1
|
Jia J, Wang X, Lin X, Zhao Y. Engineered Microorganisms for Advancing Tumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313389. [PMID: 38485221 DOI: 10.1002/adma.202313389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Engineered microorganisms have attracted significant interest as a unique therapeutic platform in tumor treatment. Compared with conventional cancer treatment strategies, engineering microorganism-based systems provide various distinct advantages, such as the intrinsic capability in targeting tumors, their inherent immunogenicity, in situ production of antitumor agents, and multiple synergistic functions to fight against tumors. Herein, the design, preparation, and application of the engineered microorganisms for advanced tumor therapy are thoroughly reviewed. This review presents a comprehensive survey of innovative tumor therapeutic strategies based on a series of representative engineered microorganisms, including bacteria, viruses, microalgae, and fungi. Specifically, it offers extensive analyses of the design principles, engineering strategies, and tumor therapeutic mechanisms, as well as the advantages and limitations of different engineered microorganism-based systems. Finally, the current challenges and future research prospects in this field, which can inspire new ideas for the design of creative tumor therapy paradigms utilizing engineered microorganisms and facilitate their clinical applications, are discussed.
Collapse
Affiliation(s)
- Jinxuan Jia
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiaocheng Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Xiang Lin
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Yuanjin Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| |
Collapse
|
2
|
Marquez CA, Oh CI, Ahn G, Shin WR, Kim YH, Ahn JY. Synergistic vesicle-vector systems for targeted delivery. J Nanobiotechnology 2024; 22:6. [PMID: 38167116 PMCID: PMC10763086 DOI: 10.1186/s12951-023-02275-6] [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: 07/28/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
With the immense progress in drug delivery systems (DDS) and the rise of nanotechnology, challenges such as target specificity remain. The vesicle-vector system (VVS) is a delivery system that uses lipid-based vesicles as vectors for a targeted drug delivery. When modified with target-probing materials, these vesicles become powerful vectors for drug delivery with high target specificity. In this review, we discuss three general types of VVS based on different modification strategies: (1) vesicle-probes; (2) vesicle-vesicles; and (3) genetically engineered vesicles. The synthesis of each VVS type and their corresponding properties that are advantageous for targeted drug delivery, are also highlighted. The applications, challenges, and limitations of VVS are briefly examined. Finally, we share a number of insights and perspectives regarding the future of VVS as a targeted drug delivery system at the nanoscale.
Collapse
Affiliation(s)
- Christine Ardelle Marquez
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Cho-Im Oh
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Gna Ahn
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Woo-Ri Shin
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
- Department of Bioengineering, University of Pennsylvania, 210 S 33rd St, Philadelphia, PA, 19104, USA
| | - Yang-Hoon Kim
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea.
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| | - Ji-Young Ahn
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea.
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
3
|
Cho Y, Kim JH, Choi W, Park DY, Cho BK, Kim YH, Min J. Reassembled Vacuoles for Drug Delivery Carriers Using Yeast Vacuoles for Enhanced Antibacterial Activity. Biomacromolecules 2023; 24:4915-4922. [PMID: 37861681 DOI: 10.1021/acs.biomac.3c00616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
In this study, we aimed to develop an efficient drug delivery system by reassembling vacuoles isolated from Saccharomyces cerevisiae. Initially, we assessed the impact of vacuolar enzymes on the efficacy of the loaded antibiotic polymyxin B (PMB), by conducting antibacterial activity tests using Shigella flexneri and Salmonella enteritidis. The results showed that vacuolar enzymes inhibited the effectiveness of PMB, highlighting the limitations of using natural vacuoles as drug carriers. To overcome this, we proposed a new drug delivery system called reassembled vacuoles (ReV). ReV particles were created by removing vacuolar enzymes and reassembling the vacuolar membrane through extrusion. ReV demonstrated improved structural stability, a more uniform size, and enhanced PMB release compared to natural vacuoles. Encapsulation efficiency tests revealed high loading efficiency for both normal vacuoles (NorV) and ReV, with over 80% efficiency at concentrations up to 600 μg/mL. The antibacterial activity of PMB-loaded ReV showed comparable results to PMB alone, indicating the potential of ReV as a drug delivery system. In conclusion, reassembled vacuoles offer a promising approach for drug delivery, addressing the limitations of natural vacuoles and providing opportunities for targeted and efficient drug release.
Collapse
Affiliation(s)
- Yunyoung Cho
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk 54896, South Korea
| | - Ji Hun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-Gu, Daejeon 34141, South Korea
| | - Wooil Choi
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk 54896, South Korea
| | - Dae-Young Park
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju 28644, South Korea
| | - Byung-Kwan Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-Gu, Daejeon 34141, South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju 28644, South Korea
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk 54896, South Korea
| |
Collapse
|
4
|
Hu J, Liu Y, Du Y, Peng X, Liu Z. Cellular organelles as drug carriers for disease treatment. J Control Release 2023; 363:114-135. [PMID: 37742846 DOI: 10.1016/j.jconrel.2023.09.038] [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: 04/24/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
Organelles not only constitute the basic structure of the cell but also are important in maintaining the normal physiological activities of the cell. With the development of biomimetic nanoscience, researchers have developed technologies to use organelles as drug carriers for disease treatment. Compared with traditional drug carriers, organelle drug carriers have the advantages of good biocompatibility, high drug loading efficiency, and modifiability, and the surface biomarkers of organelles can also participate in intracellular signal transduction to enhance intracellular and intercellular communication, and assist in enhancing the therapeutic effect of drugs. Among different types of organelles, extracellular vesicles, lipid droplets, lysosomes, and mitochondria have been used as drug carriers. This review briefly reviews the biogenesis, isolation methods, and drug-loading methods of four types of organelles, and systematically summarizes the research progress in using organelles as drug-delivery systems for disease treatment. Finally, the challenges faced by organelle-based drug delivery systems are discussed. Although the organelle-based drug delivery systems still face challenges before they can achieve clinical translation, they offer a new direction and vision for the development of next-generation drug carriers.
Collapse
Affiliation(s)
- Jiaxin Hu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Yimin Du
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Xingxing Peng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China; Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan Province, PR China.
| |
Collapse
|
5
|
Choi HJ, Kim JH, Le VQA, Kim BN, Cho BK, Kim YH, Min J. Yeast vacuolar enzymes as novel hatching inhibitors for aquatic organisms, Daphnia magna and Danio rerio eggs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115446. [PMID: 37688866 DOI: 10.1016/j.ecoenv.2023.115446] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/11/2023]
Abstract
Concerns over the spread of non-native species in aquatic environments have led to the need for effective methods to prevent and control their spread while protecting native species. This study investigated the potential of yeast vacuolar enzymes as a natural hatching inhibitor for controlling aquatic organisms. Hatching experiments with Daphnia magna eggs demonstrated that exposure to yeast vacuole enzymes inhibited hatching in a concentration-dependent manner, suggesting their potential as an effective inhibitor of egg hatching in aquatic organisms. Interestingly, the protease used for comparative purposes did not inhibit hatching, but instead increased the mortality of hatched D. magna. Additionally, chorionic changes were observed in non-hatched D. magna eggs and zebrafish eggs exposed to yeast vacuole enzymes, suggesting that the enzyme can alter the chorion and interfere with hatching. These findings suggest that yeast vacuolar enzymes may be a promising and natural management tool for controlling the spread of harmful aquatic organisms, and further research is warranted to explore their potential for species-specific control.
Collapse
Affiliation(s)
- Hyo Jin Choi
- School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, South Korea
| | - Ji Hun Kim
- Department of Biological Sciences, Korea Advanced institute of Science and Technology, 291 Daehak-ro, Yuseong-Gu, Daejeon 34141, South Korea
| | - Vu Quynh Anh Le
- School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, South Korea
| | - Bit-Na Kim
- School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, South Korea.
| | - Byung-Kwan Cho
- Department of Biological Sciences, Korea Advanced institute of Science and Technology, 291 Daehak-ro, Yuseong-Gu, Daejeon 34141, South Korea.
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju 28644, South Korea.
| | - Jiho Min
- School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, South Korea.
| |
Collapse
|
6
|
Choi W, Shin WR, Kim YH, Min J. Inducing a Proinflammatory Response with Bioengineered Yeast Vacuoles with TLR2-Binding Peptides (Vac T2BP) as a Drug Carrier for Daunorubicin Delivery. ACS APPLIED MATERIALS & INTERFACES 2023; 15:41258-41270. [PMID: 37615983 DOI: 10.1021/acsami.3c06669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Immune adjuvants have roles in immune activation for cancer therapy, and adjuvants derived from microbes have been applied. In this study, we propose the use of bioengineered vacuoles, derived from recombinant yeast with acute myeloid leukemia (AML) specificity and having a TLR-2-binding peptide (VacT2BP) on their surface, to induce a proinflammatory response as a dual-function nanomaterial for daunorubicin (DNR) delivery. Our results demonstrate that nanosized, isolated VacT2BP induced HL-60 cell-specific DNR delivery and apoptosis. Furthermore, we observed the selective release of high-mobility group box 1 from apoptotic HL-60 cells by DNR@VacT2BP. We concluded that DNR@VacT2BP exhibited target selectivity, and the indiscriminate occurrence of damage-associated molecular patterns (DAMPs) was inhibited by the VacT2BP carrier. The therapeutic efficacy of DNR@VacT2BP was confirmed in AML xenograft mice, with about 82% tumor growth inhibition. Following drug delivery, apoptotic cells and DAMPs with residual VacT2BP (apopDNR@VacT2BP) upregulated the proinflammatory immune response of macrophages. In addition, apopDNR@VacT2BP enhanced phagocytosis activity. Macrophages stimulated by apopDNR@VacT2BP suppressed cancer proliferation by about 40%. In summary, our results suggest that dual-functional vacuoles with a target-specific peptide can be a potential strategy for selective drug delivery and construction of an immune environment to fight cancer, thereby improving prognosis.
Collapse
Affiliation(s)
- Wooil Choi
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk 54896, South Korea
| | - Woo-Ri Shin
- School of Biological Sciences, Chungbuk National University, 1, Chungdae-Ro, Seowon-Gu, Cheongju 28644, South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, 1, Chungdae-Ro, Seowon-Gu, Cheongju 28644, South Korea
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk 54896, South Korea
| |
Collapse
|
7
|
Lee J, Nguyen NT, Tran LM, Kim YH, Min J. Targeted Killing of Staphylococcus aureus Using Specific Peptides Displayed on Yeast Vacuoles. Microbiol Spectr 2023; 11:e0092023. [PMID: 37098917 PMCID: PMC10269669 DOI: 10.1128/spectrum.00920-23] [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: 03/02/2023] [Accepted: 04/04/2023] [Indexed: 04/27/2023] Open
Abstract
Staphylococcus aureus is a common pathogen that causes health care-related and community-associated infections. In this study, we provide a novel system that can recognize and kill S. aureus bacteria. The system is specifically based on a combination of the phage display library technique and yeast vacuoles. A phage clone displaying a peptide capable of specific binding to a whole S. aureus cell was selected from a 12-mer phage peptide library. The peptide sequence was SVPLNSWSIFPR. The selected phage's ability to bind specifically with S. aureus was confirmed using an enzyme-linked immunosorbent assay, and the chosen peptide was then synthesized. The results showed that the synthesized peptides displayed high affinity with S. aureus but low binding ability with other strains, including Gram-negative and Gram-positive bacteria such as Salmonella sp., Shigella spp., Escherichia coli, and Corynebacterium glutamicum. In addition, yeast vacuoles were used as a drug carrier by encapsulating daptomycin, a lipopeptide antibiotic used to treat Gram-positive bacterial infections. The expression of specific peptides at the encapsulated vacuole membrane created an efficient system that can specifically recognize and kill S. aureus bacteria. IMPORTANCE The phage display method was used to select peptides with high affinity and specificity for S. aureus, and these peptides were then induced to be expressed on the surface of yeast vacuoles. These surface-modified vacuoles can act as drug carriers, with drugs such as the lipopeptide antibiotic daptomycin loaded inside. An advantage of using yeast vacuoles as a drug carrier is that they can be easily produced through yeast culture, making the approach cost-effective and suitable for large-scale production and potential implementation in clinical settings. This novel approach offers a promising way to specifically target and eliminate S. aureus that could ultimately lead to improved treatment of bacterial infections and reduced risk of antibiotic resistance.
Collapse
Affiliation(s)
- Jaewoong Lee
- School of Chemical Engineering, Jeonbuk National University, Deokjin-Gu Jeonju, Jeonbuk, South Korea
- Center for Ecology and Environmental Toxicology (CEET), Chungbuk National University, Seowon-Gu, Cheongju, South Korea
| | - Ngoc-Tu Nguyen
- School of Chemical Engineering, Jeonbuk National University, Deokjin-Gu Jeonju, Jeonbuk, South Korea
- Center for Ecology and Environmental Toxicology (CEET), Chungbuk National University, Seowon-Gu, Cheongju, South Korea
| | - Le-Minh Tran
- School of Chemical Engineering, Jeonbuk National University, Deokjin-Gu Jeonju, Jeonbuk, South Korea
| | - Yang-Hoon Kim
- Center for Ecology and Environmental Toxicology (CEET), Chungbuk National University, Seowon-Gu, Cheongju, South Korea
- School of Biological Sciences, Chungbuk National University, Seowon-Gu, Cheongju, South Korea
| | - Jiho Min
- School of Chemical Engineering, Jeonbuk National University, Deokjin-Gu Jeonju, Jeonbuk, South Korea
| |
Collapse
|
8
|
Raguraman R, Bhavsar D, Kim D, Ren X, Sikavitsas V, Munshi A, Ramesh R. Tumor-targeted exosomes for delivery of anticancer drugs. Cancer Lett 2023; 558:216093. [PMID: 36822543 PMCID: PMC10025995 DOI: 10.1016/j.canlet.2023.216093] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
Exosomes are small phospholipid bilayer vesicles that are naturally produced by all living cells, both prokaryotes and eukaryotes. The exosomes due to their unique size, reduced immunogenicity, and their ability to mimic synthetic liposomes in carrying various anticancer drugs have been tested as drug delivery vehicles for cancer treatment. An added advantage of developing exosomes as a drug carrier is the ease of manipulating their intraluminal content and their surface modification to achieve tumor-targeted drug delivery. In the past ten-years, there has been an exponential increase in the number of exosome-related studies in cancer. Preclinical studies demonstrate exosomes-mediated delivery of chemotherapeutics, biologicals and natural products produce potent anticancer activity both, in vitro and in vivo. In contrast, the number of exosome-based clinical trials are few due to challenges in the manufacturing and scalability related to large-scale production of exosomes and their storage and stability. Herein, we discuss recent advances in exosome-based drug delivery for cancer treatment in preclinical and clinical studies and conclude with challenges to be overcome for translating a larger number of exosome-based therapies into the clinic.
Collapse
Affiliation(s)
- Rajeswari Raguraman
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Dhaval Bhavsar
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Dongin Kim
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Xiaoyu Ren
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Vassilios Sikavitsas
- School of Chemical, Biological and Material Engineering, The University of Oklahoma, Norman, Oklahoma, 73019, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Anupama Munshi
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Rajagopal Ramesh
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Graduate Program in Biomedical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
9
|
Nguyen NHT, Nguyen NT, Kim YH, Min J. Yeast-derived vacuoles as a novel carrier with enhanced hCMEC/D3 cell monolayer penetration. Biotechnol J 2023; 18:e2200393. [PMID: 36321515 DOI: 10.1002/biot.202200393] [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: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 12/31/2022]
Abstract
The blood-brain barrier (BBB) is a brain protection structure that restricts drug delivery from the blood to the central nervous system. Thus, we developed a novel drug carrier using yeast vacuoles to overcome this problem. The purpose of this study was to assess the drug transportability of yeast vacuoles using a human cerebral microvascular endothelial cell line (hCMEC/D3) cell monolayer. Here, we used daunorubicin (DNR) as a microtubule-targeting agent with the ability to disaggregate pre-formed fibrils and prevent Tau fibrillization. An in vitro model was developed by culturing hCMEC/D3 cells on Transwell inserts in EBM-2 endothelial basal medium until the cells formed a monolayer. Next, nano-sized yeast vacuoles were loaded with DNR, and the signals inside and outside the hMEC/D3 cell monolayer were detected using the GloMax® Explorer fluorometer. DNR penetrated the cell monolayer and was regulated by endocytosis via receptor-mediated macropinocytosis on the surface of the cell. Confocal imaging showed a significant increase in intracellular DNR fluorescence when the cells were treated with the vacuole-encapsulated drug. These results indicate that the drug penetrated the hCMEC/D3 cell monolayer via encapsulation into the vacuoles. Overall, yeast-derived vacuoles are promising candidates as drug carriers to the brain.
Collapse
Affiliation(s)
- Ngoc-Han Thi Nguyen
- Department of Bioprocess Engineering, Jeonbuk National University, Jeonju, South Korea
| | - Ngoc-Tu Nguyen
- School of Chemical Engineering, Jeonbuk National University, Jeonju, South Korea
| | - Yang Hoon Kim
- School of Biological Sciences, Chungbuk National University, Cheongju, South Korea
| | - Jiho Min
- Department of Bioprocess Engineering, Jeonbuk National University, Jeonju, South Korea.,School of Chemical Engineering, Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
10
|
Lee SM, Kim BN, Kim YH, Min J. Identification of TLR2/4-mediated phagocytosis and immune response activation pathways by vacuoles isolated from Saccharomyces cerevisiae. J Cell Biochem 2023; 124:59-71. [PMID: 36302152 DOI: 10.1002/jcb.30342] [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: 07/01/2022] [Revised: 09/13/2022] [Accepted: 10/11/2022] [Indexed: 01/28/2023]
Abstract
The vacuoles of the yeast Saccharomyces cerevisiae are closely related to mammalian lysosomes and play a role in macromolecular degradation due to the hydrolytic enzymes present inside. The vacuoles also regulate osmotic pressure and control cellular homeostasis. In previous results, vacuoles were shown to activate the immune response of macrophages by promoting the production of immune-mediated transporters nitric oxide (NO), reactive oxygen species (ROS), and pro-inflammatory cytokines. In this study, the effects of vacuoles on the phagocytosis activity of RAW264.7 cells and their potential as immune enhancers were evaluated, and receptors capable of recognizing vacuoles were examined. An investigation using the phagocytes assay showed that phagocytosis activity increased by the vacuole. Besides, after treatment with TLR2/4 inhibitor, the expression of pro-inflammatory cytokines by vacuoles was significantly reduced and the inducible nitric oxide synthase (iNOS) protein was also significantly reduced. However, treatment with a TLR2 inhibitor did not reduce the production of interleukin-6 (IL)-6, a pro-inflammatory cytokine. As a result of confirming the activation of TLR2/4 using Western blot and immunofluorescence (IF), the TLR2/4 protein expression and fluorescence intensity increased depending on the concentration of vacuoles. Yeast vacuoles significantly upregulate protein expression of p-p65/p-p38 MAPKs. In summary, the vacuoles isolated from S. cerevisiae in macrophages have increased phagocytic ability at a concentration of 20 (µg/ml) and can function as immune-enhancing agent suggesting that TLR2/4 mediated the p38 MAPK/nuclear factor kappa B signaling pathway.
Collapse
Affiliation(s)
- Su-Min Lee
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, Deokjin-Gu, Jeonju, South Korea
| | - Bit-Na Kim
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, Deokjin-Gu, Jeonju, South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, Seowon-Gu, Cheongju, South Korea
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, Deokjin-Gu, Jeonju, South Korea
| |
Collapse
|
11
|
Choi W, Kim YH, Min J. Surface-modified vacuole-based daunorubicin delivery system for acute myeloid leukaemia (AML) and their selective therapeutics. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2022; 50:147-157. [PMID: 35635271 DOI: 10.1080/21691401.2022.2078339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
The vacuoles in Saccharomyces cerevisiae are the key players digesting the waste within the cell. This functional organelle corresponding to the lysosome of mammalians contains acidic hydrolases and specific membrane proteins. Vacuoles have more than 60 hydrolytic enzymes and can easily be modified by genetic engineering. In previous study, we optimised the encapsulation condition with appropriate time and concentration and confirmed the use of vacuole as drug delivery carrier for acute myeloid leukaemia treatment. In this study, recombinant vacuole that could target the acute myeloid leukaemia cell line was constructed. The vacuoles derived from genetic engineered yeast were decorated with targeting peptide that has specific affinity with TLR2 on AML cell membrane. The anti-cancer efficacy of AML targeting vacuoles carriers with encapsulated daunorubicin was shown to be higher than normal vacuole carriers and the crude daunorubicin. The results confirmed that target selective chemotherapy using the vacuole drug delivery system is effective and offers potential for cancer therapy.
Collapse
Affiliation(s)
- Wooil Choi
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, Jeonbuk, South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, Cheongju, South Korea
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, Jeonbuk, South Korea
| |
Collapse
|
12
|
Jeon G, Choi H, Park DJ, Nguyen NT, Kim YH, Min J. Melanin Treatment Effect of Vacuoles-Zinc Oxide Nanoparticles Combined with Ascorbic Acid. Mol Biotechnol 2022:10.1007/s12033-022-00608-8. [PMID: 36445610 PMCID: PMC9707414 DOI: 10.1007/s12033-022-00608-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/07/2022] [Indexed: 11/30/2022]
Abstract
Currently, ascorbic acid (AA) is widely used as a skin whitening material, but, AA, an unstable hydrophilic molecule, cannot penetrate the skin easily, due to the hydrophobic character of the stratum corneum. Therefore, we conjugated AA with hydrated zinc oxide-an inorganic matrix with positive surface charge, to improve the stability of AA. The metal-conjugated-ascorbic acid (ZnAA) was then combined with yeast vacuole through the vacuolar membrane proteins that relate to metal transportation to create an enhanced vacuole that contained ZnAA. The characteristics of vacuole with ZnAA (ZnAA_Vac) were next examined by various tests that included X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FT-IR), Field emission scanning electron microscopy (FE-SEM), and energy-dispersive X-ray (EDX) analysis. Furthermore, the ability of ZnAA_Vac to degrade melanin was confirmed in both melanoma cell line B16F10, and the artificial human skin MelanoDerm. The results showed that ZnAA_Vac possessed a higher depigmenting effect than the wild-type vacuole or ascorbic acid by reducing 75% of melanin color. Interestingly, ZnAA_Vac was found to be harmless, and did not cause any cytotoxicity to the cells. Overall, ZnAA_Vac is expected to provide a robust, harmless, and effective whitening agent for the skin.
Collapse
Affiliation(s)
- Gyeongchan Jeon
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-Daero, Deokjin-Gu, Jeonju-Si, Jeollabuk-do 54896 Republic of Korea
| | - Hyojin Choi
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-Daero, Deokjin-Gu, Jeonju-Si, Jeollabuk-do 54896 Republic of Korea
| | - Dong-Jun Park
- Department of Surgery, University of California, San Diego, USA
| | - Ngoc-Tu Nguyen
- Center for Ecology and Environmental Toxicology (CEET), Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, Chungbuk-Do 28644 South Korea
| | - Yang-Hoon Kim
- Center for Ecology and Environmental Toxicology (CEET), Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, Chungbuk-Do 28644 South Korea
- School of Biological Science, Chungbuk National University, Chungdae-Ro 1, Seowon-Gu, Cheongju, Chungbuk-do 28644 Republic of Korea
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-Daero, Deokjin-Gu, Jeonju-Si, Jeollabuk-do 54896 Republic of Korea
| |
Collapse
|
13
|
Liu N, Mishra K, Stiel AC, Gujrati V, Ntziachristos V. The sound of drug delivery: Optoacoustic imaging in pharmacology. Adv Drug Deliv Rev 2022; 189:114506. [PMID: 35998826 DOI: 10.1016/j.addr.2022.114506] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/14/2022] [Accepted: 08/17/2022] [Indexed: 01/24/2023]
Abstract
Optoacoustic (photoacoustic) imaging offers unique opportunities for visualizing biological function in vivo by achieving high-resolution images of optical contrast much deeper than any other optical technique. The method detects ultrasound waves that are generated inside tissue by thermo-elastic expansion, i.e., the conversion of light absorption by tissue structures to ultrasound when the tissue is illuminated by the light of varying intensity. Listening instead of looking to light offers the major advantage of image formation with a resolution that obeys ultrasonic diffraction and not photon diffusion laws. While the technique has been widely used to explore contrast from endogenous photo-absorbing molecules, such as hemoglobin or melanin, the use of exogenous agents can extend applications to a larger range of biological and possible clinical applications, such as image-guided surgery, disease monitoring, and the evaluation of drug delivery, biodistribution, and kinetics. This review summarizes recent developments in optoacoustic agents, and highlights new functions visualized and potent pharmacology applications enabled with the use of external contrast agents.
Collapse
Affiliation(s)
- Nian Liu
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany; Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany; PET Center, Department of Nuclear Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Kanuj Mishra
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Andre C Stiel
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Vipul Gujrati
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany; Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Vasilis Ntziachristos
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany; Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany; Munich Institute of Robotics and Machine Intelligence (MIRMI), Technical University of Munich, Munich 80992, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
14
|
Liu H, Zhang H, Han Y, Hu Y, Geng Z, Su J. Bacterial extracellular vesicles-based therapeutic strategies for bone and soft tissue tumors therapy. Theranostics 2022; 12:6576-6594. [PMID: 36185613 PMCID: PMC9516228 DOI: 10.7150/thno.78034] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/02/2022] [Indexed: 11/26/2022] Open
Abstract
Bone and soft tissue tumors are complex mesenchymal neoplasms that seriously endanger human health. Over the past decade, the relationship between microorganisms and human health and diseases is getting more attention. The extracellular vesicles derived from bacteria have been shown to regulate bacterial-host cell communication by transferring their contents, including nucleic acids, proteins, metabolites, lipopolysaccharides, and peptidoglycans. Bacteria extracellular vesicles (BEVs) are promising lipid-bilayer nanocarriers for the treatment of many diseases due to their low toxicity, drug loading capacity, ease of modification and industrialization. Specially, BEVs-based cancer therapy has attracted much attention because of their ability to effectively stimulate immune responses. In this review, we provide an overview of the biogenesis, composition, isolation, classification, and internalization of BEVs. We then comprehensively summarize the sources of BEVs in cancer therapy and the BEVs-related cancer treatment strategies. We further highlight the great potential of BEVs in bone and soft tissue tumors. Finally, we conclude the major advantages and challenges of BEVs-based cancer therapy. We believe that the comprehensive understanding of BEVs in the field of cancer therapy will generate innovative solutions to bone and soft tissue tumors and achieve clinical applications.
Collapse
Affiliation(s)
- Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Yafei Han
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Yan Hu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
15
|
Chen QW, Qiao JY, Liu XH, Zhang C, Zhang XZ. Customized materials-assisted microorganisms in tumor therapeutics. Chem Soc Rev 2021; 50:12576-12615. [PMID: 34605834 DOI: 10.1039/d0cs01571g] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microorganisms have been extensively applied as active biotherapeutic agents or drug delivery vehicles for antitumor treatment because of their unparalleled bio-functionalities. Taking advantage of the living attributes of microorganisms, a new avenue has been opened in anticancer research. The integration of customized functional materials with living microorganisms has demonstrated unprecedented potential in solving existing questions and even conferring microorganisms with updated antitumor abilities and has also provided an innovative train of thought for enhancing the efficacy of microorganism-based tumor therapy. In this review, we have summarized the emerging development of customized materials-assisted microorganisms (MAMO) (including bacteria, viruses, fungi, microalgae, as well as their components) in tumor therapeutics with an emphasis on the rational utilization of chosen microorganisms and tailored materials, the ingenious design of biohybrid systems, and the efficacious antitumor mechanisms. The future perspectives and challenges in this field are also discussed.
Collapse
Affiliation(s)
- Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Ji-Yan Qiao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Xin-Hua Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| |
Collapse
|
16
|
Enhanced immune response by vacuoles isolated from Saccharomyces cerevisiae in RAW 264.7 macrophages. Biosci Rep 2021; 41:229846. [PMID: 34558607 PMCID: PMC8482065 DOI: 10.1042/bsr20211158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 12/30/2022] Open
Abstract
Vacuoles are membrane vesicles in eukaryotic cells, the digestive system of cells that break down substances absorbed outside the cell and digest the useless components of the cell itself. Researches on anticancer and intractable diseases using vacuoles are being actively conducted. The practical application of the present study to animals requires the determination of the biocompatibility of vacuole. In the present study, we evaluated the effects of vacuoles isolated from Saccharomyces cerevisiae in RAW 264.7 cells. This showed a significant increase in the production of nitric oxide (NO) produced by macrophage activity. Using Reactive Oxygen Species (ROS) assay, we identified that ROS is increased in a manner dependent on vacuole concentration. Western blot analysis showed that vacuole concentration-dependently increased protein levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2). Therefore, iNOS expression was stimulated to induce NO production. In addition, pro-inflammatory cytokines levels promoted, such as interleukin (IL) 6 (IL-6) and tumor necrosis factor (TNF) α (TNF-α). In summary, vacuoles activate the immune response of macrophages by promoting the production of immune-mediated transporters NO, ROS, and pro-inflammatory cytokines.
Collapse
|
17
|
Rawal S, Patel M. Bio-Nanocarriers for Lung Cancer Management: Befriending the Barriers. NANO-MICRO LETTERS 2021; 13:142. [PMID: 34138386 PMCID: PMC8196938 DOI: 10.1007/s40820-021-00630-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/23/2021] [Indexed: 05/03/2023]
Abstract
Lung cancer is a complex thoracic malignancy developing consequential to aberrations in a myriad of molecular and biomolecular signaling pathways. It is one of the most lethal forms of cancers accounting to almost 1.8 million new annual incidences, bearing overall mortality to incidence ratio of 0.87. The dismal prognostic scenario at advanced stages of the disease and metastatic/resistant tumor cell populations stresses the requisite of advanced translational interdisciplinary interventions such as bionanotechnology. This review article deliberates insights and apprehensions on the recent prologue of nanobioengineering and bionanotechnology as an approach for the clinical management of lung cancer. The role of nanobioengineered (bio-nano) tools like bio-nanocarriers and nanobiodevices in secondary prophylaxis, diagnosis, therapeutics, and theranostics for lung cancer management has been discussed. Bioengineered, bioinspired, and biomimetic bio-nanotools of considerate translational value have been reviewed. Perspectives on existent oncostrategies, their critical comparison with bio-nanocarriers, and issues hampering their clinical bench side to bed transformation have also been summarized.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India
| | - Mayur Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India.
| |
Collapse
|
18
|
Liu N, Gujrati V, Malekzadeh-Najafabadi J, Werner JPF, Klemm U, Tang L, Chen Z, Prakash J, Huang Y, Stiel A, Mettenleiter G, Aichler M, Blutke A, Walch A, Kleigrewe K, Razansky D, Sattler M, Ntziachristos V. Croconaine-based nanoparticles enable efficient optoacoustic imaging of murine brain tumors. PHOTOACOUSTICS 2021; 22:100263. [PMID: 33948433 PMCID: PMC8080078 DOI: 10.1016/j.pacs.2021.100263] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/24/2021] [Accepted: 03/11/2021] [Indexed: 05/17/2023]
Abstract
Contrast enhancement in optoacoustic (photoacoustic) imaging can be achieved with agents that exhibit high absorption cross-sections, high photostability, low quantum yield, low toxicity, and preferential bio-distribution and clearance profiles. Based on advantageous photophysical properties of croconaine dyes, we explored croconaine-based nanoparticles (CR780RGD-NPs) as highly efficient contrast agents for targeted optoacoustic imaging of challenging preclinical tumor targets. Initial characterization of the CR780 dye was followed by modifications using polyethylene glycol and the cancer-targeting c(RGDyC) peptide, resulting in self-assembled ultrasmall particles with long circulation time and active tumor targeting. Preferential bio-distribution was demonstrated in orthotopic mouse brain tumor models by multispectral optoacoustic tomography (MSOT) imaging and histological analysis. Our findings showcase particle accumulation in brain tumors with sustainable strong optoacoustic signals and minimal toxic side effects. This work points to CR780RGD-NPs as a promising optoacoustic contrast agent for potential use in the diagnosis and image-guided resection of brain tumors.
Collapse
Affiliation(s)
- Nian Liu
- Chair of Biological Imaging, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Vipul Gujrati
- Chair of Biological Imaging, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
- Corresponding authors at: Chair of Biological Imaging, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany.
| | - Jaber Malekzadeh-Najafabadi
- Chair of Biological Imaging, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany
| | | | - Uwe Klemm
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Longguang Tang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361005, China
| | - Zhenyue Chen
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, University of Zurich and ETH Zurich, Zurich 8093, Switzerland
| | - Jaya Prakash
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
- Department of Instrumentation and Applied Physics, Indian Institute of Science, C. V. Raman Road, Bengaluru 560012, India
| | - Yuanhui Huang
- Chair of Biological Imaging, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Andre Stiel
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Gabriele Mettenleiter
- Research Unit Analytical Pathology, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Michaela Aichler
- Research Unit Analytical Pathology, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Andreas Blutke
- Research Unit Analytical Pathology, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Karin Kleigrewe
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich, Freising 85354, Germany
| | - Daniel Razansky
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, University of Zurich and ETH Zurich, Zurich 8093, Switzerland
| | - Michael Sattler
- Bavarian NMR Center and Center for Integrated Protein Science Munich, Department of Chemistry, Technical University of Munich, Garching 85747, Germany
- Institute of Structural Biology, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
| | - Vasilis Ntziachristos
- Chair of Biological Imaging, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg 85764, Germany
- Corresponding authors at: Chair of Biological Imaging, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany.
| |
Collapse
|
19
|
Choi W, Heo MY, Kim SY, Wee JH, Kim YH, Min J. Encapsulation of daunorubicin into Saccharomyces cerevisiae-derived lysosome as drug delivery vehicles for acute myeloid leukemia (AML) treatment. J Biotechnol 2020; 308:118-123. [PMID: 31846628 DOI: 10.1016/j.jbiotec.2019.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/02/2019] [Accepted: 12/13/2019] [Indexed: 01/05/2023]
Abstract
Lysosome, an intracellular organelle with an acid interior, contains acidic hydrolases and specific membrane proteins. Saccharomyces cerevisiae contains vacuoles (corresponding to lysosomes) that have similar lipid composition membrane to mammalian cell membrane. However, yeast vacuoles do not cause significant immune stimulation in vivo. Taking advantage of these structural similarities and bio-derived strengths, the present study describes encapsulation of daunorubicin into lysosome derived from S. cerevisiae as drug delivery vehicles for acute myeloid leukemia (AML) treatment. Daunorubicin is a chemotherapy medication used to treat cancer, specifically for AML. In this study, recombinant S. cerevisiae that could keep the small size of lysosomal vacuoles was constructed. Appropriate time and concentration to encapsulate the drug were then identified. In addition, release profile and anticancer effect of the drug in lysosome carriers were confirmed. According to this study, a more accurate encapsulation condition into lysosome can be optimized and potential application of S. cerevisiae derived lysosomes as drug carriers is confirmed.
Collapse
Affiliation(s)
- Wooil Choi
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk, 54896, South Korea
| | - Mi Young Heo
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk, 54896, South Korea
| | - Sang Yong Kim
- Department of Food Science and Biotechnology, Shin Ansan University, 135, Sinansandaehak-Ro, Danwon-Gu, Ansan, 15435, South Korea
| | - Ji-Hyang Wee
- Department of Food Science and Biotechnology, Shin Ansan University, 135, Sinansandaehak-Ro, Danwon-Gu, Ansan, 15435, South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, South Korea.
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk, 54896, South Korea.
| |
Collapse
|
20
|
Gujrati V, Prakash J, Malekzadeh-Najafabadi J, Stiel A, Klemm U, Mettenleiter G, Aichler M, Walch A, Ntziachristos V. Bioengineered bacterial vesicles as biological nano-heaters for optoacoustic imaging. Nat Commun 2019; 10:1114. [PMID: 30846699 PMCID: PMC6405847 DOI: 10.1038/s41467-019-09034-y] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 02/07/2019] [Indexed: 11/08/2022] Open
Abstract
Advances in genetic engineering have enabled the use of bacterial outer membrane vesicles (OMVs) to deliver vaccines, drugs and immunotherapy agents, as a strategy to circumvent biocompatibility and large-scale production issues associated with synthetic nanomaterials. We investigate bioengineered OMVs for contrast enhancement in optoacoustic (photoacoustic) imaging. We produce OMVs encapsulating biopolymer-melanin (OMVMel) using a bacterial strain expressing a tyrosinase transgene. Our results show that upon near-infrared light irradiation, OMVMel generates strong optoacoustic signals appropriate for imaging applications. In addition, we show that OMVMel builds up intense heat from the absorbed laser energy and mediates photothermal effects both in vitro and in vivo. Using multispectral optoacoustic tomography, we noninvasively monitor the spatio-temporal, tumour-associated OMVMel distribution in vivo. This work points to the use of bioengineered vesicles as potent alternatives to synthetic particles more commonly employed for optoacoustic imaging, with the potential to enable both image enhancement and photothermal applications.
Collapse
Affiliation(s)
- Vipul Gujrati
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Munich, 81675, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Jaya Prakash
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Jaber Malekzadeh-Najafabadi
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Munich, 81675, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Andre Stiel
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Uwe Klemm
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Gabriele Mettenleiter
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Michaela Aichler
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Vasilis Ntziachristos
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Munich, 81675, Germany.
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, 85764, Germany.
| |
Collapse
|
21
|
Eleutherio E, Brasil ADA, França MB, de Almeida DSG, Rona GB, Magalhães RSS. Oxidative stress and aging: Learning from yeast lessons. Fungal Biol 2018; 122:514-525. [DOI: 10.1016/j.funbio.2017.12.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 02/06/2023]
|
22
|
Zhang Q, Hu S, Wang K, Cui M, Li X, Wang M, Hu X. Engineering a yeast double-molecule carrier for drug screening. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:386-396. [PMID: 29611428 DOI: 10.1080/21691401.2018.1457539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
With the advantages of unicellular eukaryotic structure and easy manipulation, yeast becomes a popular tool for biochemical, genetic and medicinal studies. In order to construct an efficient anti-inflammatory drug screening platform, we engineered yeast as a double-molecule carrier, of which an inserted domain (I domain) of lymphocyte function-associated antigen 1 was displayed on yeast surface and a green fluorescent protein (GFP) was expressed inside cytosol. The I domain specifically targeted a surface marker of mammalian cells, intercellular adhesion molecule 1, whose number is correlated with the level of cellular inflammation. Examination of GFP intensity enables swift quantification of the yeast-mammalian cell binding and thus it reflects inflammatory potency, herein the inflammatory index, of a chemical imposed to cells. The inflammatory potency of a total of 1340 chemicals was indexed. Among them, 1 inflammation-inducing and 1 inflammation-reducing compounds were verified both in vitro and in vivo. Our method demonstrated a swift, facile and high-throughput screening platform at the protein level for inflammation and related diseases drug discovery without using sophisticated instruments.
Collapse
Affiliation(s)
- Qiyun Zhang
- a Laboratory of Drug Discovery and Molecular Engineering, Department of Medicinal Plants, College of Plant Science and Technology , Huazhong Agricultural University (HZAU) , Wuhan , China.,b National & Local Joint Engineering Research Center (Hubei) for Medicinal Plant Breeding and Cultivation , Wuhan , China.,c Hubei Provincial Engineering Research Center for Medicinal Plants , Wuhan , China
| | - Sheng Hu
- d Hubei Cancer Hospital , Wuhan , China
| | - Ke Wang
- e State Key Laboratory of Agricultural Microbiology , HZAU , Wuhan , China.,f College of Veterinary Medicine , HZAU , Wuhan , China
| | - Min Cui
- e State Key Laboratory of Agricultural Microbiology , HZAU , Wuhan , China.,f College of Veterinary Medicine , HZAU , Wuhan , China
| | - Xiaohua Li
- a Laboratory of Drug Discovery and Molecular Engineering, Department of Medicinal Plants, College of Plant Science and Technology , Huazhong Agricultural University (HZAU) , Wuhan , China.,b National & Local Joint Engineering Research Center (Hubei) for Medicinal Plant Breeding and Cultivation , Wuhan , China.,c Hubei Provincial Engineering Research Center for Medicinal Plants , Wuhan , China
| | - Mo Wang
- a Laboratory of Drug Discovery and Molecular Engineering, Department of Medicinal Plants, College of Plant Science and Technology , Huazhong Agricultural University (HZAU) , Wuhan , China.,b National & Local Joint Engineering Research Center (Hubei) for Medicinal Plant Breeding and Cultivation , Wuhan , China.,c Hubei Provincial Engineering Research Center for Medicinal Plants , Wuhan , China
| | - Xuebo Hu
- a Laboratory of Drug Discovery and Molecular Engineering, Department of Medicinal Plants, College of Plant Science and Technology , Huazhong Agricultural University (HZAU) , Wuhan , China.,b National & Local Joint Engineering Research Center (Hubei) for Medicinal Plant Breeding and Cultivation , Wuhan , China.,c Hubei Provincial Engineering Research Center for Medicinal Plants , Wuhan , China
| |
Collapse
|
23
|
Ståhl S, Gräslund T, Eriksson Karlström A, Frejd FY, Nygren PÅ, Löfblom J. Affibody Molecules in Biotechnological and Medical Applications. Trends Biotechnol 2017; 35:691-712. [PMID: 28514998 DOI: 10.1016/j.tibtech.2017.04.007] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 01/08/2023]
Abstract
Affibody molecules are small (6.5-kDa) affinity proteins based on a three-helix bundle domain framework. Since their introduction 20 years ago as an alternative to antibodies for biotechnological applications, the first therapeutic affibody molecules have now entered clinical development and more than 400 studies have been published in which affibody molecules have been developed and used in a variety of contexts. In this review, we focus primarily on efforts over the past 5 years to explore the potential of affibody molecules for medical applications in oncology, neurodegenerative, and inflammation disorders, including molecular imaging, receptor signal blocking, and delivery of toxic payloads. In addition, we describe recent examples of biotechnological applications, in which affibody molecules have been exploited as modular affinity fusion partners.
Collapse
Affiliation(s)
- Stefan Ståhl
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden.
| | - Torbjörn Gräslund
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | | | - Fredrik Y Frejd
- Unit of Biomedical Radiation Sciences, Uppsala University, SE-751 85 Uppsala, Sweden; Affibody AB, Gunnar Asplunds Allé 24, SE-171 69 Solna, Sweden
| | - Per-Åke Nygren
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - John Löfblom
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| |
Collapse
|
24
|
Kim H, Lee Y, Kang S, Choi M, Lee S, Kim S, Gujrati V, Kim J, Jon S. Self-assembled nanoparticles comprising aptide-SN38 conjugates for use in targeted cancer therapy. NANOTECHNOLOGY 2016; 27:48LT01. [PMID: 27804918 DOI: 10.1088/0957-4484/27/48/48lt01] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Self-assembled nanoparticles (NPs) have been intensively utilized as cancer drug delivery carriers because hydrophobic anticancer drugs may be efficiently loaded into the particle cores. In this study, we synthesized and evaluated the therapeutic index of self-assembled NPs chemically conjugated to a fibronectin extra domain B-specific peptide (APTEDB) and an anticancer agent SN38. The APTEDB-SN38 formed self-assembled structures with a diameter of 58 ± 3 nm in an aqueous solution and displayed excellent drug loading, solubility, and stability properties. A pharmacokinetic study revealed that the blood circulation half-life of SN38 following injection of the APTEDB-SN38 NPs was markedly higher than that of the small molecule CPT-11. The APTEDB-SN38 NPs delivered SN38 to tumor sites by both passive and active targeting. Finally, the APTEDB-SN38 NPs exhibited potent antitumor activities and low toxicities against EDB-expressing tumors (LLC, U87MG) in mice. This system merits further preclinical and clinical investigations for SN38 delivery.
Collapse
Affiliation(s)
- Hyungjun Kim
- KAIST Institute for the BioCentury, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Daejeon 305-701, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ding H, Gangalum PR, Galstyan A, Fox I, Patil R, Hubbard P, Murali R, Ljubimova JY, Holler E. HER2-positive breast cancer targeting and treatment by a peptide-conjugated mini nanodrug. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:631-639. [PMID: 27520726 DOI: 10.1016/j.nano.2016.07.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 11/17/2022]
Abstract
HER2+ breast cancer is one of the most aggressive forms of breast cancer. The new polymalic acid-based mini nanodrug copolymers are synthesized and specifically characterized to inhibit growth of HER2+ breast cancer. These mini nanodrugs are highly effective and in the clinic may substitute for trastuzumab (the marketed therapeutic antibody) and antibody-targeted nanobioconjugates. Novel mini nanodrugs are designed to have slender shape and small size. HER2+ cells were recognized by the polymer-attached trastuzumab-mimetic 12-mer peptide. Synthesis of the nascent cell-transmembrane HER2/neu receptors by HER2+ cells was inhibited by antisense oligonucleotides that prevented cancer cell proliferation and significantly reduced tumor size by more than 15 times vs. untreated control or PBS-treated group. We emphasize that the shape and size of mini nanodrugs can enhance penetration of multiple bio-barriers to facilitate highly effective treatment. Replacement of trastuzumab by the mimetic peptide favors reduced production costs and technical efforts, and a negligible immune response.
Collapse
Affiliation(s)
- Hui Ding
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Pallavi R Gangalum
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Anna Galstyan
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Irving Fox
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Rameshwar Patil
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Paul Hubbard
- Department of Biomedical Sciences, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ramachandran Murali
- Department of Biomedical Sciences, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Julia Y Ljubimova
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Eggehard Holler
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
26
|
Davis MI, Simeonov A, Auld D. Literature Search and Review. Assay Drug Dev Technol 2016. [DOI: 10.1089/adt.2016.29034.lit] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | | | - Doug Auld
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| |
Collapse
|