1
|
Wu L, Chen Y, Yan Y, Wang H, Guy CD, Carney J, Moreno CL, Quintanilla-Arteaga A, Monsivais F, Zheng Z, Zeng M. Identification of Potential Therapeutic Targets Against Anthrax-Toxin-Induced Liver and Heart Damage. Toxins (Basel) 2025; 17:54. [PMID: 39998071 PMCID: PMC11861023 DOI: 10.3390/toxins17020054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/17/2025] [Accepted: 01/19/2025] [Indexed: 02/26/2025] Open
Abstract
Anthrax represents a disease resulting from infection by toxin-secreting bacteria, Bacillus anthracis. This research aimed to identify new therapeutic targets to combat anthrax. We performed assays to assess cell viability, apoptosis, glycogen consumption, and compound uptake and release in hepatocytes and cardiomyocytes responding to anthrax toxins. Microarray analysis was carried out to identify the genes potentially involved in toxin-induced toxicity. Knockdown experiments were performed to validate the contributions of the identified genes. Our study showed that anthrax edema toxin (EdTx) and lethal toxin (LeTx) induced lethal damage in mouse liver and heart, respectively. Microarray assays showed that 218 genes were potentially involved in EdTx-mediated toxicity, and 18 genes were potentially associated with LeTx-mediated toxicity. Among these genes, the knockdown of Rgs1, Hcar2, Fosl2, Hcar2, Cxcl2, and Cxcl3 protected primary hepatocytes from EdTx-induced cytotoxicity. Plasminogen activator inhibitor 1 (PAI-1)-encoding Serpine1 constituted the most significantly upregulated gene in response to LeTx treatment in mouse liver. PAI-1 knockout mouse models had a higher tolerance to LeTx compared with wild-type counterparts, suggesting that PAI-1 is essential for LeTx-induced toxicity and might represent a therapeutic target in LeTx-induced tissue damage. These results provide potential therapeutic targets for combating anthrax-toxin-induced liver and heart damage.
Collapse
Affiliation(s)
- Lihong Wu
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Yanping Chen
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Yongyong Yan
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Haiyan Wang
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Cynthia D. Guy
- Liver and GI Pathology Section, Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - John Carney
- Liver and GI Pathology Section, Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Carla L. Moreno
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Anaisa Quintanilla-Arteaga
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Fernando Monsivais
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Zhichao Zheng
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Mingtao Zeng
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
2
|
Metzger D, Miller K, Lyon W, Migliozzi R, Pangburn HA, Saldanha R. Host Cell Transcriptional Tuning with CRISPR/dCas9 to Mitigate the Effects of Toxin Exposure. ACS Synth Biol 2022; 11:3657-3668. [PMID: 36318971 DOI: 10.1021/acssynbio.2c00214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anthrax infection is caused byBacillus anthracis, a bacterium that once established within the host releases lethal toxin (LeTx). Anthrax LeTx is internalized by the capillary morphogenesis protein 2/anthrax toxin receptor 2 (CMG2/ANTXR2) cell surface receptor on mammalian cells. Once inside the cell, LeTx cleaves mitogen-activated protein kinases (MAPKs), ultimately leading to cell death. Previous reports have shown that decreased expression of ANTXR2 reduces cell susceptibility to LeTx. By ablating the ANTXR2 gene in cells in vitro, we observed complete resistance to LeTx-induced cell death. Here, we directed CRISPR/dCas9-based tools to the ANTXR2 promoter to modulate ANTXR2 expression without altering the underlying gene sequence in human cell lines that express the receptor at high levels. We hypothesized that downregulating the expression of the ANTXR2 gene at the genomic level would mitigate the impact of toxin exposure. In one epigenetic editing approach, we employed the fusion of DNMT3A DNA methyltransferase and dCas9 (dCas9-DNMT3A) to methylate CpGs within the CpG island of the ANTXR2 promoter and found this repressed ANTXR2 gene expression resulting in significant resistance to LeTx-induced cell death. Furthermore, by multiplexing gRNAs to direct dCas9-DNMT3A to multiple sites in the ANTXR2 promoter, we applied a broader distribution of CpG methylation along the gene promoter resulting in enhanced repression and resistance to LeTx. In parallel, we directed the dCas9-KRAB-MeCP2 transcriptional repressor to the ANTXR2 promoter to quickly and robustly repress ANTXR2 expression. With this approach, in as little as two weeks, we created resistance to LeTx at a similar level to ANTXR2 gene-ablated cells. Overall, we present a transcriptional tuning approach to inhibit the effects of LeTx and provide a framework to repress toxin-binding cell surface receptors.
Collapse
Affiliation(s)
- David Metzger
- UES, Inc., assigned to 711 Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, Ohio 45432, United States
| | - Kennedy Miller
- UES, Inc., assigned to 711 Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, Ohio 45432, United States
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania 16801, United States
| | - Wanda Lyon
- Airman Bioengineering Division, 711 Human Performance Wing, Air Force Research Lab, Wright-Patterson AFB, Ohio 45433, United States
| | - Rebecca Migliozzi
- UES, Inc., assigned to 711 Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, Ohio 45432, United States
| | - Heather A Pangburn
- Airman Bioengineering Division, 711 Human Performance Wing, Air Force Research Lab, Wright-Patterson AFB, Ohio 45433, United States
| | - Roland Saldanha
- Airman Bioengineering Division, 711 Human Performance Wing, Air Force Research Lab, Wright-Patterson AFB, Ohio 45433, United States
| |
Collapse
|
3
|
A new self-attenuated therapeutic influenza vaccine that uses host cell-restricted attenuation by artificial microRNAs. Int J Pharm 2022; 612:121325. [PMID: 34883209 PMCID: PMC8871448 DOI: 10.1016/j.ijpharm.2021.121325] [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: 08/20/2021] [Revised: 11/15/2021] [Accepted: 11/24/2021] [Indexed: 01/27/2023]
Abstract
New strategies are urgently needed for developing vaccines and/or anti-viral drugs against influenza viruses, because antigenic shift and drift inevitably occurs in circulating strains each year, and new strains resistant to anti-viral drugs have recently emerged. In our study, we designed and incorporated artificial microRNAs (amiRNAs) into the NA segment of rescued influenza viruses to separately target two host genes, Cdc2-like kinase 1 (CLK1) and SON DNA binding protein (SON), which were found to play an essential role in virus replication. Mouse epithelial fibroblast (MEF) or human lung carcinoma A549 cells infected with engineered influenza PR8 viruses expressing amiR-30CLK1 (PR8-amiR-30CLK1) or amiR-93SON (PR8-amiR-93SON) had reduced expression of host proteins CLK1 and SON, respectively. All engineered influenza viruses functioned as attenuated vaccines, induced significantly higher antibody responses, and provided greater protective efficacy. In addition, they were found to be safe, based on the mouse weight changes and clinical signs observed. In contrast to the engineered viruses targeting SON, mice treated with engineered viruses targeting CLK1 recovered from weight loss and survived lethal infection by 6 h after lethal-dose PR8 infection, suggesting that our PR8-amiR-30CLK1 self-attenuated influenza virus (SAIV) could be used as a new therapeutic influenza vaccine.
Collapse
|
4
|
Patel VI, Booth JL, Dozmorov M, Brown BR, Metcalf JP. Anthrax Edema and Lethal Toxins Differentially Target Human Lung and Blood Phagocytes. Toxins (Basel) 2020; 12:toxins12070464. [PMID: 32698436 PMCID: PMC7405021 DOI: 10.3390/toxins12070464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/26/2022] Open
Abstract
Bacillus anthracis, the causative agent of inhalation anthrax, is a serious concern as a bioterrorism weapon. The vegetative form produces two exotoxins: Lethal toxin (LT) and edema toxin (ET). We recently characterized and compared six human airway and alveolar-resident phagocyte (AARP) subsets at the transcriptional and functional levels. In this study, we examined the effects of LT and ET on these subsets and human leukocytes. AARPs and leukocytes do not express high levels of the toxin receptors, tumor endothelium marker-8 (TEM8) and capillary morphogenesis protein-2 (CMG2). Less than 20% expressed surface TEM8, while less than 15% expressed CMG2. All cell types bound or internalized protective antigen, the common component of the two toxins, in a dose-dependent manner. Most protective antigen was likely internalized via macropinocytosis. Cells were not sensitive to LT-induced apoptosis or necrosis at concentrations up to 1000 ng/mL. However, toxin exposure inhibited B. anthracis spore internalization. This inhibition was driven primarily by ET in AARPs and LT in leukocytes. These results support a model of inhalation anthrax in which spores germinate and produce toxins. ET inhibits pathogen phagocytosis by AARPs, allowing alveolar escape. In late-stage disease, LT inhibits phagocytosis by leukocytes, allowing bacterial replication in the bloodstream.
Collapse
Affiliation(s)
- Vineet I. Patel
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.I.P.); (J.L.B.); (B.R.B.)
| | - J. Leland Booth
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.I.P.); (J.L.B.); (B.R.B.)
| | - Mikhail Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Brent R. Brown
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.I.P.); (J.L.B.); (B.R.B.)
| | - Jordan P. Metcalf
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.I.P.); (J.L.B.); (B.R.B.)
- Department of Microbiology and Immunology, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
- Correspondence:
| |
Collapse
|
5
|
Xu H, Qu Y. Protective effect of anthrax toxin receptor 2 polymorphism rs4333130 against the risk of ankylosing spondylitis. Medicine (Baltimore) 2020; 99:e19942. [PMID: 32664053 PMCID: PMC7360223 DOI: 10.1097/md.0000000000019942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The present study was performed to statistically explore the effect of anthrax toxin receptor 2 (ANTXR2) polymorphism rs4333130 on individual susceptibility to ankylosing spondylitis (AS) using the method of meta-analysis. METHODS All of the eligible reports were retrieved from well-known electronic databases. The strength of the association between ANTXR2 polymorphism rs4333130 and the susceptibility to AS was evaluated using pooled odds ratios (ORs) with 95% confidence intervals (95% CIs). In addition, subgroup analysis was also performed on the basis of ethnicity to further explore specific correlation between our studied polymorphism and the disease risk. Inter-study heterogeneity was detected with Q test, and P < .05 was considered statistically significant. Sensitivity analysis was implemented through removing each of eligible studies and then recalculating overall effects to test the reliability of final estimates. Publication bias among included studies was inspected with both Begg funnel plot and Egger regression test. RESULTS A total of 6 eligible papers were finally incorporated into the present meta-analysis. In total analysis, ANTXR2 polymorphism rs4333130 was significantly related to decreased risk of AS under CC versus TT, CC + TC versus TT, CC versus TT + TC, C versus T and TC versus TT contrasts (OR = 0.35, 95% CI = 0.20-0.64; OR = 0.81, 95% CI = 0.69-0.95; OR = 0.38, 95% CI = 0.21-0.68; OR = 0.89, 95% CI = 0.84-0.95; OR = 0.84, 95% CI = 0.72-0.99). Moreover, a similar effect was also observed in Asian and Caucasian subgroups under corresponding genetic models after stratification analysis based on ethnicity. CONCLUSION ANTXR2 polymorphism rs4333130 may function as a protective factor against AS incidence.
Collapse
|
6
|
Ayanoğlu FB, Elçin AE, Elçin YM. Bioethical issues in genome editing by CRISPR-Cas9 technology. ACTA ACUST UNITED AC 2020; 44:110-120. [PMID: 32256147 PMCID: PMC7129066 DOI: 10.3906/biy-1912-52] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Genome editing technologies have led to fundamental changes in genetic science. Among them, CRISPR-Cas9 technology particularly stands out due to its advantages such as easy handling, high accuracy, and low cost. It has made a quick introduction in fields related to humans, animals, and the environment, while raising difficult questions, applications, concerns, and bioethical issues to be discussed. Most concerns stem from the use of CRISPR-Cas9 to genetically alter human germline cells and embryos (called germline genome editing). Germline genome editing leads to serial bioethical issues, such as the occurrence of undesirable changes in the genome, from whom and how informed consent is obtained, and the breeding of the human species (eugenics). However, the bioethical issues that CRISPR-Cas9 technology could cause in the environment, agriculture and livestock should also not be forgotten. In order for CRISPR-Cas9 to be used safely in all areas and to solve potential issues, worldwide legislation should be prepared, taking into account the opinions of both life and social scientists, policy makers, and all other stakeholders of the sectors, and CRISPR-Cas9 applications should be implemented according to such legislations. However, these controls should not restrict scientific freedom. Here, various applications of CRISPR-Cas9 technology, especially in medicine and agriculture, are described and ethical issues related to genome editing using CRISPR-Cas9 technology are discussed. The social and bioethical concerns in relation to human beings, other organisms, and the environment are addressed.
Collapse
Affiliation(s)
- Fatma Betül Ayanoğlu
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science,Ankara University Biotechnology Institute, Ankara University Stem Cell Institute, Ankara Turkey
| | - Ayşe Eser Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science,Ankara University Biotechnology Institute, Ankara University Stem Cell Institute, Ankara Turkey
| | - Yaşar Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science,Ankara University Biotechnology Institute, Ankara University Stem Cell Institute, Ankara Turkey.,Biovalda Health Technologies, Inc., Ankara Turkey
| |
Collapse
|
7
|
Flori L, Moazami-Goudarzi K, Alary V, Araba A, Boujenane I, Boushaba N, Casabianca F, Casu S, Ciampolini R, Coeur D'Acier A, Coquelle C, Delgado JV, El-Beltagi A, Hadjipavlou G, Jousselin E, Landi V, Lauvie A, Lecomte P, Ligda C, Marinthe C, Martinez A, Mastrangelo S, Menni D, Moulin CH, Osman MA, Pineau O, Portolano B, Rodellar C, Saïdi-Mehtar N, Sechi T, Sempéré G, Thévenon S, Tsiokos D, Laloë D, Gautier M. A genomic map of climate adaptation in Mediterranean cattle breeds. Mol Ecol 2019; 28:1009-1029. [PMID: 30593690 DOI: 10.1111/mec.15004] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Domestic species such as cattle (Bos taurus taurus and B. t. indicus) represent attractive biological models to characterize the genetic basis of short-term evolutionary response to climate pressure induced by their post-domestication history. Here, using newly generated dense SNP genotyping data, we assessed the structuring of genetic diversity of 21 autochtonous cattle breeds from the whole Mediterranean basin and performed genome-wide association analyses with covariables discriminating the different Mediterranean climate subtypes. This provided insights into both the demographic and adaptive histories of Mediterranean cattle. In particular, a detailed functional annotation of genes surrounding variants associated with climate variations highlighted several biological functions involved in Mediterranean climate adaptation such as thermotolerance, UV protection, pathogen resistance or metabolism with strong candidate genes identified (e.g., NDUFB3, FBN1, METTL3, LEF1, ANTXR2 and TCF7). Accordingly, our results suggest that main selective pressures affecting cattle in Mediterranean area may have been related to variation in heat and UV exposure, in food resources availability and in exposure to pathogens, such as anthrax bacteria (Bacillus anthracis). Furthermore, the observed contribution of the three main bovine ancestries (indicine, European and African taurine) in these different populations suggested that adaptation to local climate conditions may have either relied on standing genomic variation of taurine origin, or adaptive introgression from indicine origin, depending on the local breed origins. Taken together, our results highlight the genetic uniqueness of local Mediterranean cattle breeds and strongly support conservation of these populations.
Collapse
Affiliation(s)
- Laurence Flori
- SELMET, INRA, CIRAD, University of Montpellier, Montpellier SupAgro, University of Montpellier, Montpellier, France
| | | | - Véronique Alary
- SELMET, INRA, CIRAD, University of Montpellier, Montpellier SupAgro, University of Montpellier, Montpellier, France.,CIRAD, UMR SELMET, ICARDA, Rabat, Morocco
| | - Abdelillah Araba
- Institut Agronomique et Vétérinaire Hassan II, Département de Productions et de Biotechnologies Animales, Rabat, Morocco
| | - Ismaïl Boujenane
- Institut Agronomique et Vétérinaire Hassan II, Département de Productions et de Biotechnologies Animales, Rabat, Morocco
| | - Nadjet Boushaba
- Université d'Oran "Mohamed Boudiaf", Département de Génétique Moléculaire Appliquée, Oran, Algeria
| | | | - Sara Casu
- Agris-Sardegna Servizio Ricerca per la Zootecnica, Olmedo, Italy
| | | | | | | | | | | | | | | | - Vincenzo Landi
- Animal Breeding Consulting SL, Laboratorio de Genetica Molecular Aplicada, Cordoba, Spain
| | - Anne Lauvie
- SELMET, INRA, CIRAD, University of Montpellier, Montpellier SupAgro, University of Montpellier, Montpellier, France
| | - Philippe Lecomte
- SELMET, INRA, CIRAD, University of Montpellier, Montpellier SupAgro, University of Montpellier, Montpellier, France.,CIRAD, UMR SELMET, Montpellier, France
| | - Christina Ligda
- HAO-Demeter, Veterinary Research Institute, Thessaloniki, Greece
| | | | - Amparo Martinez
- Animal Breeding Consulting SL, Laboratorio de Genetica Molecular Aplicada, Cordoba, Spain
| | - Salvatore Mastrangelo
- Dipartimento Scienze Agrarie, Alimentari e Forestali, Università degli Studi di Palermo, Palermo, Italy
| | - Dalal Menni
- Institut Agronomique et Vétérinaire Hassan II, Département de Productions et de Biotechnologies Animales, Rabat, Morocco
| | - Charles-Henri Moulin
- SELMET, INRA, CIRAD, University of Montpellier, Montpellier SupAgro, University of Montpellier, Montpellier, France
| | | | | | - Baldassare Portolano
- Dipartimento Scienze Agrarie, Alimentari e Forestali, Università degli Studi di Palermo, Palermo, Italy
| | - Clementina Rodellar
- LAGENBIO, Facultad de Veterinaria, Instituto Agroalimentario de Aragón-IA2, Universidad de Zaragoza-CITA, Zaragoza, Spain
| | - Nadhira Saïdi-Mehtar
- Université d'Oran "Mohamed Boudiaf", Département de Génétique Moléculaire Appliquée, Oran, Algeria
| | - Tiziana Sechi
- Agris-Sardegna Servizio Ricerca per la Zootecnica, Olmedo, Italy
| | - Guilhem Sempéré
- INTERTRYP, University of Montpellier, CIRAD, IRD, Montpellier, France.,CIRAD, UMR INTERTRYP, Montpellier, France
| | - Sophie Thévenon
- INTERTRYP, University of Montpellier, CIRAD, IRD, Montpellier, France.,CIRAD, UMR INTERTRYP, Montpellier, France
| | | | - Denis Laloë
- GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Mathieu Gautier
- CBGP, INRA, CIRAD, IRD, University of Montpellier, Montferrier-sur-Lez, France.,Institut de Biologie Computationnelle (IBC), Montpellier, France
| |
Collapse
|
8
|
Greene M, Master Z. Ethical Issues of Using CRISPR Technologies for Research on Military Enhancement. JOURNAL OF BIOETHICAL INQUIRY 2018; 15:327-335. [PMID: 29968018 DOI: 10.1007/s11673-018-9865-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 05/07/2018] [Indexed: 06/08/2023]
Abstract
This paper presents an overview of the key ethical questions of performing gene editing research on military service members. The recent technological advance in gene editing capabilities provided by CRISPR/Cas9 and their path towards first-in-human trials has reinvigorated the debate on human enhancement for non-medical purposes. Human performance optimization has long been a priority of military research in order to close the gap between the advancement of warfare and the limitations of human actors. In spite of this focus on temporary performance improvement, biomedical enhancement is an extension of these endeavours and the ethical issues of such research should be considered. In this paper, we explore possible applications of CRISPR to military human gene editing research and how it could be specifically applied towards protection of service members against biological or chemical weapons. We analyse three normative areas including risk-benefit analysis, informed consent, and inequality of access as it relates to CRISPR applications for military research to help inform and provide considerations for military institutional review boards and policymakers.
Collapse
Affiliation(s)
- Marsha Greene
- Senior Policy Analyst, Peraton, Contracted to the Office of the Under Secretary of Defense for Research and Engineering, 4800 Mark Center Drive, Suite 17E08, Alexandria, VA, 22350-3600, USA
| | - Zubin Master
- Biomedical Ethics Research Program, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
9
|
Bacillus anthracis Edema Toxin Increases Fractional Free Water and Sodium Reabsorption in an Isolated Perfused Rat Kidney Model. Infect Immun 2017; 85:IAI.00264-17. [PMID: 28438974 DOI: 10.1128/iai.00264-17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 04/13/2017] [Indexed: 11/20/2022] Open
Abstract
Bacillus anthracis edema toxin (ET) consists of protective antigen (PA), necessary for host cell toxin uptake, and edema factor (EF), the toxic moiety which increases host cell cyclic AMP (cAMP). Since vasopressin stimulates renal water and sodium reabsorption via increased tubular cell cAMP levels, we hypothesized the ET would also do so. To test this hypothesis, we employed an isolated perfused rat kidney model. Kidneys were isolated and perfused with modified Krebs-Henseleit buffer. Perfusate and urine samples were obtained at baseline and every 10 min over 150 min following the addition of challenges with or without treatments to the perfusate. In kidneys perfused under constant flow or constant pressure, compared to PA challenge (n = 14 or 15 kidneys, respectively), ET (13 or 15 kidneys, respectively) progressively increased urine cAMP levels, water and sodium reabsorption, and urine osmolality and decreased urine output (P ≤ 0.04, except for sodium reabsorption under constant pressure [P = 0.17]). In ET-challenged kidneys, compared to placebo treatment, adefovir, an EF inhibitor, decreased urine cAMP levels, water and sodium reabsorption, and urine osmolality and increased urine output, while raxibacumab, a PA-directed monoclonal antibody (MAb), decreased urine cAMP levels, free water reabsorption, and urine osmolality and increased urine output (P ≤ 0.03 except for urine output with raxibacumab [P = 0.17]). Upon immunohistochemistry, aquaporin 2 was concentrated along the apical membrane of tubular cells with ET but not PA, and urine aquaporin 2 levels were higher with ET (5.52 ± 1.06 ng/ml versus 1.51 ± 0.44 ng/ml [means ± standard errors of the means {SEM}; P = 0.0001). Edema toxin has renal effects that could contribute to extravascular fluid collection characterizing anthrax infection clinically.
Collapse
|
10
|
Liu X, Yuan W, Li J, Yang L, Cai J. ANTXR2 Knock-Out Does Not Result in the Development of Hypertension in Rats. Am J Hypertens 2017; 30:182-187. [PMID: 28077422 DOI: 10.1093/ajh/hpw125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 09/13/2016] [Accepted: 09/15/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Our recent genetic study as well as robust evidences reported by previous genome-wide association studies (GWASs) have indicated that the single nucleotide polymorphism rs16998073, located near gene anthrax toxin receptor 2 (ANTXR2), was significantly associated with hypertension in Asians and Europeans. The aim of the present study was to determine whether ANTXR2 is the causal gene of hypertension at the 4q21 locus using an ANTXR2 knock-out model. METHODS Relative expression of ANTXR2 in Wistar-Kyoto rats (WKYs) and spontaneously hypertensive rats (SHRs) were determined by real-time quantitative polymerase chain reaction and western blot analysis. ANTXR2 knock-out rats were created using CRISPR/Cas9-mediated genome editing and blood pressure values were measured in ANTXR2-/- and wild type (WT) rats by tail-cuff method and carotid arterial catheterization method. RESULTS Neither the mRNA nor protein levels of ANTXR2 were significantly different between tissues from SHRs and WKYs. To create ANTXR2-/- rats, 67 base pairs were deleted in exon 1 of ANTXR2 using CRISPR/Cas9-mediated genome editing. ANTXR2 protein decreased significantly in aortas of ANTXR2-/- rats, suggesting sufficient efficiency of ANTXR2 knock-out in this model. However, ANTXR2-/- rats exhibited nearly the same blood pressure as WT rats at baseline conditions as well as during Angiotensin II (400ng/kg/min) infusion or high-salt diet treatment. CONCLUSIONS These findings suggest that ANTXR2 might not be associated with hypertension and thus further functional analysis is warranted to identify the causal gene at this locus.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wen Yuan
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jing Li
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypertension, Beijing, China
| | - Lei Yang
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jun Cai
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China;
- Beijing Key Laboratory of Hypertension, Beijing, China
| |
Collapse
|
11
|
Arévalo MT, Li J, Diaz-Arévalo D, Chen Y, Navarro A, Wu L, Yan Y, Zeng M. A dual purpose universal influenza vaccine candidate confers protective immunity against anthrax. Immunology 2016; 150:276-289. [PMID: 27775159 DOI: 10.1111/imm.12683] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/17/2016] [Accepted: 10/20/2016] [Indexed: 01/08/2023] Open
Abstract
Preventive influenza vaccines must be reformulated annually because of antigen shift and drift of circulating influenza viral strains. However, seasonal vaccines do not always match the circulating strains, and there is the ever-present threat that avian influenza viruses may adapt to humans. Hence, a universal influenza vaccine is needed to provide protective immunity against a broad range of influenza viruses. We designed an influenza antigen consisting of three tandem M2e repeats plus HA2, in combination with a detoxified anthrax oedema toxin delivery system (EFn plus PA) to enhance immune responses. The EFn-3×M2e-HA2 plus PA vaccine formulation elicited robust, antigen-specific, IgG responses; and was protective against heterologous influenza viral challenge when intranasally delivered to mice three times. Moreover, use of the detoxified anthrax toxin system as an adjuvant had the additional benefit of generating protective immunity against anthrax. Hence, this novel vaccine strategy could potentially address two major emerging public health and biodefence threats.
Collapse
Affiliation(s)
- Maria T Arévalo
- Center of Emphasis in Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| | - Junwei Li
- Center of Emphasis in Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| | - Diana Diaz-Arévalo
- Center of Emphasis in Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| | - Yanping Chen
- Center of Emphasis in Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| | - Ashley Navarro
- Center of Emphasis in Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| | - Lihong Wu
- Center of Emphasis in Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA.,Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yongyong Yan
- Center of Emphasis in Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA.,Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mingtao Zeng
- Center of Emphasis in Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| |
Collapse
|
12
|
Sun J, Jacquez P. Roles of Anthrax Toxin Receptor 2 in Anthrax Toxin Membrane Insertion and Pore Formation. Toxins (Basel) 2016; 8:34. [PMID: 26805886 PMCID: PMC4773787 DOI: 10.3390/toxins8020034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 01/18/2016] [Accepted: 01/19/2016] [Indexed: 12/04/2022] Open
Abstract
Interaction between bacterial toxins and cellular surface receptors is an important component of the host-pathogen interaction. Anthrax toxin protective antigen (PA) binds to the cell surface receptor, enters the cell through receptor-mediated endocytosis, and forms a pore on the endosomal membrane that translocates toxin enzymes into the cytosol of the host cell. As the major receptor for anthrax toxin in vivo, anthrax toxin receptor 2 (ANTXR2) plays an essential role in anthrax toxin action by providing the toxin with a high-affinity binding anchor on the cell membrane and a path of entry into the host cell. ANTXR2 also acts as a molecular clamp by shifting the pH threshold of PA pore formation to a more acidic pH range, which prevents premature pore formation at neutral pH before the toxin reaches the designated intracellular location. Most recent studies have suggested that the disulfide bond in the immunoglobulin (Ig)-like domain of ANTXR2 plays an essential role in anthrax toxin action. Here we will review the roles of ANTXR2 in anthrax toxin action, with an emphasis on newly updated knowledge.
Collapse
Affiliation(s)
- Jianjun Sun
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA.
| | - Pedro Jacquez
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA.
| |
Collapse
|
13
|
Ohanjanian L, Remy KE, Li Y, Cui X, Eichacker PQ. An overview of investigational toxin-directed therapies for the adjunctive management of Bacillus anthracis infection and sepsis. Expert Opin Investig Drugs 2015; 24:851-65. [PMID: 25920540 DOI: 10.1517/13543784.2015.1041587] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Sepsis with Bacillus anthracis infection has a very high mortality rate despite appropriate antibiotic and supportive therapies. Over the past 15 years, recent outbreaks in the US and in Europe, coupled with anthrax's bioterrorism weapon potential, have stimulated efforts to develop adjunctive therapies to improve clinical outcomes. Since lethal toxin and edema toxin (LT and ET) make central contributions to the pathogenesis of B. anthracis, these have been major targets in this effort. AREAS COVERED Here, the authors review different investigative biopharmaceuticals that have been recently identified for their therapeutic potential as inhibitors of LT or ET. Among these inhibitors are two antibody preparations that have been included in the Strategic National Stockpile (SNS) and several more that have reached Phase I testing. Presently, however, many of these candidate agents have only been studied in vitro and very few tested in bacteria-challenged models. EXPERT OPINION Although a large number of drugs have been identified as potential therapeutic inhibitors of LT and ET, in most cases their testing has been limited. The use of the two SNS antibody therapies during a large-scale exposure to B. anthracis will be difficult. Further testing and development of agents with oral bioavailability and relatively long shelf lives should be a focus for future research.
Collapse
Affiliation(s)
- Lernik Ohanjanian
- National Institutes of Health, Clinical Center, Critical Care Medicine Department , Building 10, Room 2C145, Bethesda, MD 20892 , USA +1 301 402 2914 ; +1 301 402 1213 ;
| | | | | | | | | |
Collapse
|