1
|
Liu Y, Lin Y, Zhu W. Systemic Effects of a Phage Cocktail on Healthy Weaned Piglets. BIOLOGY 2024; 13:271. [PMID: 38666883 PMCID: PMC11048100 DOI: 10.3390/biology13040271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024]
Abstract
Numerous studies have demonstrated that bacteriophages (phages) can effectively treat intestinal bacterial infections. However, research on the impact of phages on overall body health once they enter the intestine is limited. This study utilized weaned piglets as subjects to evaluate the systemic effects of an orally administered phage cocktail on their health. Twelve 21-day-old weaned piglets were divided into control (CON) and phage gavage (Phages) groups. The phage cocktail consisted of five lytic phages, targeting Salmonella enterica serovar Choleraesuis (S. choleraesuis), Enteropathogenic Escherichia coli (EPEC), and Shiga tox-in-producing Escherichia coli (STEC). The phages group received 10 mL of phage cocktail orally for 20 consecutive days. The results show that the phage gavage did not affect the piglets' growth performance, serum biochemical indices, or most organ indices, except for the pancreas. However, the impact on the intestine was complex. Firstly, although the pancreatic index decreased, it did not affect the secretion of digestive enzymes in the intestine. Secondly, phages increased the pH of jejunum chyme and relative weight of the ileum, and enhanced intestinal barrier function without affecting the morphology of the intestine. Thirdly, phages did not proliferate in the intestine, but altered the intestinal microbiota structure and increased concentrations of microbial metabolites isobutyric acid and isovaleric acid in the colonic chyme. In addition, phages impacted the immune status, significantly increasing serum IgA, IgG, and IgM, as well as serum and intestinal mucosal IFN-γ, IL-1β, IL-17, and TGF-β, and decreasing IL-4 and IL-10. They also activated toll-like receptors TLR-4 and TLR-9. Apart from an increase in basophil numbers, the counts of other immune cells in the blood did not change. This study indicates that the impact of phages on body health is complex, especially regarding immune status, warranting further attention. Short-term phage gavage did not have significant negative effects on health but could enhance intestinal barrier function.
Collapse
Affiliation(s)
- Yankun Liu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (W.Z.)
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Yan Lin
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (W.Z.)
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
- National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (W.Z.)
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
- National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
2
|
Sánchez Romano J, Simón-Santamaría J, McCourt P, Smedsrød B, Mortensen KE, Sagona AP, Sørensen KK, Larsen AK. Liver sinusoidal cells eliminate blood-borne phage K1F. mSphere 2024; 9:e0070223. [PMID: 38415633 PMCID: PMC10964407 DOI: 10.1128/msphere.00702-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/30/2024] [Indexed: 02/29/2024] Open
Abstract
Phage treatment has regained attention due to an increase in multiresistant bacteria. For phage therapy to be successful, phages must reach their target bacteria in sufficiently high numbers. Blood-borne phages are believed to be captured by macrophages in the liver and spleen. Since liver sinusoids also consist of specialized scavenger liver sinusoidal endothelial cells (LSECs) and Kupffer cells (KCs), this study investigated the contribution of both cell types in the elimination of Escherichia coli phage K1Fg10b::gfp (K1Fgfp) in mice. Circulatory half-life, organ, and hepatocellular distribution of K1Fgfp were determined following intravenous administration. Internalization of K1Fgfp and effects of phage opsonization on uptake were explored using primary mouse and human LSEC and KC cultures. When inoculated with 107 virions, >95% of the total K1Fgfp load was eliminated from the blood within 20 min, and 94% of the total retrieved K1Fgfp was localized to the liver. Higher doses resulted in slower elimination, possibly reflecting temporary saturation of liver scavenging capacity. Phage DNA was detected in both cell types, with a KC:LSEC ratio of 12:1 per population following cell isolation. Opsonization with plasma proteins increased time-dependent cellular uptake in both LSECs and KCs in vitro. Internalized phages were rapidly transported along the endocytic pathway to lysosomal compartments. Reduced viability of intracellular K1Fgfp corroborated inactivation following endocytosis. This study is the first to identify phage distribution in the liver at the hepatocellular level, confirming clearance of K1Fgfp performed mostly by KCs with a significant uptake also in LSECs.IMPORTANCEFaced with the increasing amounts of bacteria with multidrug antimicrobial resistance, phage therapy has regained attention as a possible treatment option. The phage field has recently experienced an emergence in commercial interest as research has identified new and more efficient ways of identifying and matching phages against resistant superbugs. Currently, phages are unapproved drugs in most parts of the world. For phages to reach broad clinical use, they must be shown to be clinically safe and useful. The results presented herein contribute to increased knowledge about the pharmacokinetics of the T7-like phage K1F in the mammalian system. The cell types of the liver that are responsible for rapid phage blood clearance are identified. Our results highlight the need for more research about appropriate dose regimens when phage therapy is delivered intravenously and advise essential knowledge about cell systems that should be investigated further for detailed phage pharmacodynamics.
Collapse
Affiliation(s)
| | | | - Peter McCourt
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Bård Smedsrød
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Kim Erlend Mortensen
- Gastrointestinal Surgery Unit, University Hospital of North Norway, Tromsø, Norway
| | - Antonia P. Sagona
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | | | - Anett Kristin Larsen
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
3
|
Bakhrushina E, Mikhel I, Kondratieva V, Demina N, Grebennikova T, Krasnyuk I, Krasnyuk I. Main Aspects of Pharmaceutical Development of In situ Immunobiological Drugs for Intranasal Administration. Curr Pharm Biotechnol 2024; 25:1394-1405. [PMID: 37842893 DOI: 10.2174/0113892010260017231002075152] [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] [Received: 04/28/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 10/17/2023]
Abstract
INTRODUCTION The review presents the latest developments in the area of intranasal in situ delivery systems of immunobiological drugs (IBDs). Interest in intranasal administration for IBDs has increased significantly due to the COVID-19 pandemic. However, not only intranasal delivery of vaccines is developing, but also bacteriophages, interferons, etc. In situ systems that make a selective phase transition can be a modern solution to intranasal delivery problems caused by mucociliary clearance. In addition, smart-polymers used as the main excipients in in situ systems can be used as specific adjuvants. METHODS A scientific search was conducted on the PubMed database of medical publications for the period from 2000 to 2022, using the keywords "intranasal in situ vaccine"; "intranasal in situ immunization". There were analyzed in detail more than 70 scientific studies on intranasal in situ delivery of IBDs. RESULTS AND CONCLUSIONS Despite the large number of new studies, the potential of possibilities of intranasal in situ systems is not being realized. Based on the results of the literature review an algorithm was created for the development of in situ systems for intranasal delivery of IBDs. Such algorithms and the methods of study design organization described in the review will help to facilitate the R&D process and bring the drug to commercial market, which will help to improve the quality of medical care.
Collapse
Affiliation(s)
- Elena Bakhrushina
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Iosif Mikhel
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Valeriya Kondratieva
- Centre of Epidemiology and Microbiology, The Gamaleya National Centre of Epidemiology and Microbiology, Moscow, Russian Federation
| | - Natalia Demina
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Tatyana Grebennikova
- Centre of Epidemiology and Microbiology, The Gamaleya National Centre of Epidemiology and Microbiology, Moscow, Russian Federation
| | - Ivan Krasnyuk
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Ivan Krasnyuk
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
4
|
Borg RE, Ozbakir HF, Xu B, Li E, Fang X, Peng H, Chen IA, Mukherjee A. Genetically engineered filamentous phage for bacterial detection using magnetic resonance imaging. SENSORS & DIAGNOSTICS 2023; 2:948-955. [PMID: 38405385 PMCID: PMC10888512 DOI: 10.1039/d3sd00026e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Detecting bacterial cells with high specificity in deep tissues is challenging. Optical probes provide specificity, but are limited by the scattering and absorption of light in biological tissues. Conversely, magnetic resonance imaging (MRI) allows unfettered access to deep tissues, but lacks contrast agents for detecting specific bacterial strains. Here, we introduce a biomolecular platform that combines both capabilities by exploiting the modularity of M13 phage to target bacteria with tunable specificity and allow deep-tissue imaging using T1-weighted MRI. We engineered two types of phage probes: one for detecting the phage's natural host, viz., F-pilus expressing E. coli; and the other for detecting a different (F-negative) bacterial target, V. cholerae. We show that these phage sensors generate 3-9-fold stronger T1 relaxation upon recognizing target cells relative to non-target bacteria. We further establish a preliminary proof-of-concept for in vivo applications, by demonstrating that phage-labeled bacteria can be detected in mice using MRI. The framework developed in this study may have potential utility in a broad range of applications, from basic biomedical research to in situ diagnostics, which require methods to detect and track specific bacteria in the context of intact living systems.
Collapse
Affiliation(s)
- Raymond E Borg
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
| | - Harun F Ozbakir
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Binzhi Xu
- Biomolecular Science and Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Eugene Li
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Xiwen Fang
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Huan Peng
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA
| | - Irene A Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA
| | - Arnab Mukherjee
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
- Biological Engineering, University of California, Santa Barbara, CA 93106, USA
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
5
|
Intranasal delivery of biotechnology-based therapeutics. Drug Discov Today 2022; 27:103371. [PMID: 36174965 DOI: 10.1016/j.drudis.2022.103371] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/09/2022] [Accepted: 09/21/2022] [Indexed: 11/21/2022]
Abstract
Biotechnology-based therapeutics include a wide range of products, such as recombinant hormones, stem cells, therapeutic enzymes, monoclonal antibodies, genes, vaccines, among others. The administration of these macromolecules has been studied via various routes. The nasal route is one of the promising routes of administration for biotechnology products owing to its easy delivery, the rich vascularity of the nasal mucosa, high absorption and targeted action. Several preclinical studies have been reported for nasal delivery of these products and many are at the clinical stage. This review focuses on biotechnology-based therapeutics administered via the intranasal route for treating various diseases.
Collapse
|
6
|
González-Mora A, Calvillo-Rodríguez KM, Hernández-Pérez J, Rito-Palomares M, Martínez-Torres AC, Benavides J. Evaluation of the Immune Response of a Candidate Phage-Based Vaccine against Rhipicephalus microplus (Cattle Tick). Pharmaceutics 2021; 13:pharmaceutics13122018. [PMID: 34959300 PMCID: PMC8706106 DOI: 10.3390/pharmaceutics13122018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
Cattle tick (Rhipicephalus microplus) represents a severe problem causing substantial economic losses, estimated in billions of dollars annually. Currently, chemical acaricides represent the most widely used control method. However, several problems such as resistance have been described. Phage-based vaccines represent a fast and low-cost tool for antigen delivery. In this regard, the objective of the present work was to develop a candidate phage-based vaccine displaying a cattle tick antigen (Bm86-derived Sbm7462 antigen) on the surface of bacteriophage M13. Phage ELISA and dot blotting analysis confirmed the display of the antigen. Vaccine immunogenicity was evaluated using a bovine monocyte-derived dendritic cell-based ex vivo assay and a murine in vivo assay. The ex vivo model showed the maturation of dendritic cells after being pulsed with the phage-based vaccine. The humoral response was confirmed in the in vivo assay. These results demonstrated the capacity of the phage-based vaccine to induce both humoral and cellular immune-specific responses. Importantly, this is the first report describing a control method for cattle ticks using a candidate phage-based vaccine. Further studies to evaluate the immunogenicity in a bovine model are needed. The current approach represents a promising alternative to control cattle tick infestations.
Collapse
Affiliation(s)
- Alejandro González-Mora
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L. CP, Mexico; (A.G.-M.); (J.H.-P.)
| | - Kenny Misael Calvillo-Rodríguez
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, N.L. CP, Mexico;
| | - Jesús Hernández-Pérez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L. CP, Mexico; (A.G.-M.); (J.H.-P.)
| | - Marco Rito-Palomares
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000 Pte, Monterrey 64710, N.L. CP, Mexico;
| | - Ana Carolina Martínez-Torres
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, N.L. CP, Mexico;
- Correspondence: (A.C.M.-T.); (J.B.); Tel.: +52-(81)-835294000 (ext. 6424) (A.C.M.-T.); +52-(81)-83582000 (ext. 4821) (J.B.)
| | - Jorge Benavides
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L. CP, Mexico; (A.G.-M.); (J.H.-P.)
- Correspondence: (A.C.M.-T.); (J.B.); Tel.: +52-(81)-835294000 (ext. 6424) (A.C.M.-T.); +52-(81)-83582000 (ext. 4821) (J.B.)
| |
Collapse
|
7
|
Modification of a Tumor-Targeting Bacteriophage for Potential Diagnostic Applications. Molecules 2021; 26:molecules26216564. [PMID: 34770973 PMCID: PMC8588016 DOI: 10.3390/molecules26216564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Tumor-targeting bacteriophages can be used as a versatile new platform for the delivery of diagnostic imaging agents and therapeutic cargo. This became possible due to the development of viral capsid modification method. Earlier in our laboratory and using phage display technology, phages to malignant breast cancer cells MDA-MB 231 were obtained. The goal of this study was the optimization of phage modification and the assessment of the effect of the latter on the efficiency of phage particle penetration into MDA-MB 231 cells. METHODS In this work, we used several methods, such as chemical phage modification using FAM-NHS ester, spectrophotometry, phage amplification, sequencing, phage titration, flow cytometry, and confocal microscopy. RESULTS We performed chemical phage modification using different concentrations of FAM-NHS dye (0.5 mM, 1 mM, 2 mM, 4 mM, 8 mM). It was shown that with an increase of the modification degree, the phage titer decreases. The maximum modification coefficient of the phage envelope with the FAM-NHS dye was observed with 4 mM modifying agent and had approximately 804,2 FAM molecules per phage. Through the immunofluorescence staining and flow cytometry methods, it was shown that the modified bacteriophage retains the ability to internalize into MDA-MB-231 cells. The estimation of the number of phages that could have penetrated into one tumor cell was conducted. CONCLUSIONS Optimizing the conditions for phage modification can be an effective strategy for producing tumor-targeting diagnostic and therapeutic agents, i.e., theranostic drugs.
Collapse
|
8
|
Podlacha M, Grabowski Ł, Kosznik-Kawśnicka K, Zdrojewska K, Stasiłojć M, Węgrzyn G, Węgrzyn A. Interactions of Bacteriophages with Animal and Human Organisms-Safety Issues in the Light of Phage Therapy. Int J Mol Sci 2021; 22:8937. [PMID: 34445641 PMCID: PMC8396182 DOI: 10.3390/ijms22168937] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022] Open
Abstract
Bacteriophages are viruses infecting bacterial cells. Since there is a lack of specific receptors for bacteriophages on eukaryotic cells, these viruses were for a long time considered to be neutral to animals and humans. However, studies of recent years provided clear evidence that bacteriophages can interact with eukaryotic cells, significantly influencing the functions of tissues, organs, and systems of mammals, including humans. In this review article, we summarize and discuss recent discoveries in the field of interactions of phages with animal and human organisms. Possibilities of penetration of bacteriophages into eukaryotic cells, tissues, and organs are discussed, and evidence of the effects of phages on functions of the immune system, respiratory system, central nervous system, gastrointestinal system, urinary tract, and reproductive system are presented and discussed. Modulations of cancer cells by bacteriophages are indicated. Direct and indirect effects of virulent and temperate phages are discussed. We conclude that interactions of bacteriophages with animal and human organisms are robust, and they must be taken under consideration when using these viruses in medicine, especially in phage therapy, and in biotechnological applications.
Collapse
Affiliation(s)
- Magdalena Podlacha
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Łukasz Grabowski
- Laboratory of Phage Therapy, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdansk, Poland; (Ł.G.); (K.K.-K.)
| | - Katarzyna Kosznik-Kawśnicka
- Laboratory of Phage Therapy, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdansk, Poland; (Ł.G.); (K.K.-K.)
| | - Karolina Zdrojewska
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Małgorzata Stasiłojć
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Alicja Węgrzyn
- Laboratory of Phage Therapy, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdansk, Poland; (Ł.G.); (K.K.-K.)
| |
Collapse
|
9
|
Nose-to-brain delivery: exploring newer domains for glioblastoma multiforme management. Drug Deliv Transl Res 2021; 10:1044-1056. [PMID: 32221847 DOI: 10.1007/s13346-020-00747-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive form of the primary brain tumors in humans. The intricate pathophysiology, the development of resistance by tumor cells, and the inability of the drugs to effectively cross the blood-brain and blood-tumor barriers result in poor prognosis for GBM patients, with a median survival time of only 1 to 2 years. Nose-to-brain delivery offers an attractive, noninvasive strategy to enhance drug penetration or transport novel drug/gene carriers into the brain. Although the exact mechanism of intranasal delivery remains elusive, the olfactory and trigeminal nerve pathways have been found to play a vital role in circumventing the traditional barriers of brain targeting. This review discusses the intranasal pathway as a novel domain for delivering drugs and nanocarriers encapsulating drugs/genes, as well as stem cell carriers specifically to the glioma cells. Considering the fact that most of these studies are still in preclinical stage, translating such intranasal delivery strategies from bench to bedside would be a critical step for better management and prognosis of GBM. Graphical abstract.
Collapse
|
10
|
Bichet MC, Chin WH, Richards W, Lin YW, Avellaneda-Franco L, Hernandez CA, Oddo A, Chernyavskiy O, Hilsenstein V, Neild A, Li J, Voelcker NH, Patwa R, Barr JJ. Bacteriophage uptake by mammalian cell layers represents a potential sink that may impact phage therapy. iScience 2021; 24:102287. [PMID: 33855278 PMCID: PMC8024918 DOI: 10.1016/j.isci.2021.102287] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/15/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
It is increasingly apparent that bacteriophages, viruses that infect bacteria and more commonly referred to as simply phages, have tropisms outside their bacterial hosts. Using live tissue culture cell imaging, we demonstrate that cell type, phage size, and morphology play a major role in phage internalization. Uptake was validated under physiological conditions using a microfluidic device. Phages adhered to mammalian tissues, with adherent phages being subsequently internalized by macropinocytosis, with functional phages accumulating intracellularly. We incorporated these results into a pharmacokinetic model demonstrating the potential impact of phage accumulation by cell layers, which represents a potential sink for circulating phages in the body. During phage therapy, high doses of phages are directly administered to a patient in order to treat a bacterial infection, thereby facilitating broad interactions between phages and mammalian cells. Understanding these interactions will have important implications on innate immune responses, phage pharmacokinetics, and the efficacy of phage therapy.
Collapse
Affiliation(s)
- Marion C. Bichet
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Wai Hoe Chin
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - William Richards
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Yu-Wei Lin
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Laura Avellaneda-Franco
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Catherine A. Hernandez
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Arianna Oddo
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, VIC, 3800, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, 3168, Australia
| | | | - Volker Hilsenstein
- Monash Micro Imaging, Monash University, Clayton Campus, Clayton, VIC, 3800, Australia
| | - Adrian Neild
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton Campus, Clayton, VIC 3800, Australia
| | - Jian Li
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Nicolas Hans Voelcker
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, VIC, 3800, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, 3168, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC 3168, Australia
| | - Ruzeen Patwa
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Jeremy J. Barr
- School of Biological Sciences, Monash University, Clayton Campus, 25 Rainforest Walk, Clayton, VIC, 3800, Australia
| |
Collapse
|
11
|
Goswami A, Sharma PR, Agarwal R. Combatting intracellular pathogens using bacteriophage delivery. Crit Rev Microbiol 2021; 47:461-478. [PMID: 33818246 DOI: 10.1080/1040841x.2021.1902266] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Intracellular pathogens reside in specialised compartments within the host cells restricting the access of antibiotics. Insufficient intracellular delivery of antibiotics along with several other resistance mechanisms weaken the efficacy of current therapies. An alternative to antibiotic therapy could be bacteriophage (phage) therapy. Although phage therapy has been in practice for a century against various bacterial infections, the efficacy of phages against intracellular bacteria is still being explored. In this review, we will discuss the advancement and challenges in phage therapy, particularly against intracellular bacterial pathogens. Finally, we will highlight the uptake mechanisms and approaches to overcome the challenges to phage therapy against intracellular bacteria.
Collapse
Affiliation(s)
- Avijit Goswami
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Pallavi Raj Sharma
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Rachit Agarwal
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
12
|
Żaczek M, Górski A, Skaradzińska A, Łusiak-Szelachowska M, Weber-Dąbrowska B. Phage penetration of eukaryotic cells: practical implications. Future Virol 2019. [DOI: 10.2217/fvl-2019-0110] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The inability to infect eukaryotic cells has been considered as the most undeniable feature of all bacterial viruses. Such specificity, limited only for bacterial hosts, raises questions about the paths and challenges phages should overcome when circulating through the human body. Recently, it has been shown that phages are able to continually penetrate human organs and tissues. Latest reports revealed that phages can cross eukaryotic cell barriers both para- and transcellularly and even reach the nucleus. Further, phages are capable of internalizing within cells through different endocytic mechanisms. Such phenomenon indicates that phages could shape human microbiome composition and affect all aspects of human health. Thus, herein, we summarize the current state of knowledge and describe this phenomenon with a particular emphasis on endocytic pathways.
Collapse
Affiliation(s)
- Maciej Żaczek
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
| | - Andrzej Górski
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
| | - Aneta Skaradzińska
- Department of Biotechnology & Food Microbiology, Faculty of Biotechnology & Food Science, Wrocław University of Environmental & Life Sciences, Chełmońskiego 37, 51-630 Wrocław, Poland
| | - Marzanna Łusiak-Szelachowska
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
| | - Beata Weber-Dąbrowska
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
| |
Collapse
|
13
|
Sartorius R, D'Apice L, Prisco A, De Berardinis P. Arming Filamentous Bacteriophage, a Nature-Made Nanoparticle, for New Vaccine and Immunotherapeutic Strategies. Pharmaceutics 2019; 11:E437. [PMID: 31480551 PMCID: PMC6781307 DOI: 10.3390/pharmaceutics11090437] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 12/17/2022] Open
Abstract
The pharmaceutical use of bacteriophages as safe and inexpensive therapeutic tools is collecting renewed interest. The use of lytic phages to fight antibiotic-resistant bacterial strains is pursued in academic and industrial projects and is the object of several clinical trials. On the other hand, filamentous bacteriophages used for the phage display technology can also have diagnostic and therapeutic applications. Filamentous bacteriophages are nature-made nanoparticles useful for their size, the capability to enter blood vessels, and the capacity of high-density antigen expression. In the last decades, our laboratory focused its efforts in the study of antigen delivery strategies based on the filamentous bacteriophage 'fd', able to trigger all arms of the immune response, with particular emphasis on the ability of the MHC class I restricted antigenic determinants displayed on phages to induce strong and protective cytotoxic responses. We showed that fd bacteriophages, engineered to target mouse dendritic cells (DCs), activate innate and adaptive responses without the need of exogenous adjuvants, and more recently, we described the display of immunologically active lipids. In this review, we will provide an overview of the reported applications of the bacteriophage carriers and describe the advantages of exploiting this technology for delivery strategies.
Collapse
Affiliation(s)
- Rossella Sartorius
- Institute of Biochemistry and Cell Biology (IBBC), 80131 CNR Naples, Italy
| | - Luciana D'Apice
- Institute of Biochemistry and Cell Biology (IBBC), 80131 CNR Naples, Italy.
| | - Antonella Prisco
- Institute of Genetics and Biophysics "A. Buzzati-Traverso" (IGB), 80131 CNR Naples, Italy
| | | |
Collapse
|
14
|
Samoylova TI, Braden TD, Spencer JA, Bartol FF. Immunocontraception: Filamentous Bacteriophage as a Platform for Vaccine Development. Curr Med Chem 2017; 24:3907-3920. [PMID: 28901276 PMCID: PMC5738698 DOI: 10.2174/0929867324666170911160426] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/19/2017] [Accepted: 08/23/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Population control of domestic, wild, invasive, and captive animal species is a global issue of importance to public health, animal welfare and the economy. There is pressing need for effective, safe, and inexpensive contraceptive technologies to address this problem. Contraceptive vaccines, designed to stimulate the immune system in order to block critical reproductive events and suppress fertility, may provide a solution. Filamentous bacteriophages can be used as platforms for development of such vaccines. OBJECTIVE In this review authors highlight structural and immunogenic properties of filamentous phages, and discuss applications of phage-peptide vaccines for advancement of immunocontraception technology in animals. RESULTS Phages can be engineered to display fusion (non-phage) peptides as coat proteins. Such modifications can be accomplished via genetic manipulation of phage DNA, or by chemical conjugation of synthetic peptides to phage surface proteins. Phage fusions with antigenic determinants induce humoral as well as cell-mediated immune responses in animals, making them attractive as vaccines. Additional advantages of the phage platform include environmental stability, low cost, and safety for immunized animals and those administering the vaccines. CONCLUSION Filamentous phages are viable platforms for vaccine development that can be engineered with molecular and organismal specificity. Phage-based vaccines can be produced in abundance at low cost, are environmentally stable, and are immunogenic when administered via multiple routes. These features are essential for a contraceptive vaccine to be operationally practical in animal applications. Adaptability of the phage platform also makes it attractive for design of human immunocontraceptive agents.
Collapse
Affiliation(s)
- Tatiana I Samoylova
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Timothy D Braden
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Jennifer A Spencer
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Frank F Bartol
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.,Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
15
|
Wang X, Wang L, Xu Y, Yu Q, Li L, Guo Y. Intranasal administration of Exendin-4 antagonizes Aβ31-35-induced disruption of circadian rhythm and impairment of learning and memory. Aging Clin Exp Res 2016; 28:1259-1266. [PMID: 26920423 DOI: 10.1007/s40520-016-0548-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/09/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND The deposition of β-amyloid protein (Aβ) is one of the pathological characteristics of Alzheimer's disease (AD) and can disrupt circadian rhythm and impair learning and memory. Exendin-4, a therapeutic drug for type II diabetes mellitus (T2DM), exerts neuroprotective effects from the toxicity of Aβ. However, it is not clear whether Exendin-4 protects against Aβ-induced disruption of circadian rhythm. The neuroprotective effects of Exendin-4 have been studied using injection of Exendin-4 into the lateral ventricle and abdomen. However, these procedures are not suitable for clinical application. METHODS First, male C57BL/6 mice received triple distilled water or Exendin-4 (0.1 nmol, 0.5 nmol) by intranasal administration. Exendin-4 levels were measured in the hippocampal samples using an ELISA Kit. Then, the study examined whether intranasal or hippocampal administration of Exendin-4 antagonized Aβ-induced disruption of circadian rhythm as well as impairment of learning and memory using the wheel-running activity assay and the Morris water maze test. RESULTS The study showed that intranasally administered Exendin-4 passed through the blood-brain barrier. Aβ31-35 given by intrahippocampal injection disrupted circadian rhythm and impaired learning and memory in C57BL/6 mice, and Exendin-4 given by nasal cavity or hippocampal administration ameliorated Aβ31-35-induced circadian rhythm disturbance of locomotor activity and impairment of learning and memory. CONCLUSIONS These findings provide pivotal experimental support for further study of the neuroprotective effects and clinical application of Exendin-4.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China.
| | - Li Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China
| | - Yunyun Xu
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China
| | - Qianqian Yu
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China
| | - Lin Li
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Yanlin Guo
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
16
|
Rueda F, Céspedes MV, Sánchez-Chardi A, Seras-Franzoso J, Pesarrodona M, Ferrer-Miralles N, Vázquez E, Rinas U, Unzueta U, Mamat U, Mangues R, García-Fruitós E, Villaverde A. Structural and functional features of self-assembling protein nanoparticles produced in endotoxin-free Escherichia coli. Microb Cell Fact 2016; 15:59. [PMID: 27059706 PMCID: PMC4826532 DOI: 10.1186/s12934-016-0457-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/28/2016] [Indexed: 11/10/2022] Open
Abstract
Background Production of recombinant drugs in process-friendly endotoxin-free bacterial factories targets to a lessened complexity of the purification process combined with minimized biological hazards during product application. The development of nanostructured recombinant materials in innovative nanomedical activities expands such a need beyond plain functional polypeptides to complex protein assemblies. While Escherichia coli has been recently modified for the production of endotoxin-free proteins, no data has been so far recorded regarding how the system performs in the fabrication of smart nanostructured materials. Results We have here explored the nanoarchitecture and in vitro and in vivo functionalities of CXCR4-targeted, self-assembling protein nanoparticles intended for intracellular delivery of drugs and imaging agents in colorectal cancer. Interestingly, endotoxin-free materials exhibit a distinguishable architecture and altered size and target cell penetrability than counterparts produced in conventional E. coli strains. These variant nanoparticles show an eventual proper biodistribution and highly specific and exclusive accumulation in tumor upon administration in colorectal cancer mice models, indicating a convenient display and function of the tumor homing peptides and high particle stability under physiological conditions. Discussion The observations made here support the emerging endotoxin-free E. coli system as a robust protein material producer but are also indicative of a particular conformational status and organization of either building blocks or oligomers. This appears to be promoted by multifactorial stress-inducing conditions upon engineering of the E. coli cell envelope, which impacts on the protein quality control of the cell factory.
Collapse
Affiliation(s)
- Fabián Rueda
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - María Virtudes Céspedes
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Biomedical Research Institute Sant Pau (IIB-SantPau) and Josep Carreras Leukemia Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
| | - Alejandro Sánchez-Chardi
- Servei de Microscòpia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Joaquin Seras-Franzoso
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Cibbim-Nanomedicine, Hospital Vall d'Hebron, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Mireia Pesarrodona
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Neus Ferrer-Miralles
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Ursula Rinas
- Leibniz University of Hannover, Technical Chemistry & Life Science, Hannover, Germany.,Helmholtz Centre for Infection Research, Inhoffenstraße 7, Brunswick, Germany
| | - Ugutz Unzueta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Biomedical Research Institute Sant Pau (IIB-SantPau) and Josep Carreras Leukemia Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
| | - Uwe Mamat
- Division of Structural Biochemistry, Priority Area Asthma and Allergy, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), 23845, Borstel, Germany
| | - Ramón Mangues
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Biomedical Research Institute Sant Pau (IIB-SantPau) and Josep Carreras Leukemia Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
| | - Elena García-Fruitós
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Torre Marimon, Caldes de Montbui, 08140, Barcelona, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain. .,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain. .,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.
| |
Collapse
|