is an expansion of T cells from cynomolgus monkey, which is a valuable non-human primate model for T cell therapy transferring to the clinic. Here, we compared several strategies to expand cynomolgus monkey T cell and developed an appropriate method. Our study demonstrated that 100 ng/ml CD3 mAb + 1% PHA+ 1000 U/ml IL2 therapy significantly expanded peripheral blood CD3+ T cells without compromising T cell phenotype in vitro. The results of this study could be used for T cell remodeling, which has significant therapeutic potential in Chimeric Antigen Receptor-T (CAR-T) cell immunotherapy.
Collapse
Affiliation(s)
- Change Gao
- Department of Medical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Qian Song
- Department of Medical Oncology, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Kunming, 650032, Yunnan, China
| | - Ming Zhang
- Department of Radiotherapy, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jian Li
- Kunming Biological Diversity Regional Center of Instruments, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Miao Yi
- Department of Medical Oncology, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Kunming, 650032, Yunnan, China
| | - Jian Dong
- Department of Medical Oncology, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Kunming, 650032, Yunnan, China.
| |
Collapse
|
10
|
Yu M, Lu B, Liu Y, Me Y, Wang L, Zhang P. Tim-3 is upregulated in human colorectal carcinoma and associated with tumor progression. Mol Med Rep 2016; 15:689-695. [PMID: 28035413 PMCID: PMC5364832 DOI: 10.3892/mmr.2016.6065] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 08/16/2016] [Indexed: 12/28/2022] Open
Abstract
T cell immunoglobulin mucin-3 (Tim-3) has previously been implicated in the immune response and tumor biology. Colorectal carcinoma (CRC) is a malignancy, which is closely associated with inflammation. However, the role of Tim-3 in the progression of CRC remains to be fully elucidated. The present study aimed to investigate the role of Tim-3 in the progressive activities of human CRC. A total of 30 clinical CRC tissues and their adjacent tissues were collected. Slides from another 112 cases that underwent CRC surgical resection were also obtained. The protein and mRNA levels of Tim-3 in the clinical tissues and in CRC cell lines were initially examined using western blot and reverse transcription-quantitative polymerase chain reaction analyses, respectively. Immunohistochemical staining was performed to detect Tim-3 in the CRC samples. Specific small interfering (si)RNA against Tim-3 (siTim-3) was synthesized to knock down the expression of Tim-3, and the subsequent effects of Tim-3 knockdown on cell proliferation, migration and invasion were assessed. The data obtained showed that Tim-3 was expressed at high levels in the CRC tissues, compared with the non-cancerous tissues. The expression of Tim-3 in the clinical tissues was significantly associated with tumor size (P=0.007), tumor-node-metastasis staging (P<0.0001) and distant metastasis (P<0.0001). Knockdown of Tim-3 significantly reduced the cell proliferative rate of HCT116 and HT-29 cells. Wound closure activity was also inhibited by knockdown of Tim-3 in these two cell lines, and the migration and invasive abilities of these two cell lines were consistently decreased following knockdown of Tim-3. Taken together, Tim-3 was found to be a critical mediator in the progression of CRC and may serve as a potential therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Muming Yu
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin 300071, P.R. China
| | - Bin Lu
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin 300071, P.R. China
| | - Yancun Liu
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin 300071, P.R. China
| | - Ying Me
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin 300071, P.R. China
| | - Lijun Wang
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin 300071, P.R. China
| | - Peng Zhang
- School of Basic Medical Sciences, Medical Institution of Peking University, Beijing 100191, P.R. China
| |
Collapse
|
11
|
Heterologous Expression of Toxins from Bacterial Toxin-Antitoxin Systems in Eukaryotic Cells: Strategies and Applications. Toxins (Basel) 2016; 8:49. [PMID: 26907343 PMCID: PMC4773802 DOI: 10.3390/toxins8020049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 02/03/2016] [Accepted: 02/15/2016] [Indexed: 11/21/2022] Open
Abstract
Toxin-antitoxin (TA) systems are found in nearly all prokaryotic genomes and usually consist of a pair of co-transcribed genes, one of which encodes a stable toxin and the other, its cognate labile antitoxin. Certain environmental and physiological cues trigger the degradation of the antitoxin, causing activation of the toxin, leading either to the death or stasis of the host cell. TA systems have a variety of functions in the bacterial cell, including acting as mediators of programmed cell death, the induction of a dormant state known as persistence and the stable maintenance of plasmids and other mobile genetic elements. Some bacterial TA systems are functional when expressed in eukaryotic cells and this has led to several innovative applications, which are the subject of this review. Here, we look at how bacterial TA systems have been utilized for the genetic manipulation of yeasts and other eukaryotes, for the containment of genetically modified organisms, and for the engineering of high expression eukaryotic cell lines. We also examine how TA systems have been adopted as an important tool in developmental biology research for the ablation of specific cells and the potential for utility of TA systems in antiviral and anticancer gene therapies.
Collapse
|
12
|
Chan WT, Balsa D, Espinosa M. One cannot rule them all: Are bacterial toxins-antitoxins druggable? FEMS Microbiol Rev 2015; 39:522-40. [PMID: 25796610 PMCID: PMC4487406 DOI: 10.1093/femsre/fuv002] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2015] [Indexed: 01/31/2023] Open
Abstract
Type II (proteic) toxin–antitoxin (TA) operons are widely spread in bacteria and archaea. They are organized as operons in which, usually, the antitoxin gene precedes the cognate toxin gene. The antitoxin generally acts as a transcriptional self-repressor, whereas the toxin acts as a co-repressor, both proteins constituting a harmless complex. When bacteria encounter a stressful environment, TAs are triggered. The antitoxin protein is unstable and will be degraded by host proteases, releasing the free toxin to halt essential processes. The result is a cessation of cell growth or even death. Because of their ubiquity and the essential processes targeted, TAs have been proposed as good candidates for development of novel antimicrobials. We discuss here the possible druggability of TAs as antivirals and antibacterials, with focus on the potentials and the challenges that their use may find in the ‘real’ world. We present strategies to develop TAs as antibacterials in view of novel technologies, such as the use of very small molecules (fragments) as inhibitors of protein–protein interactions. Appropriate fragments could disrupt the T:A interfaces leading to the release of the targeted TA pair. Possible ways of delivery and formulation of Tas are also discussed. We consider various approaches to develop the toxins of the type II family as possible candidates to drug discovery; druggability of toxins-antitoxins could be possible as antivirals. As antibacterials, they might be considered as druggable but delivery and formulation may not be simple so far.
Collapse
Affiliation(s)
- Wai Ting Chan
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, 9, 28006-Madrid, Spain
| | - Dolors Balsa
- Immunology & Vaccines, Laboratorios LETI, Gran Via de les Corts Catalanes 184. 08034-Barcelona, Spain
| | - Manuel Espinosa
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, 9, 28006-Madrid, Spain
| |
Collapse
|
13
|
Saito N, Chono H, Shibata H, Ageyama N, Yasutomi Y, Mineno J. CD4(+) T Cells Modified by the Endoribonuclease MazF Are Safe and Can Persist in SHIV-infected Rhesus Macaques. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e168. [PMID: 24914931 PMCID: PMC4078760 DOI: 10.1038/mtna.2014.20] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/27/2014] [Indexed: 12/31/2022]
Abstract
MazF, an endoribonuclease encoded by Escherichia coli,
specifically cleaves the ACA (adenine–cytosine–adenine) sequence of
single-stranded RNAs. Conditional expression of MazF under the control of the HIV-1 LTR
promoter rendered CD4+ T cells resistant to HIV-1 replication without affecting
cell growth. To investigate the safety, persistence and efficacy of MazF-modified
CD4+ T cells in a nonhuman primate model in vivo,
rhesus macaques were infected with a pathogenic simian/human immunodeficiency virus (SHIV)
and transplanted with autologous MazF-modified CD4+ T cells. MazF-modified
CD4+ T cells were clearly detected throughout the experimental period of more
than 6 months. The CD4+ T cell count values increased in all four rhesus
macaques. Moreover, the transplantation of the MazF-modified CD4+ T cells was
not immunogenic, and did not elicit cellular or humoral immune responses. These data
suggest that the autologous transplantation of MazF-modified CD4+ T cells in
the presence of SHIV is effective, safe and not immunogenic, indicating that this is an
attractive strategy for HIV-1 gene therapy.
Collapse
Affiliation(s)
- Naoki Saito
- Center for Cell and Gene Therapy, Takara Bio Inc, Seta, Otsu, Shiga, Japan
| | - Hideto Chono
- Center for Cell and Gene Therapy, Takara Bio Inc, Seta, Otsu, Shiga, Japan
| | - Hiroaki Shibata
- Tsukuba Primate Research Center, National Institute of Biomedical Innovation, Tsukuba, Ibaraki, Japan
| | - Naohide Ageyama
- Tsukuba Primate Research Center, National Institute of Biomedical Innovation, Tsukuba, Ibaraki, Japan
| | - Yasuhiro Yasutomi
- Tsukuba Primate Research Center, National Institute of Biomedical Innovation, Tsukuba, Ibaraki, Japan
| | - Junichi Mineno
- Center for Cell and Gene Therapy, Takara Bio Inc, Seta, Otsu, Shiga, Japan
| |
Collapse
|
14
|
An efficient large-scale retroviral transduction method involving preloading the vector into a RetroNectin-coated bag with low-temperature shaking. PLoS One 2014; 9:e86275. [PMID: 24454964 PMCID: PMC3893289 DOI: 10.1371/journal.pone.0086275] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 12/10/2013] [Indexed: 01/15/2023] Open
Abstract
In retroviral vector-mediated gene transfer, transduction efficiency can be hampered by inhibitory molecules derived from the culture fluid of virus producer cell lines. To remove these inhibitory molecules to enable better gene transduction, we had previously developed a transduction method using a fibronectin fragment-coated vessel (i.e., the RetroNectin-bound virus transduction method). In the present study, we developed a method that combined RetroNectin-bound virus transduction with low-temperature shaking and applied this method in manufacturing autologous retroviral-engineered T cells for adoptive transfer gene therapy in a large-scale closed system. Retroviral vector was preloaded into a RetroNectin-coated bag and incubated at 4°C for 16 h on a reciprocating shaker at 50 rounds per minute. After the supernatant was removed, activated T cells were added to the bag. The bag transduction method has the advantage of increasing transduction efficiency, as simply flipping over the bag during gene transduction facilitates more efficient utilization of the retroviral vector adsorbed on the top and bottom surfaces of the bag. Finally, we performed validation runs of endoribonuclease MazF-modified CD4(+) T cell manufacturing for HIV-1 gene therapy and T cell receptor-modified T cell manufacturing for MAGE-A4 antigen-expressing cancer gene therapy and achieved over 200-fold (≥ 10(10)) and 100-fold (≥ 5 × 10(9)) expansion, respectively. In conclusion, we demonstrated that the large-scale closed transduction system is highly efficient for retroviral vector-based T cell manufacturing for adoptive transfer gene therapy, and this technology is expected to be amenable to automation and improve current clinical gene therapy protocols.
Collapse
|
15
|
Kurosaki M, Bolis M, Fratelli M, Barzago MM, Pattini L, Perretta G, Terao M, Garattini E. Structure and evolution of vertebrate aldehyde oxidases: from gene duplication to gene suppression. Cell Mol Life Sci 2013; 70:1807-30. [PMID: 23263164 PMCID: PMC11113236 DOI: 10.1007/s00018-012-1229-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 11/29/2012] [Accepted: 12/03/2012] [Indexed: 12/20/2022]
Abstract
Aldehyde oxidases (AOXs) and xanthine dehydrogenases (XDHs) belong to the family of molybdo-flavoenzymes. Although AOXs are not identifiable in fungi, these enzymes are represented in certain protists and the majority of plants and vertebrates. The physiological functions and substrates of AOXs are unknown. Nevertheless, AOXs are major drug metabolizing enzymes, oxidizing a wide range of aromatic aldehydes and heterocyclic compounds of medical/toxicological importance. Using genome sequencing data, we predict the structures of AOX genes and pseudogenes, reconstructing their evolution. Fishes are the most primitive organisms with an AOX gene (AOXα), originating from the duplication of an ancestral XDH. Further evolution of fishes resulted in the duplication of AOXα into AOXβ and successive pseudogenization of AOXα. AOXβ is maintained in amphibians and it is the likely precursors of reptilian, avian, and mammalian AOX1. Amphibian AOXγ is a duplication of AOXβ and the likely ancestor of reptilian and avian AOX2, which, in turn, gave rise to mammalian AOX3L1. Subsequent gene duplications generated the two mammalian genes, AOX3 and AOX4. The evolution of mammalian AOX genes is dominated by pseudogenization and deletion events. Our analysis is relevant from a structural point of view, as it provides information on the residues characterizing the three domains of each mammalian AOX isoenzyme. We cloned the cDNAs encoding the AOX proteins of guinea pig and cynomolgus monkeys, two unique species as to the evolution of this enzyme family. We identify chimeric RNAs from the human AOX3 and AOX3L1 pseudogenes with potential to encode a novel microRNA.
Collapse
Affiliation(s)
- Mami Kurosaki
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche “Mario Negri”, via La Masa 19, 20156 Milan, Italy
| | - Marco Bolis
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche “Mario Negri”, via La Masa 19, 20156 Milan, Italy
| | - Maddalena Fratelli
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche “Mario Negri”, via La Masa 19, 20156 Milan, Italy
| | - Maria Monica Barzago
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche “Mario Negri”, via La Masa 19, 20156 Milan, Italy
| | - Linda Pattini
- Department of Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy
| | - Gemma Perretta
- Istututo di Biologia Cellulare e Neurobiologia, Consiglio Nazionale delle Ricerche, via Anguillarese 301, 00123 Rome, Italy
| | - Mineko Terao
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche “Mario Negri”, via La Masa 19, 20156 Milan, Italy
| | - Enrico Garattini
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche “Mario Negri”, via La Masa 19, 20156 Milan, Italy
| |
Collapse
|
16
|
Okamoto M, Chono H, Kawano Y, Saito N, Tsuda H, Inoue K, Kato I, Mineno J, Baba M. Sustained Inhibition of HIV-1 Replication by Conditional Expression of theE. coli-Derived Endoribonuclease MazF in CD4+T cells. Hum Gene Ther Methods 2013; 24:94-103. [DOI: 10.1089/hgtb.2012.131] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mika Okamoto
- Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Hideto Chono
- Center for Cell and Gene Therapy, Takara Bio Inc., Otsu 520-2193, Japan
| | - Yasuhiro Kawano
- Center for Cell and Gene Therapy, Takara Bio Inc., Otsu 520-2193, Japan
| | - Naoki Saito
- Center for Cell and Gene Therapy, Takara Bio Inc., Otsu 520-2193, Japan
| | - Hiroshi Tsuda
- Center for Cell and Gene Therapy, Takara Bio Inc., Otsu 520-2193, Japan
| | - Koichi Inoue
- Center for Cell and Gene Therapy, Takara Bio Inc., Otsu 520-2193, Japan
| | - Ikunoshin Kato
- Center for Cell and Gene Therapy, Takara Bio Inc., Otsu 520-2193, Japan
| | - Junichi Mineno
- Center for Cell and Gene Therapy, Takara Bio Inc., Otsu 520-2193, Japan
| | - Masanori Baba
- Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| |
Collapse
|
17
|
Intramolecular regulation of the sequence-specific mRNA interferase activity of MazF fused to a MazE fragment with a linker cleavable by specific proteases. Appl Environ Microbiol 2012; 78:3794-9. [PMID: 22447587 DOI: 10.1128/aem.00364-12] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The genomes of human immunodeficiency virus type 1 (HIV-1) and hepatitis C virus (HCV) consist of single-stranded RNA encoding polyproteins, which are processed to individual functional proteins by virus-encoded specific proteases. These proteases have been used as targets for drug development. Here, instead of targeting these proteases to inhibit viral infection, we utilized the protease activity to activate a toxic protein to prevent viral infection. We engineered the MazE-MazF antitoxin-toxin system of Escherichia coli to fuse a C-terminal 41-residue fragment of antitoxin MazE to the N-terminal end of toxin MazF with a linker having a specific protease cleavage site for either HIV PR (HIV-1 protease), NS3 protease (HCV protease), or factor Xa. These fusion proteins formed a stable dimer (instead of the MazF(2)-MazE(2)-MazF(2) heterohexamer in nature) to inactivate the ACA (sequence)-specific mRNA interferase activity of MazF. When the fusion proteins were incubated with the corresponding proteases, the MazE fragment was cleaved from the fusion proteins, releasing active MazF, which then acted as an ACA-specific mRNA interferase cleaving single-stranded MS2 phage RNA. The intramolecular regulation of MazF toxicity by proteases as demonstrated may provide a novel approach for preventive and therapeutic treatments of infection by HIV-1, HCV, and other single-stranded RNA viruses.
Collapse
|