1
|
Guo Y, Sun Y, Li Z, Zuo C, Liu X, Chen Y, Xun Z, Liu J, Mei Y, Min JJ, Wen M, Zheng JH, Tan W. S100a8/a9 regulated by LPS/TLR4 axis plays an important role in Salmonella-based tumor therapy and host defense. Int J Cancer 2024; 155:2080-2093. [PMID: 39129048 DOI: 10.1002/ijc.35128] [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: 04/22/2024] [Revised: 06/02/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024]
Abstract
Bacteria are ideal anticancer agents and carriers due to their unique capabilities that are convenient in genetic manipulation, tumor-specific targeting, and deep-tissue penetration. However, the specific molecular mechanisms of bacteria-mediated cancer therapy (BMCT) have not been clarified. In this study, we found that TLR4 signaling pathway is critical for Salmonella-mediated tumor targeting, tumor suppression, and liver and spleen protection. TLR4 knockout in mice decreased the levels of cytokines and chemokines, such as S100a8, S100a9, TNF-α, and IL-1β, in tumor microenvironments (TMEs) after Salmonella treatment, which inhibited tumor cell death and nutrient release, led to reduced bacterial contents in tumors and attenuated antitumor efficacy in a negative feedback manner. Importantly, we found that S100a8 and S100a9 played a leading role in Salmonella-mediated cancer therapy (SMCT). The antitumor efficacy was abrogated and liver damage was prominent when blocked with a specific inhibitor. These findings elucidated the mechanism of Salmonella-mediated tumor targeting, suppression, and host antibacterial defense, providing insights into clinical cancer therapeutics.
Collapse
Affiliation(s)
- Yanxia Guo
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
| | - Yujie Sun
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
| | - Zhongying Li
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Xiaoqing Liu
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
| | - Yu Chen
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
| | - Zhen Xun
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Jinling Liu
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
- College of Biology, Hunan University, Changsha, China
| | - Yang Mei
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Min Wen
- Department of Neurosurgery, the second affiliated hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jin Hai Zheng
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
| | - Wenzhi Tan
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China
- School of Food Science and Bioengineering, Changsha University of Science & Technology, Changsha, Hunan, China
| |
Collapse
|
2
|
Wu Y, Qu H, Li X, Liu X, Wang L, Xia X, Wu X. Excessive autophagy-inducing and highly penetrable biomineralized bacteria for multimodal imaging-guided and mild hyperthermia-enhanced immunogenic cell death. J Colloid Interface Sci 2024; 679:181-196. [PMID: 39362143 DOI: 10.1016/j.jcis.2024.09.246] [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: 09/10/2024] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
The tumor microenvironment, characterized by hypoxia, supports the efficacy of anaerobic bacteria like attenuated S. typhimurium in cancer therapies. These bacteria target and penetrate deep tumor regions, significantly reducing tumor size but often lead to tumor regrowth due to limited long-term efficacy. To enhance the therapeutic impact, a novel biohybrid system, S@UIL, has been developed by coating S. typhimurium with a zirconium-based nanoscale metal-organic framework (UiO-66-NH2) loaded with indocyanine green (ICG) and luteolin (LUT). This system maintains the bacteria's tumor-targeting ability while increasing the penetration and therapeutic effectiveness through excessive autophagy and mild hyperthermia. In a subcutaneous colon cancer model, the integration of LUT and ICG promotes autophagic cell death and photothermal sensitization, leading to the release of damage-associated molecular patterns (DAMPs). These DAMPs activate immune responses through dendritic cells and T-cells, enhancing immunogenic cell death (ICD) and potentially reducing immune evasion by tumors. This single-administration approach also integrates multimodal imaging capabilities, providing a promising strategy for improved tumor ICD induction and cancer progression inhibition.
Collapse
Affiliation(s)
- Yundi Wu
- School of Biomedical Engineering, State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou 570228, China; School of Life and Health Sciences, Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou 571199, China
| | - Huanran Qu
- School of Biomedical Engineering, State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou 570228, China; School of Life and Health Sciences, Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Xiangying Li
- Department of Radiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou 570208, China
| | - Xiande Liu
- School of Life and Health Sciences, Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Xiaojing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Xilong Wu
- School of Biomedical Engineering, State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou 570228, China; School of Life and Health Sciences, Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China.
| |
Collapse
|
3
|
Huang Y, Piao L, Liu X. Enhancing Tumor-Specific immunity with SL dacA: A attenuated Salmonella-mediated c-di-AMP delivery system targeting the STING pathway. Int J Pharm 2024; 666:124759. [PMID: 39332458 DOI: 10.1016/j.ijpharm.2024.124759] [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: 05/31/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
The STING agonist stimulates an anti-tumor immune response by activating T cells, but its limited tumor-targeting specificity poses risks of cytokine storms or autoimmune reactions. Conversely, attenuated Salmonella typhimurium △ppGpp (S.t△ppGpp) exhibits superior tumor-targeting specificity and potent anti-tumor immunogenicity. However, the anti-tumor effects of Salmonella carrying STING agonists remain underexplored. In this study, we engineered a strain called SLdacA, utilizing S.t△ppGpp as a carrier, to produce c-di-AMP. This engineered strain effectively enhances dendritic cell maturation and M1-type macrophage polarization by inducing type I interferon production, thereby recruiting and activating effector T cells against tumor progression. This process is regulated by the STING/type I interferon pathway. Our findings indicate that utilizing S.t△ppGpp as a delivery vehicle for STING agonists holds promise as a strategy for synergistic bacterial-mediated immunotherapy.
Collapse
Affiliation(s)
- Yuanjia Huang
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation center of One Health, Hainan University, No. 58 Renmin Avenue, Haikou 570228, China
| | - Linghua Piao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Science, Hainan Medical University, No. 3 Xueyuan Avenue, Haikou 57119, China.
| | - Xiande Liu
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation center of One Health, Hainan University, No. 58 Renmin Avenue, Haikou 570228, China.
| |
Collapse
|
4
|
Staedtke V, Sun N, Bai R. Hypoxia-targeting bacteria in cancer therapy. Semin Cancer Biol 2024; 100:39-48. [PMID: 38554791 PMCID: PMC11344594 DOI: 10.1016/j.semcancer.2024.03.003] [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: 11/14/2023] [Revised: 03/10/2024] [Accepted: 03/20/2024] [Indexed: 04/02/2024]
Abstract
Tumor hypoxia plays a crucial role in driving cancer progression and fostering resistance to therapies by contributing significantly to chemoresistance, radioresistance, angiogenesis, invasiveness, metastasis, altered cell metabolism, and genomic instability. Despite the challenges encountered in therapeutically addressing tumor hypoxia with conventional drugs, a noteworthy alternative has emerged through the utilization of anaerobic oncolytic bacteria. These bacteria exhibit a preference for accumulating and proliferating within the hypoxic regions of tumors, where they can initiate robust antitumor effects and immune responses. Through simple genetic manipulation or sophisticated synthetic bioengineering, these bacteria can be further optimized to improve safety and antitumor activities, or they can be combined synergistically with chemotherapies, radiation, or other immunotherapies. In this review, we explore the potential benefits and challenges associated with this innovative anticancer approach, addressing issues related to clinical translation, particularly as several strains have progressed to clinical evaluation.
Collapse
Affiliation(s)
- Verena Staedtke
- Department of Neurology, Johns Hopkins University, 600 North Wolfe Street, Meyer 8-149 J, Baltimore, MD 21287, USA.
| | - Nihao Sun
- Kennedy Krieger Institute, Lab 520, 707 N Broadway, Baltimore, MD 21205, USA
| | - Renyuan Bai
- Kennedy Krieger Institute, Lab 520, 707 N Broadway, Baltimore, MD 21205, USA; Department of Neurosurgery, Johns Hopkins University, Lab 520, 707 N Broadway, Baltimore, MD 21205, USA
| |
Collapse
|
5
|
Wang C, Zhong L, Xu J, Zhuang Q, Gong F, Chen X, Tao H, Hu C, Huang F, Yang N, Li J, Zhao Q, Sun X, Huo Y, Chen Q, Zhao Y, Peng R, Liu Z. Oncolytic mineralized bacteria as potent locally administered immunotherapeutics. Nat Biomed Eng 2024; 8:561-578. [PMID: 38514774 DOI: 10.1038/s41551-024-01191-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 02/17/2024] [Indexed: 03/23/2024]
Abstract
Oncolytic bacteria can trigger innate immune activity. However, the antitumour efficacy of inactivated bacteria is poor, and attenuated live bacteria pose substantial safety risks. Here we show that intratumourally injected paraformaldehyde-fixed bacteria coated with manganese dioxide potently activate innate immune activity, modulate the immunosuppressive tumour microenvironment and trigger tumour-specific immune responses and abscopal antitumour responses. A single intratumoural administration of mineralized Salmonella typhimurium suppressed the growth of multiple types of subcutaneous and orthotopic tumours in mice, rabbits and tree shrews and protected the cured animals against tumour rechallenge. We also show that mineralized bacteria can be administered via arterial embolization to treat orthotopic liver cancer in rabbits. Our findings support the further translational testing of oncolytic mineralized bacteria as potent and safe antitumour immunotherapeutics.
Collapse
Affiliation(s)
- Chenya Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Liping Zhong
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Jiachen Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Zhuang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Fei Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Xiaojing Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Huiquan Tao
- InnoBM Pharmaceuticals Co. Ltd., Suzhou, China
| | - Cong Hu
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Fuquan Huang
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Junyan Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Qi Zhao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
- InnoBM Pharmaceuticals Co. Ltd., Suzhou, China
| | - Xinjun Sun
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Yu Huo
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Yongxiang Zhao
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China.
| | - Rui Peng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China.
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China.
- InnoBM Pharmaceuticals Co. Ltd., Suzhou, China.
| |
Collapse
|
6
|
Inpanathan S, Ospina-Escobar E, Li VC, Adamji Z, Lackraj T, Cho YH, Porco N, Choy CH, McPhee JB, Botelho RJ. Salmonella actively modulates TFEB in murine macrophages in a growth-phase and time-dependent manner. Microbiol Spectr 2024; 12:e0498122. [PMID: 38051049 PMCID: PMC10783059 DOI: 10.1128/spectrum.04981-22] [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: 12/04/2022] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
IMPORTANCE Activation of the host transcription factor TFEB helps mammalian cells adapt to stresses such as starvation and infection by upregulating lysosome, autophagy, and immuno-protective gene expression. Thus, TFEB is generally thought to protect host cells. However, it may also be that pathogenic bacteria like Salmonella orchestrate TFEB in a spatio-temporal manner to harness its functions to grow intracellularly. Indeed, the relationship between Salmonella and TFEB is controversial since some studies showed that Salmonella actively promotes TFEB, while others have observed that Salmonella degrades TFEB and that compounds that promote TFEB restrict bacterial growth. Our work provides a path to resolve these apparent discordant observations since we showed that stationary-grown Salmonella actively delays TFEB after infection, while late-log Salmonella is permissive of TFEB activation. Nevertheless, the exact function of this manipulation remains unclear, but conditions that erase the conditional control of TFEB by Salmonella may be detrimental to the microbe.
Collapse
Affiliation(s)
- Subothan Inpanathan
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Erika Ospina-Escobar
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Vanessa Cruz Li
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Zainab Adamji
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Tracy Lackraj
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Youn Hee Cho
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Natasha Porco
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Christopher H. Choy
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Joseph B. McPhee
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Roberto J. Botelho
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Abstract
This review is focused on describing and analyzing means by which Salmonella enterica serotype strains have been genetically modified with the purpose of developing safe, efficacious vaccines to present Salmonella-induced disease in poultry and to prevent Salmonella colonization of poultry to reduce transmission through the food chain in and on eggs and poultry meat. Emphasis is on use of recently developed means to generate defined deletion mutations to eliminate genetic sequences conferring antimicrobial resistance or residual elements that might lead to genetic instability. Problems associated with prior means to develop vaccines are discussed with presentation of various means by which these problems have been lessened, if not eliminated. Practical considerations are also discussed in hope of facilitating means to move lab-proven successful vaccination procedures and vaccine candidates to the marketplace to benefit the poultry industry.
Collapse
Affiliation(s)
- Roy Curtiss
- College of Veterinary Medicine, University of Florida, Gainesville, Florida,
| |
Collapse
|
8
|
Lim D, Kim K, Duysak T, So E, Jeong JH, Choy HE. Bacterial cancer therapy using the attenuated fowl-adapted Salmonella enterica serovar Gallinarum. Mol Ther Oncolytics 2023; 31:100745. [PMID: 38053546 PMCID: PMC10694566 DOI: 10.1016/j.omto.2023.100745] [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] [Indexed: 12/07/2023] Open
Abstract
We report here a novel anti-cancer therapy based on an avian-host-specific serotype Salmonella enterica serovar Gallinarum (S. Gallinarum) deficient in ppGpp synthesis. To monitor the tumor targeting, a bioluminescent ΔppGpp S. Gallinarum was constructed and injected intravenously into mice bearing syngeneic and human xenograft tumors. Strong bioluminescent signals were detected specifically in all grafted tumors at 2 days post-injection (dpi). The bacterial counts in normal and tumor tissue at 1 dpi revealed that ΔppGpp S. Gallinarum reached >108 CFU/g in tumor tissue and 106-107 CFU/g in endothelial organs; counts were much lower in other organs. At 16 dpi, ΔppGpp S. Gallinarum counts in tumor tissue decreased to ∼106 CFU/g, while those in the other organs became undetectable. A strong anti-cancer effect was observed after the injection of ΔppGpp S. Gallinarum into BALB/c mice grafted with CT26 colon cancer cells. This could be attributed to reduced virulence, which allowed the administration of at least a 10-fold greater dose (108 CFU) of ΔppGpp S. Gallinarum than other attenuated strains of S. enterica serovar Typhimurium (≤107 CFU). An advantage of the avian-specific S. Gallinarum as a cancer therapeutic should be a reduced capacity to cause infections or harm in humans.
Collapse
Affiliation(s)
- Daejin Lim
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Kwangsoo Kim
- Odysseus Bio, Basic Medical Research Building, Chonnam National University Medical College, 322 Seoyangro, Hwasun, Jeonnam 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Republic of Korea
| | - Taner Duysak
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Republic of Korea
| | - EunA. So
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Republic of Korea
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Republic of Korea
| | - Hyon E. Choy
- Odysseus Bio, Basic Medical Research Building, Chonnam National University Medical College, 322 Seoyangro, Hwasun, Jeonnam 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Republic of Korea
| |
Collapse
|
9
|
Tanniche I, Behkam B. Engineered live bacteria as disease detection and diagnosis tools. J Biol Eng 2023; 17:65. [PMID: 37875910 PMCID: PMC10598922 DOI: 10.1186/s13036-023-00379-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
Sensitive and minimally invasive medical diagnostics are essential to the early detection of diseases, monitoring their progression and response to treatment. Engineered bacteria as live sensors are being developed as a new class of biosensors for sensitive, robust, noninvasive, and in situ detection of disease onset at low cost. Akin to microrobotic systems, a combination of simple genetic rules, basic logic gates, and complex synthetic bioengineering principles are used to program bacterial vectors as living machines for detecting biomarkers of diseases, some of which cannot be detected with other sensing technologies. Bacterial whole-cell biosensors (BWCBs) can have wide-ranging functions from detection only, to detection and recording, to closed-loop detection-regulated treatment. In this review article, we first summarize the unique benefits of bacteria as living sensors. We then describe the different bacteria-based diagnosis approaches and provide examples of diagnosing various diseases and disorders. We also discuss the use of bacteria as imaging vectors for disease detection and image-guided surgery. We conclude by highlighting current challenges and opportunities for further exploration toward clinical translation of these bacteria-based systems.
Collapse
Affiliation(s)
- Imen Tanniche
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Bahareh Behkam
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, 24061, USA.
- School of Biomedical Engineered and Sciences, Virginia Tech, Blacksburg, VA, 24061, USA.
- Center for Engineered Health, Institute for Critical Technology and Applied Science, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
10
|
Zhang Y, Tan W, Sultonova RD, Nguyen DH, Zheng JH, You SH, Rhee JH, Kim SY, Khim K, Hong Y, Min JJ. Synergistic cancer immunotherapy utilizing programmed Salmonella typhimurium secreting heterologous flagellin B conjugated to interleukin-15 proteins. Biomaterials 2023; 298:122135. [PMID: 37148758 DOI: 10.1016/j.biomaterials.2023.122135] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 05/08/2023]
Abstract
The use of appropriately designed immunotherapeutic bacteria is an appealing approach to tumor therapy because the bacteria specifically target tumor tissue and deliver therapeutic payloads. The present study describes the engineering of an attenuated strain of Salmonella typhimurium deficient in ppGpp biosynthesis (SAM) that could secrete Vibrio vulnificus flagellin B (FlaB) conjugated to human (hIL15/FlaB) and mouse (mIL15/FlaB) interleukin-15 proteins in the presence of L-arabinose (L-ara). These strains, named SAMphIF and SAMpmIF, respectively, secreted fusion proteins that retained bioactivity of both FlaB and IL15. SAMphIF and SAMpmIF inhibited the growth of MC38 and CT26 subcutaneous (sc) tumors in mice and increased mouse survival rate more efficiently than SAM expressing FlaB alone (SAMpFlaB) or IL15 alone (SAMpmIL15 and SAMphIL15), although SAMpmIF had slightly greater antitumor activity than SAMphIF. The mice treated with these bacteria showed enhanced macrophage phenotype shift, from M2-like to M1-like, as well as greater proliferation and activation of CD4+ T, CD8+ T, NK, and NKT cells in tumor tissues. After tumor eradication by these bacteria, ≥50% of the mice show no evidence of tumor recurrence upon rechallenge with the same tumor cells, indicating that they had acquired long-term immune memory. Treatment of mice of 4T1 and B16F10 highly malignant sc tumors with a combination of these bacteria and an immune checkpoint inhibitor, anti-PD-L1 antibody, significantly suppressed tumor metastasis and increased mouse survival rate. Taken together, these findings suggest that SAM secreting IL15/FlaB is a novel therapeutic candidate for bacterial-mediated cancer immunotherapy and that its antitumor activity is enhanced by combination with anti-PD-L1 antibody.
Collapse
Affiliation(s)
- Ying Zhang
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Wenzhi Tan
- School of Food Science and Bioengineering, Changsha University of Science & Technology, Changsha, Hunan, 410114, China
| | - Rukhsora D Sultonova
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Dinh-Huy Nguyen
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Jin Hai Zheng
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | | | - Joon Haeng Rhee
- Department of Microbiology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - So-Young Kim
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Koemchhoy Khim
- Department of Microbiology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Yeongjin Hong
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea; Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, 58128, Republic of Korea.
| |
Collapse
|
11
|
ECM-targeting bacteria enhance chemotherapeutic drug efficacy by lowering IFP in tumor mouse models. J Control Release 2023; 355:199-210. [PMID: 36750146 DOI: 10.1016/j.jconrel.2023.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023]
Abstract
Bacterial cancer therapies aim to manipulate bacteria to effectively deploy therapeutic payloads to tumors. Attenuated bacteria alone often cannot eradicate solid tumors. Attenuated Salmonella can be engineered to deliver cytotoxic drugs to either trigger an immune response or increase antitumor efficacy when combined with chemotherapeutic drugs. However, the extracellular matrix (ECM) surrounding cancer cells forms a barrier that often limits the ability of chemotherapeutic and cytotoxic drugs to penetrate and eliminate tumors. To overcome this limitation, we developed a strategy to combine chemotherapy with an attenuated Salmonella typhimurium strain engineered to secrete HysA protein (from Staphylococcus aureus; Hyaluronidase, HAase) in tumors. The engineered Salmonella effectively degraded hyaluronan (HA), which is a major ECM constituent in tumors, and suppressed tumor growth in mouse models of pancreatic adenocarcinoma (ASPC-1) and breast cancer (4T1). Furthermore, it prolonged survival when combined with chemotherapeutic drugs (doxorubicin or gemcitabine). Upon bacterial colonization, the HAase-mediated ECM degradation decreased interstitial fluid pressure (IFP) in the tumor microenvironment. Additionally, HA degradation using HAase-expressing bacteria in vivo led to decreased binding to the receptor, CD44, expressed in tumors. This may modulate proliferation- and apoptosis-related signal pathways. Therefore, ECM-targeting bacteria can be used as a synergistic anticancer therapeutic agent to maximize chemotherapeutic drug delivery into highly invasive tumors.
Collapse
|
12
|
<italic>Salmonella typhimurium</italic> may support cancer treatment: a review. Acta Biochim Biophys Sin (Shanghai) 2023; 55:331-342. [PMID: 36786073 PMCID: PMC10160236 DOI: 10.3724/abbs.2023007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
<p indent="0mm">Antitumour treatments are evolving, including bacteria-mediated cancer therapy which is concurrently an ancient and cutting-edge approach. <italic>Salmonella typhimurium</italic> is a widely studied bacterial species that colonizes tumor tissues, showing oncolytic and immune system-regulating properties. It can be used as a delivery vector for genes and drugs, supporting conventional treatments that lack tumor-targeting abilities. This article summarizes recent evidence on the anticancer mechanisms of <italic>S</italic>. <italic>typhimurium</italic> alone and in combination with other anticancer treatments, suggesting that it may be a suitable approach to disease management. </p>.
Collapse
|
13
|
Qin Y, You SH, Zhang Y, Venu A, Hong Y, Min JJ. Genetic Programming by Nitric Oxide-Sensing Gene Switch System in Tumor-Targeting Bacteria. BIOSENSORS 2023; 13:266. [PMID: 36832032 PMCID: PMC9954711 DOI: 10.3390/bios13020266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/01/2023] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
Recent progress in synthetic biology has enabled bacteria to respond to specific disease signals to perform diagnostic and/or therapeutic tasks. Salmonella enterica subsp. enterica serovar Typhimurium (S. Typhimurium) colonization of tumors results in increases in nitric oxide (NO) levels, suggesting that NO may act as a candidate inducer of tumor-specific gene expression. The present study describes a NO-sensing gene switch system for triggering tumor-specific gene expression in an attenuated strain of S. Typhimurium. The genetic circuit was designed to sense NO via NorR, thus initiating the expression of FimE DNA recombinase. This was found to lead sequentially to the unidirectional inversion of a promoter region (fimS), which induced the expression of target genes. Target gene expression in bacteria transformed with the NO-sensing switch system was triggered in the presence of a chemical source of NO, diethylenetriamine/nitric oxide (DETA/NO) in vitro. In vivo results revealed that the gene expression is tumor-targeted, and specific to NO generated by inducible nitric oxide synthase (iNOS) after S. Typhimurium colonization. These results showed that NO was a promising inducer to finely tune the expression of target genes carried by tumor-targeting bacteria.
Collapse
Affiliation(s)
- Yeshan Qin
- Department of Molecular Medicine, Chonnam National University Graduate School, Gwangju 61469, Republic of Korea
- Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Gwangju 58128, Republic of Korea
| | - Sung-Hwan You
- Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Gwangju 58128, Republic of Korea
| | - Ying Zhang
- Department of Molecular Medicine, Chonnam National University Graduate School, Gwangju 61469, Republic of Korea
- Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Gwangju 58128, Republic of Korea
| | - Akhil Venu
- Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Gwangju 58128, Republic of Korea
| | - Yeongjin Hong
- Department of Microbiology, Chonnam National University Medical School, Gwangju 58128, Republic of Korea
| | - Jung-Joon Min
- Department of Molecular Medicine, Chonnam National University Graduate School, Gwangju 61469, Republic of Korea
- Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Gwangju 58128, Republic of Korea
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
14
|
Guo Y, Song M, Liu X, Chen Y, Xun Z, Sun Y, Tan W, He J, Zheng JH. Photodynamic therapy-improved oncolytic bacterial immunotherapy with FAP-encoding S. typhimurium. J Control Release 2022; 351:860-871. [PMID: 36181917 DOI: 10.1016/j.jconrel.2022.09.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 10/31/2022]
Abstract
Genetically engineered bacterial cancer therapy presents several advantages over conventional therapies. However, the anticancer effects of bacterium-based therapies remain insufficient, and serious side effects may be incurred with the increase in therapeutic dosages. Photodynamic therapy (PDT) suppresses tumor growth by producing reactive oxygen species (ROS) through specific laser-activated photosensitizers (PSs). Tumor-specific PS delivery and activatable ROS generation are two critical aspects for PDT advancement. Here, we propose PDT-enhanced oncolytic bacterial immunotherapy (OBI) by using genetically engineered avirulent Salmonella expressing a fluorogen-activating protein (FAP). Upon binding to a fluorogen, FAP could be activated and generate fluorescence and ROS. The instant activation of persistent fluorescence was detected in tumors through bacterium-based imaging. In a colon cancer model, PDT-OBI showed an enhanced tumor inhibition effect and prolonged animal survival. Mechanically, PDT generated ROS, resulting in the killing of cancer cells and over-accumulated bacteria. The pathogen-associated molecular patterns and damage-associated molecular patterns released from the destroyed bacteria and cancer cells recruited and activated immune cells (macrophages, neutrophils, and dendritic cells), which released additional proinflammatory cytokines (TNF-α and IL-1β); reduced anti-inflammatory cytokines (IL-10); and further enhanced immune cell infiltration in a positive-feedback manner, thus reducing bacterium-induced side effects and improving anticancer activities. This synergistic therapy has promising potential for cancer immunotherapy.
Collapse
Affiliation(s)
- Yanxia Guo
- School of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Mingxia Song
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Xiaoqing Liu
- School of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Yu Chen
- School of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Zhen Xun
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yujie Sun
- School of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Wenzhi Tan
- School of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Jianjun He
- School of Biomedical Sciences, Hunan University, Changsha 410082, China.
| | - Jin Hai Zheng
- School of Biomedical Sciences, Hunan University, Changsha 410082, China.
| |
Collapse
|
15
|
Becerra-Báez EI, Meza-Toledo SE, Muñoz-López P, Flores-Martínez LF, Fraga-Pérez K, Magaño-Bocanegra KJ, Juárez-Hernández U, Mateos-Chávez AA, Luria-Pérez R. Recombinant Attenuated Salmonella enterica as a Delivery System of Heterologous Molecules in Cancer Therapy. Cancers (Basel) 2022; 14:cancers14174224. [PMID: 36077761 PMCID: PMC9454573 DOI: 10.3390/cancers14174224] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Cancer is among the main causes of death of millions of individuals worldwide. Although survival has improved with conventional treatments, the appearance of resistant cancer cells leads to patient relapses. It is, therefore, necessary to find new antitumor therapies that can completely eradicate transformed cells. Bacteria-based tumor therapy represents a promising alternative treatment, particularly the use of live-attenuated Salmonella enterica, with its potential use as a delivery system of antitumor heterologous molecules such as tumor-associated antigens, cytotoxic molecules, immunomodulatory molecules, pro-apoptotic proteins, nucleic acids, and nanoparticles. In this review, we present the state of the art of current preclinical and clinical research on the use of Salmonella enterica as a potential therapeutic ally in the war against cancer. Abstract Over a century ago, bacterial extracts were found to be useful in cancer therapy, but this treatment modality was obviated for decades. Currently, in spite of the development and advances in chemotherapies and radiotherapy, failure of these conventional treatments still represents a major issue in the complete eradication of tumor cells and has led to renewed approaches with bacteria-based tumor therapy as an alternative treatment. In this context, live-attenuated bacteria, particularly Salmonella enterica, have demonstrated tumor selectivity, intrinsic oncolytic activity, and the ability to induce innate or specific antitumor immune responses. Moreover, Salmonella enterica also has strong potential as a delivery system of tumor-associated antigens, cytotoxic molecules, immunomodulatory molecules, pro-apoptotic proteins, and nucleic acids into eukaryotic cells, in a process known as bactofection and antitumor nanoparticles. In this review, we present the state of the art of current preclinical and clinical research on the use of Salmonella enterica as a potential therapeutic ally in the war against cancer.
Collapse
Affiliation(s)
- Elayne Irene Becerra-Báez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Sergio Enrique Meza-Toledo
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Paola Muñoz-López
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Luis Fernando Flores-Martínez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Karla Fraga-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| | - Kevin Jorge Magaño-Bocanegra
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico
| | - Uriel Juárez-Hernández
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico
| | - Armando Alfredo Mateos-Chávez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| | - Rosendo Luria-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Correspondence: ; Tel.: +52-55-52289917 (ext. 4401)
| |
Collapse
|
16
|
Elpers L, Deiwick J, Hensel M. Effect of Environmental Temperatures on Proteome Composition of Salmonella enterica Serovar Typhimurium. Mol Cell Proteomics 2022; 21:100265. [PMID: 35788066 PMCID: PMC9396072 DOI: 10.1016/j.mcpro.2022.100265] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 06/17/2022] [Accepted: 06/30/2022] [Indexed: 12/29/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (STM) is a major cause of gastroenteritis and transmitted by consumption of contaminated food. STM is associated to food originating from animals (pork, chicken, eggs) or plants (vegetables, fruits, nuts, and herbs). Infection of warm-blooded mammalian hosts by STM and the underlying complex regulatory network of virulence gene expression depend on various environmental conditions encountered in hosts. However, less is known about the proteome and possible regulatory networks for gene expression of STM outside the preferred host. Nutritional limitations and changes in temperature are the most obvious stresses outside the native host. Thus, we analyzed the proteome profile of STM grown in rich medium (LB medium) or minimal medium (PCN medium) at temperatures ranging from 8 °C to 37 °C. LB medium mimics the nutritional rich environment inside the host, whereas minimal PCN medium represents nutritional limitations outside the host, found during growth of fresh produce (field conditions). Further, the range of temperatures analyzed reflects conditions within natural hosts (37 °C), room temperature (20 °C), during growth under agricultural conditions (16 °C and 12 °C), and during food storage (8 °C). Implications of altered nutrient availability and growth temperature on STM proteomes were analyzed by HPLC/MS-MS and label-free quantification. Our study provides first insights into the complex adaptation of STM to various environmental temperatures, which allows STM not only to infect mammalian hosts but also to enter new infection routes that have been poorly studied so far. With the present dataset, global virulence factors, their impact on infection routes, and potential anti-infective strategies can now be investigated in detail. Especially, we were able to demonstrate functional flagella at 12 °C growth temperature for STM with an altered motility behavior.
Collapse
Affiliation(s)
- Laura Elpers
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
| | - Jörg Deiwick
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
| | - Michael Hensel
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany; CellNanOs - Center of Cellular Nanoanalytics Osnabrück, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany.
| |
Collapse
|
17
|
Pattern of F-18 FDG Uptake in Colon Cancer after Bacterial Cancer Therapy Using Engineered Salmonella Typhimurium: A Preliminary In Vivo Study. Mol Imaging 2022; 2022:9222331. [PMID: 35517712 PMCID: PMC9042370 DOI: 10.1155/2022/9222331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 02/16/2022] [Accepted: 03/17/2022] [Indexed: 12/21/2022] Open
Abstract
Purpose. Bacterial cancer therapy (BCT) research using engineered Salmonella typhimurium has increased in recent years. 2-Deoxy-2[18F] fluoro-D-glucose positron emission tomography (FDG PET) is widely used in cancer patients to detect cancer, monitor treatment responses, and predict prognoses. The aim of this pilot study was to investigate FDG uptake patterns in a mouse tumor model after BCT. Procedures. BCT was performed via the intravenous injection of attenuated S. typhimurium (SLΔppGpp/lux) into female mice bearing a tumor (derived from CT26 murine colon cancer cells) in the right thigh. 18F-FDG PET images acquired before BCT and at different time points after BCT. In vivo bioluminescence imaging confirmed bacterial presence in the tumor. The tumor volume, standardized uptake value (SUV) of FDG (SUVmax and SUVmean), early SUV reduction%, and normalized tumor volume change were analyzed. Results. Early after BCT (1 or 2 days post-injection (dpi)), FDG tumor uptake decreased in 10 out of 11 mice and then increased at later stages. FDG uptake before BCT was correlated with normalized tumor volume change after BCT. Early FDG reduction% after BCT was correlated with normalized volume change after BCT. Conclusions. Early after BCT, FDG tumor uptake decreased and then increased at later stages. The higher the FDG tumor uptake before BCT, the better the BCT response. FDG uptake patterns were related to tumor volume change after BCT. Therefore, FDG uptake was a good candidate for evaluating BCT.
Collapse
|
18
|
Ek V, Fattinger SA, Florbrant A, Hardt WD, Di Martino ML, Eriksson J, Sellin ME. A Motile Doublet Form of Salmonella Typhimurium Diversifies Target Search Behaviour at the Epithelial Surface. Mol Microbiol 2022; 117:1156-1172. [PMID: 35332598 PMCID: PMC9325389 DOI: 10.1111/mmi.14898] [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: 11/04/2021] [Revised: 02/24/2022] [Accepted: 03/21/2022] [Indexed: 12/01/2022]
Abstract
The behaviors of infectious bacteria are commonly studied in bulk. This is effective to define the general properties of a given isolate, but insufficient to resolve subpopulations and unique single‐microbe behaviors within the bacterial pool. We here employ microscopy to study single‐bacterium characteristics among Salmonella enterica serovar Typhimurium (S.Tm), as they prepare for and launch invasion of epithelial host cells. We find that during the bacterial growth cycle, S.Tm populations switch gradually from fast planktonic growth to a host cell‐invasive phenotype, characterized by flagellar motility and expression of the Type‐three‐secretion‐system‐1. The indistinct nature of this shift leads to the establishment of a transient subpopulation of S.Tm “doublets”—waist‐bearing bacteria anticipating cell division—which simultaneously express host cell invasion machinery. In epithelial cell culture infections, these S.Tm doublets outperform their “singlet” brethren and represent a hyperinvasive subpopulation. Atop both glass and enteroid‐derived monolayers, doublets swim along markedly straighter trajectories than singlets, thereby diversifying search patterns and improving the surface exploration capacity of the total bacterial population. The straighter swimming, combined with an enhanced cell‐adhesion propensity, suffices to account for the hyperinvasive doublet phenotype. This work highlights bacterial cell length heterogeneity as a key determinant of target search patterns atop epithelia.
Collapse
Affiliation(s)
- Viktor Ek
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| | - Stefan A Fattinger
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden.,Institute of Microbiology, Department of Biology, ETH, Zurich, Zurich, Switzerland
| | - Alexandra Florbrant
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH, Zurich, Zurich, Switzerland
| | - Maria Letizia Di Martino
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| | - Jens Eriksson
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| | - Mikael E Sellin
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| |
Collapse
|
19
|
Tan W, Duong MTQ, Zuo C, Qin Y, Zhang Y, Guo Y, Hong Y, Zheng JH, Min JJ. Targeting of pancreatic cancer cells and stromal cells using engineered oncolytic Salmonella typhimurium. Mol Ther 2022; 30:662-671. [PMID: 34400328 PMCID: PMC8821930 DOI: 10.1016/j.ymthe.2021.08.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/14/2021] [Accepted: 08/08/2021] [Indexed: 02/04/2023] Open
Abstract
Pancreatic cancer is resistant to conventional therapeutic interventions, mainly due to abundant cancer stromal cells and poor immune cell infiltration. Here, we used a targeted cancer therapy approach based on attenuated Salmonella typhimurium engineered to express cytolysin A (ClyA) to target cancer stromal cells and cancer cells and treat pancreatic cancer in mice. Nude mice bearing subcutaneous or orthotopic human pancreatic cancers were treated with engineered S. typhimurium expressing ClyA. The tumor microenvironment was monitored to analyze stromal cell numbers, stromal cell marker expression, and immune cell infiltration. The attenuated bacteria accumulated and proliferated specifically in tumor tissues after intravenous injection. The bacteria secreted ClyA into the tumor microenvironment. A single dose of ClyA-expressing Salmonella markedly inhibited growth of pancreatic cancer both in subcutaneous xenograft- and orthotopic tumor-bearing nude mice. Histological analysis revealed a marked decrease in expression of stromal cell markers and increased immune cell (neutrophils and macrophages) infiltration into tumors after colonization by ClyA-expressing bacteria. ClyA-expressing S. typhimurium destroyed cancer stromal cells and cancer cells in mouse models of human pancreatic cancer. This approach provides a novel strategy for combining anticancer and anti-stromal therapy to treat pancreatic cancer.
Collapse
Affiliation(s)
- Wenzhi Tan
- School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Mai Thi-Quynh Duong
- Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam 58128, Republic of Korea
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital, Changsha, Hunan 410013, China
| | - Yeshan Qin
- Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam 58128, Republic of Korea
| | - Ying Zhang
- Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam 58128, Republic of Korea
| | - Yanxia Guo
- School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Yeongjin Hong
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Jin Hai Zheng
- School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China,Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam 58128, Republic of Korea,Corresponding author: Jin Hai Zheng, College of Biology, Hunan University, Changsha, Hunan 410082, China.
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam 58128, Republic of Korea,Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Jeonnam 58128, Republic of Korea,Corresponding author: Jung-Joon Min, Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam 58128, Republic of Korea.
| |
Collapse
|
20
|
Nguyen DH, You SH, Vo ATN, Ngo HTT, Van Nguyen K, Duong MTQ, Choy HE, Song M, Hong Y, Min JJ. Optimized Doxycycline-Inducible Gene Expression System for Genetic Programming of Tumor-Targeting Bacteria. Mol Imaging Biol 2021; 24:82-92. [PMID: 34403085 PMCID: PMC8760206 DOI: 10.1007/s11307-021-01624-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/29/2021] [Accepted: 06/10/2021] [Indexed: 01/21/2023]
Abstract
PURPOSE In the programming of tumor-targeting bacteria, various therapeutic or reporter genes are expressed by different gene-triggering strategies. Previously, we engineered pJL87 plasmid with an inducible bacterial drug delivery system that simultaneously co-expressed two genes for therapy and imaging by a bidirectional tet promoter system only in response to the administration of exogenous doxycycline (Doxy). In this multi-cassette expression approach, tetA promoter (PtetA) was 100-fold higher in expression strength than tetR promoter (PtetR). In the present study, we developed pJH18 plasmid with novel Doxy-inducible gene expression system based on a tet promoter. PROCEDURES In this system, Tet repressor (TetR) expressed by a weak constitutive promoter binds to tetO operator, resulting in the tight repression of gene expressions by PtetA and PtetR, and Doxy releases TetR from tetO to de-repress PtetA and PtetR. RESULTS In Salmonella transformed with pJH18, the expression balance of bidirectional tet promoters in pJH18 was remarkably improved (PtetA:PtetR = 4~6:1) compared with that of pJL87 (PtetA:PtetR = 100:1) in the presence of Doxy. Also, the expression level by novel tet system was much higher in Salmonella transformed with pJH18 than in those with pJL87 (80-fold in rluc8 and 5-fold in clyA). Interestingly, pJH18 of the transformed Salmonella was much more stably maintained than pJL87 in antibiotic-free tumor-bearing mice (about 41-fold), because only pJH18 carries bom sequence with an essential role in preventing the plasmid-free population of programmed Salmonella from undergoing cell division. CONCLUSIONS Overall, doxycycline-induced co-expression of two proteins at similar expression levels, we exploited bioluminescence reporter proteins with preclinical but no clinical utility. Future validation with clinically compatible reporter systems, for example, suitable for radionuclide imaging, is necessary to develop this system further towards potential clinical application.
Collapse
Affiliation(s)
- Dinh-Huy Nguyen
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Sung-Hwan You
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - An-Trang Ngoc Vo
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Hien Thi-Thu Ngo
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Khuynh Van Nguyen
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Mai Thi-Quynh Duong
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Hyon E Choy
- Department of Microbiology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Miryoung Song
- Department of Bioscience and Biotechnology, Hankuk University of Foreign Studies, Yongin, South Korea
| | - Yeongjin Hong
- Department of Microbiology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.
- Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea.
- Department of Nuclear Medicine, Chonnam National University Medical School, Jeonnam, Gwangju, 61469, Republic of Korea.
| |
Collapse
|
21
|
Badie F, Ghandali M, Tabatabaei SA, Safari M, Khorshidi A, Shayestehpour M, Mahjoubin-Tehran M, Morshedi K, Jalili A, Tajiknia V, Hamblin MR, Mirzaei H. Use of Salmonella Bacteria in Cancer Therapy: Direct, Drug Delivery and Combination Approaches. Front Oncol 2021; 11:624759. [PMID: 33738260 PMCID: PMC7960920 DOI: 10.3389/fonc.2021.624759] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Over the years, conventional cancer treatments, such as chemotherapy with only a limited specificity for tumors, have undergone significant improvement. Moreover, newer therapies such as immunotherapy have undergone a revolution to stimulate the innate as well as adaptive immune responses against the tumor. However, it has been found that tumors can be selectively colonized by certain bacteria, where they can proliferate, and exert direct oncolytic effects as well as stimulating the immune system. Bacterial-mediated cancer therapy (BMCT) is now one example of a hot topic in the antitumor field. Salmonella typhimurium is a Gram-negative species that generally causes self-limiting gastroenteritis in humans. This species has been designed and engineered in order to be used in cancer-targeted therapeutics. S. typhimurium can be used in combination with other treatments such as chemotherapy or radiotherapy for synergistic modification of the tumor microenvironment. Considerable benefits have been shown by using engineered attenuated strains for the diagnosis and treatment of tumors. Some of these treatment approaches have received FDA approval for early-phase clinical trials. This review summarizes the use of Salmonella bacteria for cancer therapy, which could pave the way towards routine clinical application. The benefits of this therapy include an automatic self-targeting ability, and the possibility of genetic manipulation to produce newly engineered attenuated strains. Nevertheless, Salmonella-mediated anticancer therapy has not yet been clinically established, and requires more research before its use in cancer treatment.
Collapse
Affiliation(s)
- Fereshteh Badie
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Ghandali
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Alireza Tabatabaei
- Department of Internal Medicine, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mahmood Safari
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ahmad Khorshidi
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Shayestehpour
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Korosh Morshedi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Amin Jalili
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vida Tajiknia
- Department of Surgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
22
|
Kundra S, Colomer-Winter C, Lemos JA. Survival of the Fittest: The Relationship of (p)ppGpp With Bacterial Virulence. Front Microbiol 2020; 11:601417. [PMID: 33343543 PMCID: PMC7744563 DOI: 10.3389/fmicb.2020.601417] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
The signaling nucleotide (p)ppGpp has been the subject of intense research in the past two decades. Initially discovered as the effector molecule of the stringent response, a bacterial stress response that reprograms cell physiology during amino acid starvation, follow-up studies indicated that many effects of (p)ppGpp on cell physiology occur at levels that are lower than those needed to fully activate the stringent response, and that the repertoire of enzymes involved in (p)ppGpp metabolism is more diverse than initially thought. Of particular interest, (p)ppGpp regulation has been consistently linked to bacterial persistence and virulence, such that the scientific pursuit to discover molecules that interfere with (p)ppGpp signaling as a way to develop new antimicrobials has grown substantially in recent years. Here, we highlight contemporary studies that have further supported the intimate relationship of (p)ppGpp with bacterial virulence and studies that provided new insights into the different mechanisms by which (p)ppGpp modulates bacterial virulence.
Collapse
Affiliation(s)
- Shivani Kundra
- Department of Oral Biology, UF College of Dentistry, Gainesville, FL, United States
| | | | - José A Lemos
- Department of Oral Biology, UF College of Dentistry, Gainesville, FL, United States
| |
Collapse
|
23
|
Abstract
Antibiotics constitute one of the cornerstones of modern medicine. However, individuals may succumb to a bacterial infection if a pathogen survives exposure to antibiotics. The ability of bacteria to survive bactericidal antibiotics results from genetic changes in the preexisting bacterial genome, from the acquisition of genes from other organisms, and from nonheritable phenomena that give rise to antibiotic tolerance. Nonheritable antibiotic tolerance can be exhibited by a large fraction of the bacterial population or by a small subpopulation referred to as persisters. Nonheritable resistance to antibiotics has been ascribed to the activity of toxins that are part of toxin-antitoxin modules, to the universal energy currency ATP, and to the signaling molecule guanosine (penta) tetraphosphate. However, these molecules are dispensable for nonheritable resistance to antibiotics in many organisms. By contrast, nutrient limitation, treatment with bacteriostatic antibiotics, or expression of genes that slow bacterial growth invariably promote nonheritable resistance. We posit that antibiotic persistence results from conditions promoting feedback inhibition among core cellular processes, resulting phenotypically in a slowdown or halt in bacterial growth.
Collapse
|
24
|
Xiong S, Qi Z, Ni J, Zhong J, Cao L, Yang K. Attenuated Salmonella typhimurium-mediated tumour targeting imaging based on peptides. Biomater Sci 2020; 8:3712-3719. [PMID: 32495762 DOI: 10.1039/d0bm00013b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Attenuated bacteria-mediated tumor targeting diagnosis will provide a novel strategy for further cancer treatments owing to the intrinsic facultative anaerobic characteristic of bacteria and rapid proliferation in the tumor sites. In this work, we firstly investigate the in vivo behaviour of the attenuated Salmonella typhimurium (S. typhimuriumΔppGpp/lux) after intravenous injection. S. typhimurium exhibits rapid proliferation in tumor sites after three days of injection through bioluminescence imaging, the Luria-Bertani plate and the Gram-staining assay. In contrast, S. typhimurium does not proliferate in the normal tissues and could be excreted from the body of mice. Afterwards, a targeting peptide ubiquicidin (UBI) labeled with fluorescent dye Cy5.5 or radionuclide 125I was intravenously injected into the mice with or without S. typhimurium treatments for in vivo fluorescence imaging and single-photon emission computed tomography (SPECT/CT) imaging, respectively. The results show that the peptide UBI could specifically target the two independent bacteria-infected tumor models, the 4T1 murine breast cancer model and the CT26 mouse colon cancer model, realizing the sensitive multimodal imaging of tumors. Such a strategy (bacteria-mediated tumor targeting) may further improve the sensitivity to early diagnosis of tumors. We hope that our developed strategy could further be extended to cancer theranostics in the future, bringing good news for cancer patients.
Collapse
Affiliation(s)
- Saisai Xiong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China.
| | | | | | | | | | | |
Collapse
|
25
|
Development of Dual-Scale Fluorescence Endoscopy for In Vivo Bacteria Imaging in an Orthotopic Mouse Colon Tumor Model. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10030844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Colorectal cancer is a representative cancer where early diagnosis and proper treatment monitoring are important. Recently, cancer treatment using bacteria has actively progressed and has been successfully monitored using fluorescence imaging techniques. However, because subcutaneous tumor models are limited in reflecting the actual colorectal cancer situation, new imaging approaches are needed to observe cancers growing in the colon. The fluorescence endoscopic approach is an optimal monitoring modality to evaluate the therapeutic response of bacteria in orthotopic colon cancer. In this study, we developed dual-scaled fluorescence endoscopy (DSFE) by combining wide-field fluorescence endoscopy (WFE) and confocal fluorescence endomicroscopy (CFEM) and demonstrated its usefulness for evaluating bacterial therapy. Firstly, the endoscopic probe of DSFE was developed by integrating the CFEM probe into the guide sheath of WFE. Secondly, colorectal cancer tumor growth and tumors infiltrating the fluorescent bacteria were successfully monitored at the multi-scale using DSFE. Finally, the bacterial distribution of the tumor and organs were imaged and quantitatively analyzed using CFEM. DSFE successfully exhibited fluorescent bacterial signals in an orthotopic mouse colon tumor model. Thus, it can be concluded that the DSFE system is a promising modality to monitor bacterial therapy in vivo.
Collapse
|
26
|
Guo Y, Chen Y, Liu X, Min JJ, Tan W, Zheng JH. Targeted cancer immunotherapy with genetically engineered oncolytic Salmonella typhimurium. Cancer Lett 2020; 469:102-110. [DOI: 10.1016/j.canlet.2019.10.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/29/2019] [Accepted: 10/21/2019] [Indexed: 12/31/2022]
|
27
|
The Small RNA PinT Contributes to PhoP-Mediated Regulation of the Salmonella Pathogenicity Island 1 Type III Secretion System in Salmonella enterica Serovar Typhimurium. J Bacteriol 2019; 201:JB.00312-19. [PMID: 31262841 DOI: 10.1128/jb.00312-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/26/2019] [Indexed: 11/20/2022] Open
Abstract
Salmonella enterica serovar Typhimurium induces inflammatory diarrhea and bacterial uptake into intestinal epithelial cells using the Salmonella pathogenicity island 1 (SPI1) type III secretion system (T3SS). HilA activates transcription of the SPI1 structural components and effector proteins. Expression of hilA is activated by HilD, HilC, and RtsA, which act in a complex feed-forward regulatory loop. Many environmental signals and other regulators are integrated into this regulatory loop, primarily via HilD. After the invasion of Salmonella into host intestinal epithelial cells or during systemic replication in macrophages, the SPI T3SS is no longer required or expressed. We have shown that the two-component regulatory system PhoPQ, required for intracellular survival, represses the SPI1 T3SS mostly by controlling the transcription of hilA and hilD Here we show that PinT, one of the PhoPQ-regulated small RNAs (sRNAs), contributes to this regulation by repressing hilA and rtsA translation. PinT base pairs with both the hilA and rtsA mRNAs, resulting in translational inhibition of hilA, but also induces degradation of the rts transcript. PinT also indirectly represses expression of FliZ, a posttranslational regulator of HilD, and directly represses translation of ssrB, encoding the primary regulator of the SPI2 T3SS. Our in vivo mouse competition assays support the concept that PinT controls a series of virulence genes at the posttranscriptional level in order to adapt Salmonella from the invasion stage to intracellular survival.IMPORTANCE Salmonella is one of the most important food-borne pathogens, infecting over one million people in the United States every year. These bacteria use a needle-like device to interact with intestinal epithelial cells, leading to invasion of the cells and induction of inflammatory diarrhea. A complex regulatory network controls expression of the invasion system in response to numerous environmental signals. Here we explore the molecular mechanisms by which the small RNA PinT contributes to this regulation, facilitating inactivation of the system after invasion. PinT controls several important virulence systems in Salmonella, tuning the transition between different stages of infection.
Collapse
|
28
|
Dasgupta S, Das S, Biswas A, Bhadra RK, Das S. Small alarmones (p)ppGpp regulate virulence associated traits and pathogenesis of Salmonella enterica serovar Typhi. Cell Microbiol 2019; 21:e13034. [PMID: 31013389 DOI: 10.1111/cmi.13034] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/29/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022]
Abstract
How Salmonella enterica serovar Typhi (S. Typhi), an important human pathogen, survives the stressful microenvironments inside the gastrointestinal tract and within macrophages remains poorly understood. We report here that S. Typhi has a bonafide stringent response (SR) system, which is mediated by (p)ppGpp and regulates multiple virulence-associated traits and the pathogenicity of the S. Typhi Ty2 strain. In an iron overload mouse model of S. Typhi infection, the (p)ppGpp0 (Ty2ΔRelAΔSpoT) strain showed minimal systemic spread and no mortality, as opposed to 100% death of the mice challenged with the isogenic wild-type strain. Ty2ΔRelAΔSpoT had markedly elongated morphology with incomplete septa formation and demonstrated severely attenuated motility and chemotaxis due to the loss of flagella. Absence of the Vi-polysaccharide capsule rendered the mutant strain highly susceptible to complement-mediated lysis. The phenotypes of Ty2ΔRelAΔSpoT was contributed by transcriptional repression of several genes, including fliC, tviA, and ftsZ, as found by reverse transcriptase quantitative polymerase chain reaction and gene complementation studies. Finally, Ty2ΔRelAΔSpoT had markedly reduced invasion into intestinal epithelial cells and significantly attenuated survival within macrophages. To the best of our knowledge, this was the first study that addressed SR in S. Typhi and showed that (p)ppGpp was essential for optimal pathogenic fitness of the organism.
Collapse
Affiliation(s)
- Shreya Dasgupta
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Sayan Das
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Asim Biswas
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Rupak K Bhadra
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Santasabuj Das
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| |
Collapse
|
29
|
Kröger C, MacKenzie KD, Alshabib EY, Kirzinger MWB, Suchan DM, Chao TC, Akulova V, Miranda-CasoLuengo AA, Monzon VA, Conway T, Sivasankaran SK, Hinton JCD, Hokamp K, Cameron ADS. The primary transcriptome, small RNAs and regulation of antimicrobial resistance in Acinetobacter baumannii ATCC 17978. Nucleic Acids Res 2018; 46:9684-9698. [PMID: 29986115 PMCID: PMC6182133 DOI: 10.1093/nar/gky603] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 06/21/2018] [Accepted: 06/26/2018] [Indexed: 12/12/2022] Open
Abstract
We present the first high-resolution determination of transcriptome architecture in the priority pathogen Acinetobacter baumannii. Pooled RNA from 16 laboratory conditions was used for differential RNA-seq (dRNA-seq) to identify 3731 transcriptional start sites (TSS) and 110 small RNAs, including the first identification in A. baumannii of sRNAs encoded at the 3' end of coding genes. Most sRNAs were conserved among sequenced A. baumannii genomes, but were only weakly conserved or absent in other Acinetobacter species. Single nucleotide mapping of TSS enabled prediction of -10 and -35 RNA polymerase binding sites and revealed an unprecedented base preference at position +2 that hints at an unrecognized transcriptional regulatory mechanism. To apply functional genomics to the problem of antimicrobial resistance, we dissected the transcriptional regulation of the drug efflux pump responsible for chloramphenicol resistance, craA. The two craA promoters were both down-regulated >1000-fold when cells were shifted to nutrient limited medium. This conditional down-regulation of craA expression renders cells sensitive to chloramphenicol, a highly effective antibiotic for the treatment of multidrug resistant infections. An online interface that facilitates open data access and visualization is provided as 'AcinetoCom' (http://bioinf.gen.tcd.ie/acinetocom/).
Collapse
Affiliation(s)
- Carsten Kröger
- Department of Microbiology, School of Genetics & Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Keith D MacKenzie
- Institute for Microbial Systems and Society, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
- Department of Biology, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
| | - Ebtihal Y Alshabib
- Institute for Microbial Systems and Society, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
- Department of Biology, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
| | - Morgan W B Kirzinger
- Institute for Microbial Systems and Society, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
| | - Danae M Suchan
- Institute for Microbial Systems and Society, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
- Department of Biology, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
| | - Tzu-Chiao Chao
- Department of Biology, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
- Institute of Environmental Change and Society, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
| | - Valentyna Akulova
- Institute for Microbial Systems and Society, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
| | - Aleksandra A Miranda-CasoLuengo
- Department of Microbiology, School of Genetics & Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Vivian A Monzon
- Department of Microbiology, School of Genetics & Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Tyrrell Conway
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK 74078, USA
| | - Sathesh K Sivasankaran
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jay C D Hinton
- Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Karsten Hokamp
- Department of Genetics, School of Genetics & Microbiology, Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew D S Cameron
- Institute for Microbial Systems and Society, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
- Department of Biology, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
| |
Collapse
|
30
|
Amar A, Pezzoni M, Pizarro RA, Costa CS. New envelope stress factors involved in σ E activation and conditional lethality of rpoE mutations in Salmonella enterica. MICROBIOLOGY-SGM 2018; 164:1293-1307. [PMID: 30084765 DOI: 10.1099/mic.0.000701] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Salmonella enterica serovar Typhimurium (S. typhimurium) can cause food- and water-borne illness with diverse clinical manifestations. One key factor for S. typhimurium pathogenesis is the alternative sigma factor σE, which is encoded by the rpoE gene and controls the transcription of genes required for outer-membrane integrity in response to alterations in the bacterial envelope. The canonical pathway for σE activation involves proteolysis of the antisigma factor RseA, which is triggered by unfolded outer-membrane porins (OMPs) and lipopolysaccharides (LPS) that have accumulated in the periplasm. This study reports new stress factors that are able to activate σE expression. We demonstrate that UVA radiation induces σE activity in a pathway that is dependent on the stringent response regulator ppGpp. Survival assays revealed that rpoE has a role in the defence against lethal UVA doses that is mediated by functions that are dependent on and independent of the alternative sigma factor RpoS. We also report that the envelope stress generated by phage infection requires a functional rpoE gene for optimal bacterial tolerance and that it is able to induce σE activity in an RseA-dependent fashion. σE activity is also induced by hypo-osmotic shock in the absence of osmoregulated periplasmic glucans (OPGs). It is known that the rpoE gene is not essential in S. typhimurium. However, we report here two cases of the conditional lethality of rpoE mutations in this micro-organism. We demonstrate that rpoE mutations are not tolerated in the absence of OPGs (at low to moderate osmolarity) or LPS O-antigen. The latter case resembles that of the prototypic Escherichia coli strain K12, which neither synthesizes a complete LPS nor tolerates null rpoE mutations.
Collapse
Affiliation(s)
- Agustina Amar
- Dpto. de Radiobiología, Comisión Nacional de Energía Atómica, General San Martín, Argentina
| | - Magdalena Pezzoni
- Dpto. de Radiobiología, Comisión Nacional de Energía Atómica, General San Martín, Argentina
| | - Ramón A Pizarro
- Dpto. de Radiobiología, Comisión Nacional de Energía Atómica, General San Martín, Argentina
| | - Cristina S Costa
- Dpto. de Radiobiología, Comisión Nacional de Energía Atómica, General San Martín, Argentina
| |
Collapse
|
31
|
Hüttener M, Prieto A, Espelt J, Bernabeu M, Juárez A. Stringent Response and AggR-Dependent Virulence Regulation in the Enteroaggregative Escherichia coli Strain 042. Front Microbiol 2018; 9:717. [PMID: 29692772 PMCID: PMC5902536 DOI: 10.3389/fmicb.2018.00717] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/27/2018] [Indexed: 12/22/2022] Open
Abstract
Virulence expression in the enteroaggregative Escherichia coli strain 042 requires the transcriptional activator AggR. We show in this report that, as reported for other virulence factors, the nucleotide second messenger (p)ppGpp is needed for a high expression level of AggR. As expected from these findings, expression of AggR-activated genes such as the AafA pilin subunit is downregulated in the absence of (p)ppGpp. Considering the fact that biofilm formation in strain 042 requires the AafA protein, biofilm development in strain 042 is impaired in derivatives that lack either the AggR protein, the virulence plasmid that encodes AggR (pAA2) or the ability to synthesize (p)ppGpp. These results show a direct correlation between (p)ppGpp, expression of AggR and biofilm development in strain 042.
Collapse
Affiliation(s)
- Mário Hüttener
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Alejandro Prieto
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Joan Espelt
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Manuel Bernabeu
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Antonio Juárez
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain.,Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| |
Collapse
|
32
|
Kim K, Min SY, Lim HD, You SH, Lim D, Jeong JH, Kim HJ, Rhee JH, Park K, Shin M, Kim GJ, Min JJ, Choy HE. Cell mass-dependent expression of an anticancer protein drug by tumor-targeted Salmonella. Oncotarget 2018; 9:8548-8559. [PMID: 29492216 PMCID: PMC5823552 DOI: 10.18632/oncotarget.24013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/03/2017] [Indexed: 01/09/2023] Open
Abstract
Bacterial cancer therapy relies on the properties of certain bacterial species capable of targeting and proliferating within solid malignancies. If these bacteria could be loaded with antitumor proteins, the efficacy of this approach could be greatly increased. However, because most antitumor proteins are also toxic to normal tissue, they must be expressed by bacteria that specifically target and exclusively localize to tumor tissue. As a strategy for treating solid malignancies, we recently evaluated L-asparaginase (L-ASNase) delivered by tumor-targeted Salmonella. In this system, L-ASNase was expressed under the control of the araBAD promoter (PBAD) of the E. coli arabinose operon, which is induced by injection of L-arabinose. Here, we further improved the performance of recombinant Salmonella in cancer therapy by exploiting the quorum-sensing (QS) system, which uses cell mass-dependent auto-induction logic. This approach obviates the necessity of monitoring intratumoral bacterial status and inducing cargo protein expression by administration of an exogenous compound. Recombinant Salmonella in tumors expressed and secreted active L-ASNase in a cell mass-dependent manner, yielding significant anticancer effects. These results suggest that expression of a therapeutic protein under the control of the QS system represents a promising engineering platform for the production of recombinant proteins in vivo.
Collapse
Affiliation(s)
- Kwangsoo Kim
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Sa-Young Min
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Ho-Dong Lim
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Sung-Hwan You
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Daejin Lim
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Hyun-Ju Kim
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Joon Haeng Rhee
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Kyeongil Park
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Minsang Shin
- Department of Microbiology, Kyungpook National University Medical School, Daegu, Republic of Korea
| | - Geun-Joong Kim
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Jung-Joon Min
- Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea.,Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Jeonnam, Republic of Korea
| | - Hyon E Choy
- Department of Microbiology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.,Molecular Medicine, BK21 plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| |
Collapse
|
33
|
The stringent response promotes biofilm dispersal in Pseudomonas putida. Sci Rep 2017; 7:18055. [PMID: 29273811 PMCID: PMC5741744 DOI: 10.1038/s41598-017-18518-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 12/12/2017] [Indexed: 01/08/2023] Open
Abstract
Biofilm dispersal is a genetically programmed response enabling bacterial cells to exit the biofilm in response to particular physiological or environmental conditions. In Pseudomonas putida biofilms, nutrient starvation triggers c-di-GMP hydrolysis by phosphodiesterase BifA, releasing inhibition of protease LapG by the c-di-GMP effector protein LapD, and resulting in proteolysis of the adhesin LapA and the subsequent release of biofilm cells. Here we demonstrate that the stringent response, a ubiquitous bacterial stress response, is accountable for relaying the nutrient stress signal to the biofilm dispersal machinery. Mutants lacking elements of the stringent response - (p)ppGpp sythetases [RelA and SpoT] and/or DksA - were defective in biofilm dispersal. Ectopic (p)ppGpp synthesis restored biofilm dispersal in a ∆relA ∆spoT mutant. In vivo gene expression analysis showed that (p)ppGpp positively regulates transcription of bifA, and negatively regulates transcription of lapA and the lapBC, and lapE operons, encoding a LapA-specific secretion system. Further in vivo and in vitro characterization revealed that the PbifA promoter is dependent on the flagellar σ factor FliA, and positively regulated by ppGpp and DksA. Our results indicate that the stringent response stimulates biofilm dispersal under nutrient limitation by coordinately promoting LapA proteolysis and preventing de novo LapA synthesis and secretion.
Collapse
|
34
|
Zheng JH, Nguyen VH, Jiang SN, Park SH, Tan W, Hong SH, Shin MG, Chung IJ, Hong Y, Bom HS, Choy HE, Lee SE, Rhee JH, Min JJ. Two-step enhanced cancer immunotherapy with engineered Salmonella typhimurium secreting heterologous flagellin. Sci Transl Med 2017; 9:9/376/eaak9537. [PMID: 28179508 DOI: 10.1126/scitranslmed.aak9537] [Citation(s) in RCA: 326] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/16/2016] [Accepted: 12/02/2016] [Indexed: 12/12/2022]
Abstract
We report a method of cancer immunotherapy using an attenuated Salmonella typhimurium strain engineered to secrete Vibrio vulnificus flagellin B (FlaB) in tumor tissues. Engineered FlaB-secreting bacteria effectively suppressed tumor growth and metastasis in mouse models and prolonged survival. By using Toll-like receptor 5 (TLR5)-negative colon cancer cell lines, we provided evidence that the FlaB-mediated tumor suppression upon bacterial colonization is associated with TLR5-mediated host reactions in the tumor microenvironment. These therapeutic effects were completely abrogated in TLR4 and MyD88 knockout mice, and partly in TLR5 knockout mice, indicating that TLR4 signaling is a requisite for tumor suppression mediated by FlaB-secreting bacteria, whereas TLR5 signaling augmented tumor-suppressive host reactions. Tumor microenvironment colonization by engineered Salmonella appeared to induce the infiltration of abundant immune cells such as monocytes/macrophages and neutrophils via TLR4 signaling. Subsequent secretion of FlaB from colonizing Salmonella resulted in phenotypic and functional activation of intratumoral macrophages with M1 phenotypes and a reciprocal reduction in M2-like suppressive activities. Together, these findings provide evidence that nonvirulent tumor-targeting bacteria releasing multiple TLR ligands can be used as cancer immunotherapeutics.
Collapse
Affiliation(s)
- Jin Hai Zheng
- Laboratory of In Vivo Molecular Imaging, Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam 58128, Republic of Korea.,Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju 61469, Republic of Korea
| | - Vu H Nguyen
- Laboratory of In Vivo Molecular Imaging, Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam 58128, Republic of Korea.,Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Sheng-Nan Jiang
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.,Department of Nuclear Medicine, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Hainan 570-208, China
| | - Seung-Hwan Park
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Wenzhi Tan
- Department of Microbiology and Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Seol Hee Hong
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Myung Geun Shin
- Department of Laboratory Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Ik-Joo Chung
- Department of Hemato-Oncology, Chonnam National University Medical School, Jeonnam 58128, Republic of Korea
| | - Yeongjin Hong
- Department of Microbiology and Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Hee-Seung Bom
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Hyon E Choy
- Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju 61469, Republic of Korea.,Department of Microbiology and Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Shee Eun Lee
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Joon Haeng Rhee
- Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju 61469, Republic of Korea. .,Department of Microbiology and Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Jung-Joon Min
- Laboratory of In Vivo Molecular Imaging, Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam 58128, Republic of Korea. .,Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju 61469, Republic of Korea.,Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.,Department of Microbiology and Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
35
|
Basu P, Pal RR, Dasgupta S, Bhadra RK. DksA-HapR-RpoS axis regulates haemagglutinin protease production in Vibrio cholerae. MICROBIOLOGY-SGM 2017; 163:900-910. [PMID: 28597815 DOI: 10.1099/mic.0.000469] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
DksA acts as a co-factor for the intracellular small signalling molecule ppGpp during the stringent response. We recently reported that the expression of the haemagglutinin protease (HAP), which is needed for shedding of the cholera pathogen Vibrio cholerae during the late phase of infection, is significantly downregulated in V. cholerae ∆dksA mutant (∆dksAVc) cells. So far, it has been shown that HAP production by V. cholerae cells is critically regulated by HapR and also by RpoS. Here, we provide evidence that V. cholerae DksA (DksAVc) positively regulates HapR at both the transcriptional and post-transcriptional levels. We show that in ∆dksAVc cells the CsrB/C/D sRNAs, required for the maintenance of intracellular levels of hapR transcripts during the stationary growth, are distinctly downregulated. Moreover, the expression of exponential phase regulatory protein Fis, a known negative regulator of HapR, was found to continue even during the stationary phase in ∆dksAVc cells compared to that of wild-type strain, suggesting another layer of complex regulation of HapR by DksAVc. Extensive reporter construct-based and quantitative reverse-transcriptase PCR (qRT-PCR) analyses supported that RpoS is distinctly downregulated at the post-transcriptional/translational levels in stationary phase-grown ∆dksAVc cells. Since HAP expression through HapR and RpoS is stationary phase-specific in V. cholerae, it appears that DksAVc is also a critical stationary phase regulator for fine tuning of the expression of HAP. Moreover, experimental evidence provided in this study clearly supports that DksAVc is sitting at the top of the hierarchy of regulation of expression of HAP in V. cholerae.
Collapse
Affiliation(s)
- Pallabi Basu
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata-700 032, India
| | - Ritesh Ranjan Pal
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata-700 032, India
- Present address: Department of Microbiology and Molecular Genetics, Hebrew University of Jerusalem, Israel
| | - Shreya Dasgupta
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata-700 032, India
- Present address: Department of Clinical Medicine, National Institute of Cholera and Enteric Diseases, India
| | - Rupak K Bhadra
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata-700 032, India
| |
Collapse
|
36
|
Lim D, Kim KS, Kim H, Ko KC, Song JJ, Choi JH, Shin M, Min JJ, Jeong JH, Choy HE. Anti-tumor activity of an immunotoxin (TGFα-PE38) delivered by attenuated Salmonella typhimurium. Oncotarget 2017; 8:37550-37560. [PMID: 28473665 PMCID: PMC5514929 DOI: 10.18632/oncotarget.17197] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 04/06/2017] [Indexed: 12/22/2022] Open
Abstract
The anticancer strategy underlying the use of immunotoxins is as follows: the cancer-binding domain delivers the toxin to a cancer cell, after which the toxin enters and kills the cell. TGFα-PE38 is an immunotoxin comprising transforming growth factor alpha (TGFα), a natural ligand of epidermal growth factor receptor (EGFR), and a modified Pseudomonas exotoxin A (PE38) lacking N terminal cell-binding domain, a highly potent cytotoxic protein moiety. Tumor cells with high level of EGFR undergo apoptosis upon treatment with TGFα-PE38. However, clinical trials demonstrated that this immunotoxin delivered by an intracerebral infusion technique has only a limited inhibitory effect on intracranial tumors mainly due to inconsistent drug delivery. To circumvent this problem, we turned to tumor-seeking bacterial system. Here, we engineered Salmonella typhimurium to selectively express and release TGFα-PE38. Engineered bacteria were administered to mice implanted with mouse colon or breast tumor cells expressing high level of EGFR. We observed that controlled expression and release of TGFα-PE38 from intra-tumoral Salmonellae by either an engineered phage lysis system or by a bacterial membrane transport signal led to significant inhibition of solid tumor growth. These results demonstrated that delivery by tumor-seeking bacteria would greatly augment efficacy of immunotoxin in cancer therapeutics.
Collapse
Affiliation(s)
- Daejin Lim
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Molecular Medicine, BK21 Plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Kwang Soo Kim
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Molecular Medicine, BK21 Plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Hyunju Kim
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Molecular Medicine, BK21 Plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Kyong-Cheol Ko
- Applied Microbiology Research Center, Bio-Materials Research Institute, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Jeonbuk, Republic of Korea
| | - Jae Jun Song
- Applied Microbiology Research Center, Bio-Materials Research Institute, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Jeonbuk, Republic of Korea
| | - Jong Hyun Choi
- Applied Microbiology Research Center, Bio-Materials Research Institute, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Jeonbuk, Republic of Korea
| | - Minsang Shin
- Department of Microbiology, Kyungpook National University Medical School, Daegu, Republic of Korea
| | - Jung-joon Min
- Molecular Medicine, BK21 Plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Molecular Medicine, BK21 Plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Hyon E. Choy
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Molecular Medicine, BK21 Plus, Chonnam National University Graduate School, Gwangju, Republic of Korea
| |
Collapse
|
37
|
Ning BT, Yu B, Chan S, Chan JL, Huang JD, Chan GCF. Treatment of Neuroblastoma with an Engineered "Obligate" Anaerobic Salmonella typhimurium Strain YB1. J Cancer 2017; 8:1609-1618. [PMID: 28775780 PMCID: PMC5535716 DOI: 10.7150/jca.18776] [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: 12/27/2016] [Accepted: 02/27/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose Neuroblastoma is an embryonic solid tumor derived from the progenitors of the sympathetic nervous system. More than half of the patients developed metastatic disease at the time of initial diagnosis and had poor outcome with current therapeutic approaches. In recent years, some obligate and facultative anaerobic bacteria were reported to target the hypoxic and necrotic region of solid tumor models and caused tumor regression. We recently successfully constructed an "obligate" anaerobic Salmonella strain YB1 that was applied in breast cancer nude mice model by us. Here, we report the application of YB1 in neuroblastoma treatment. Methods The anti-cancer effect and side-effects of YB1 was examined in both in vitro and in vivo experiment. Previous established orthotopic neuroblastoma SCID/beige murine model using SK-NLP/luciferase cell line was adopted. ResultsIn vitro, YB1 induced apoptosis for up to 31.4% of the neuroblastoma cells under anaerobic condition, three times more than that under aerobic condition (10.9%). The expression of both Toll like Receptor 4 and 5 (TLR4 and TLR5) in cancer cells were significantly up-regulated (p<0.05, p<0.01 respectively) after the treatment of YB1 under anaerobic condition. In mouse model, YB1 preferentially accumulated inside the core of the tumors, rather than in normal tissues as our previous reported. This is suggestive of the hypoxic nature of tumor core. Tumor growth was significantly retarded in YB1 treatment group (n=6, P<0.01). Furthermore, there was no long-term organ damage noted in all the organs examined including heart, lung, liver, spleen and brain in the YB1 treated mice. Conclusion The genetic modified Salmonella strain YB1 is a promising anti-tumor strategy against the tumor bulk for neuroblastoma. Future study can be extended to other common cancer types to verify the relative efficacy on different neoplastic cells.
Collapse
Affiliation(s)
- Bo-Tao Ning
- Pediatric Intensive Care Unit, Shanghai Children's medical Center affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.,Department of Peadiatrics & Adolescent, Queen Mary Hospital, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Bin Yu
- Department of Biochemistry, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Shing Chan
- Department of Peadiatrics & Adolescent, Queen Mary Hospital, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Jian-Liang Chan
- Department of Peadiatrics & Adolescent, Queen Mary Hospital, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Jian-Dong Huang
- Department of Biochemistry, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Godfrey Chi-Fung Chan
- Department of Peadiatrics & Adolescent, Queen Mary Hospital, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China.,Center of Cancer Research, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| |
Collapse
|
38
|
Zheng JH, Min JJ. Targeted Cancer Therapy Using Engineered Salmonella typhimurium. Chonnam Med J 2016; 52:173-84. [PMID: 27689027 PMCID: PMC5040766 DOI: 10.4068/cmj.2016.52.3.173] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 07/25/2016] [Accepted: 08/02/2016] [Indexed: 12/26/2022] Open
Abstract
Obligate or facultative anaerobic bacteria such as Bifidobacterium, Clostridium, Salmonella, or Escherichia coli specifically colonize and proliferate inside tumor tissues and inhibit tumor growth. Among them, attenuated Salmonella typhimurium (S. typhimurium) has been widely studied in animal cancer models and Phase I clinical trials in human patients. S. typhimurium genes are easily manipulated; thus diverse attenuated strains of S. typhimurium have been designed and engineered as tumor-targeting therapeutics or drug delivery vehicles that show both an excellent safety profile and therapeutic efficacy in mouse models. An attenuated strain of S. typhimurium, VNP20009, successfully targeted human metastatic melanoma and squamous cell carcinoma in Phase I clinical trials; however, the efficacy requires further refinement. Along with the characteristics of self-targeting, proliferation, and deep tissue penetration, the ease of genetic manipulation allows for the production of more attenuated strains with greater safety profiles and vector systems that deliver designable cargo molecules for cancer diagnosis and/or therapy. Here, we discuss recent progress in the field of Salmonellae-mediated cancer therapy.
Collapse
Affiliation(s)
- Jin Hai Zheng
- Laboratory of In Vivo Molecular Imaging, Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, Korea
| | - Jung-Joon Min
- Laboratory of In Vivo Molecular Imaging, Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, Korea.; Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
39
|
Park SH, Zheng JH, Nguyen VH, Jiang SN, Kim DY, Szardenings M, Min JH, Hong Y, Choy HE, Min JJ. RGD Peptide Cell-Surface Display Enhances the Targeting and Therapeutic Efficacy of Attenuated Salmonella-mediated Cancer Therapy. Theranostics 2016; 6:1672-82. [PMID: 27446500 PMCID: PMC4955065 DOI: 10.7150/thno.16135] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 05/16/2016] [Indexed: 01/10/2023] Open
Abstract
Bacteria-based anticancer therapies aim to overcome the limitations of current cancer therapy by actively targeting and efficiently removing cancer. To achieve this goal, new approaches that target and maintain bacterial drugs at sufficient concentrations during the therapeutic window are essential. Here, we examined the tumor tropism of attenuated Salmonella typhimurium displaying the RGD peptide sequence (ACDCRGDCFCG) on the external loop of outer membrane protein A (OmpA). RGD-displaying Salmonella strongly bound to cancer cells overexpressing αvβ3, but weakly bound to αvβ3-negative cancer cells, suggesting the feasibility of displaying a preferential homing peptide on the bacterial surface. In vivo studies revealed that RGD-displaying Salmonellae showed strong targeting efficiency, resulting in the regression in αvβ3-overexpressing cancer xenografts, and prolonged survival of mouse models of human breast cancer (MDA-MB-231) and human melanoma (MDA-MB-435). Thus, surface engineering of Salmonellae to display RGD peptides increases both their targeting efficiency and therapeutic effect.
Collapse
|
40
|
Phan TX, Nguyen VH, Duong MTQ, Hong Y, Choy HE, Min JJ. Activation of inflammasome by attenuated Salmonella typhimurium in bacteria-mediated cancer therapy. Microbiol Immunol 2016; 59:664-75. [PMID: 26500022 DOI: 10.1111/1348-0421.12333] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 10/13/2015] [Accepted: 10/19/2015] [Indexed: 12/13/2022]
Abstract
Escherichia coli and attenuated Salmonella both naturally accumulate in a tumor mass, yet have distinct therapeutic efficacy: the E. coli K-12 strain (MG1655) cannot induce as significant a tumor suppression as attenuated Salmonella typhimurium, despite similar levels of accumulation in the tumor. To elucidate the mechanism of the robust antitumor effect of S. typhimurium, the cytokine profiles elicited by bacterial colonization in tumors were analyzed. C57BL/6 mice bearing MC38 tumors were injected with Salmonella or MG1655 in the tail vein. Tumors were collected 3 days post-infection and homogenized. Inflammasome-related signals were measured by real-time PCR, ELISA and western blot analysis. Only attenuated Salmonella triggered significant levels of the inflammatory cytokine IL-1β in the tumor, whereas tumor growth was significantly suppressed. In addition, transcript levels of the core molecules of inflammasome signaling, IPAF, NLRP3 and P2X7, were significantly elevated only in Salmonella-treated tumors. Upon direct interaction between Salmonella and BMDM, BMDM expressed inflammasome-related proteins such as NLRP3, IPAF and caspase-1 p10, and secreted a significant amount of IL-1β in supernatants. Coincubation assays with BMDM and Salmonella-treated MC38 cells (damaged cancer cells) revealed secretion of IL-1β only when TLR4 and inflammasome were activated by both LPS and damaged cancer cells. ATP released from damaged cancer cells was also identified as a mechanism of NLRP3 activation. In conclusion, Salmonella activate the inflammasome pathway using damage signals released from cancer cells and through direct interaction with macrophages.
Collapse
Affiliation(s)
- Thuy Xuan Phan
- Laboratory of In Vivo Imaging, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital
| | - Vu Hong Nguyen
- Laboratory of In Vivo Imaging, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital
| | - Mai Thi-Quynh Duong
- Laboratory of In Vivo Imaging, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital
| | - Yeongjin Hong
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Korea
| | - Hyon E Choy
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Korea
| | - Jung-Joon Min
- Laboratory of In Vivo Imaging, Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital.,Department of Microbiology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
41
|
Sridhar S, Steele-Mortimer O. Inherent Variability of Growth Media Impacts the Ability of Salmonella Typhimurium to Interact with Host Cells. PLoS One 2016; 11:e0157043. [PMID: 27280414 PMCID: PMC4900594 DOI: 10.1371/journal.pone.0157043] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/24/2016] [Indexed: 12/20/2022] Open
Abstract
Efficient invasion of non-phagocytic cells, such as intestinal epithelial cells, by Salmonella Typhimurium is dependent on the Salmonella Pathogenicity Island 1 (SPI-1)-encoded Type Three Secretion System. The environmental cues involved in SPI-1 induction are not well understood. In vitro, various conditions are used to induce SPI-1 and the invasive phenotype. Although lysogeny broth (LB) is widely used, multiple formulations exist, and variation can arise due to intrinsic differences in complex components. Minimal media are also susceptible to variation. Still, the impact of these inconsistencies on Salmonella virulence gene expression has not been well studied. The goal of this project is to identify growth conditions in LB and minimal medium that affect SPI-1 induction in vitro using both whole population and single cell analysis. Here we show, using a fluorescent reporter of the SPI-1 gene prgH, that growth of Salmonella in LB yields variable induction. Deliberate modification of media components can influence the invasive profile. Finally, we demonstrate that changes in SPI-1 inducing conditions can affect the ability of Salmonella to replicate intracellularly. These data indicate that the specific media growth conditions impact how the bacteria interact with host cells.
Collapse
Affiliation(s)
- Sushmita Sridhar
- Laboratory of Bacteriology, Rocky Mountain Labs, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Olivia Steele-Mortimer
- Laboratory of Bacteriology, Rocky Mountain Labs, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
- * E-mail:
| |
Collapse
|
42
|
Zhang T, Zhu J, Wei S, Luo Q, Li L, Li S, Tucker A, Shao H, Zhou R. The roles of RelA/(p)ppGpp in glucose-starvation induced adaptive response in the zoonotic Streptococcus suis. Sci Rep 2016; 6:27169. [PMID: 27255540 PMCID: PMC4891663 DOI: 10.1038/srep27169] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 05/16/2016] [Indexed: 01/02/2023] Open
Abstract
The (p)ppGpp signal molecules play a central role in the stringent response (SR) to adapt to nutrient starvation in bacteria, yet the carbohydrate starvation induced adaptive response and the roles of SR in this response is not well characterized, especially in Gram-positives. Here, two (p)ppGpp synthetases RelA and RelQ are identified in Streptococcus suis, an important emerging zoonotic Gram-positive bacterium, while only RelA is functional under glucose starvation. To characterize the roles of RelA/(p)ppGpp in glucose starvation response in S. suis, the growth curves and transcriptional profiles were compared between the mutant strain ΔrelA [a (p)ppGpp0 strain under glucose starvation] and its parental strain SC-19 [(p)ppGpp+]. The results showed great difference between SC-19 and ΔrelA on adaptive responses when suffering glucose starvation, and demonstrated that RelA/(p)ppGpp plays important roles in adaptation to glucose starvation. Besides the classic SR including inhibition of growth and related macromolecular synthesis, the extended adaptive response also includes inhibited glycolysis, and carbon catabolite repression (CCR)-mediated carbohydrate-dependent metabolic switches. Collectively, the pheno- and genotypic characterization of the glucose starvation induced adaptive response in S. suis makes a great contribution to understanding better the mechanism of SR.
Collapse
Affiliation(s)
- Tengfei Zhang
- State Key Laboratory of Agricultural Microbiology and Key Laboratory of Veterinary Diagnosis (Ministry of Agriculture), College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.,Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Jiawen Zhu
- State Key Laboratory of Agricultural Microbiology and Key Laboratory of Veterinary Diagnosis (Ministry of Agriculture), College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Shun Wei
- State Key Laboratory of Agricultural Microbiology and Key Laboratory of Veterinary Diagnosis (Ministry of Agriculture), College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingping Luo
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Lu Li
- State Key Laboratory of Agricultural Microbiology and Key Laboratory of Veterinary Diagnosis (Ministry of Agriculture), College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan 430070, China
| | - Shengqing Li
- Department of Chemistry, College of Science, Huazhong Agricultural University, Wuhan 430070, China
| | - Alexander Tucker
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Huabin Shao
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology and Key Laboratory of Veterinary Diagnosis (Ministry of Agriculture), College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan 430070, China
| |
Collapse
|
43
|
Intestinal Long-Chain Fatty Acids Act as a Direct Signal To Modulate Expression of the Salmonella Pathogenicity Island 1 Type III Secretion System. mBio 2016; 7:e02170-15. [PMID: 26884427 PMCID: PMC4752608 DOI: 10.1128/mbio.02170-15] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Salmonella enterica serovar Typhimurium uses the Salmonella pathogenicity island 1 (SPI1) type III secretion system (T3SS) to induce inflammatory diarrhea and bacterial uptake into intestinal epithelial cells. The expression of hilA, encoding the transcriptional activator of the T3SS structural genes, is directly controlled by three AraC-like regulators, HilD, HilC, and RtsA, each of which can activate hilD, hilC, rtsA, and hilA genes, forming a complex feed-forward regulatory loop. Expression of the SPI1 genes is tightly controlled by numerous regulatory inputs to ensure proper timing in production of the T3SS apparatus. Loss of FadD, an acyl coenzyme A (acyl-CoA) synthetase required for degradation of long-chain fatty acids (LCFAs), was known to decrease hilA expression. We show that free external LCFAs repress expression of hilA independently of FadD and the LCFA degradation pathway. Genetic and biochemical evidence suggests that LCFAs act directly to block primarily HilD activity. Further analyses show that in the absence of FadD, hilA expression is downregulated due to endogenous production of free LCFAs, which are excreted into the culture medium via TolC and then transported back into the bacterial cell via FadL. A fadL mutant is more virulent than the wild-type strain in mouse oral competition assays independently of LCFA degradation, showing that, in the host, dietary LCFAs serve as a signal for proper regulation of SPI1 expression, rather than an energy source. To cause disease, Salmonella must respond to diverse environmental cues to express its invasion machinery at the appropriate location in the host intestine. We show that host intestinal free long-chain fatty acids (LCFAs) affect Salmonella invasion by reducing expression of the SPI1 type III secretion system, acting primarily via the AraC-like activator HilD. Degradation of LCFAs is not required for this regulation, showing that free LCFAs serve as a cue to proper intestinal localization to invade host epithelial cells and not as a nutrient source.
Collapse
|
44
|
Zheng LL, Huang CZ. Selective and sensitive colorimetric detection of stringent alarmone ppGpp with Fenton-like reagent. Analyst 2015; 139:6284-9. [PMID: 25315398 DOI: 10.1039/c4an01632g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stringent alarmone, namely, guanosine 3'-diphosphate-5'-diphosphate (ppGpp), is a global regulator that plays a critical role in the survival, growth, metabolism, and many other vital processes of microorganisms. Because of its structural similarity to normal nucleotides, it is also a challenge for the selective and sensitive detection of ppGpp nowadays. Herein, we developed a colorimetric method for the selective detection of ppGpp by inhibiting the redox reaction between Fenton-like reagent (composed of Fe(3+) and H2O2) with 2,2'-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid) (ABTS). Owing to the strong coordination affinity between ppGpp and Fe(3+), the chromogenic reaction between ABTS and Fenton-like reagent, occurred in aqueous medium at 37 °C and resulted in a bluish-green solution, which was inhibited with the addition of ppGpp. This phenomenon forms the basis for the colorimetric detection of ppGpp, with a detection limit of 0.19 μM and good selectivity for ppGpp over other nucleotides and anions. Furthermore, the results could be visualized by the naked eye, and the sensitivity of the naked-eye observation could even be further improved with the aid of the introduction of a background color.
Collapse
Affiliation(s)
- Lin Ling Zheng
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China.
| | | |
Collapse
|
45
|
Kim JE, Phan TX, Nguyen VH, Dinh-Vu HV, Zheng JH, Yun M, Park SG, Hong Y, Choy HE, Szardenings M, Hwang W, Park JA, Park S, Im SH, Min JJ. Salmonella typhimurium Suppresses Tumor Growth via the Pro-Inflammatory Cytokine Interleukin-1β. Am J Cancer Res 2015; 5:1328-42. [PMID: 26516371 PMCID: PMC4615736 DOI: 10.7150/thno.11432] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 08/23/2015] [Indexed: 12/23/2022] Open
Abstract
Although strains of attenuated Salmonella typhimurium and wild-type Escherichia coli show similar tumor-targeting capacities, only S. typhimurium significantly suppresses tumor growth in mice. The aim of the present study was to examine bacteria-mediated immune responses by conducting comparative analyses of the cytokine profiles and immune cell populations within tumor tissues colonized by E. coli or attenuated Salmonellae. CT26 tumor-bearing mice were treated with two different bacterial strains: S. typhimurium defective in ppGpp synthesis (ΔppGpp Salmonellae) or wild-type E. coli MG1655. Cytokine profiles and immune cell populations in tumor tissue colonized by these two bacterial strains were examined at two time points based on the pattern of tumor growth after ΔppGpp Salmonellae treatment: 1) when tumor growth was suppressed ('suppression stage') and 2) when they began to re-grow ('re-growing stage'). The levels of IL-1β and TNF-α were markedly increased in tumors colonized by ΔppGpp Salmonellae. This increase was associated with tumor regression; the levels of both IL-1β and TNF-α returned to normal level when the tumors started to re-grow. To identify the immune cells primarily responsible for Salmonellae-mediated tumor suppression, we examined the major cell types that produce IL-1β and TNF-α. We found that macrophages and dendritic cells were the main producers of TNF-α and IL-1β. Inhibiting IL-1β production in Salmonellae-treated mice restored tumor growth, whereas tumor growth was suppressed for longer by local administration of recombinant IL-1β or TNF-α in conjunction with Salmonella therapy. These findings suggested that IL-1β and TNF-α play important roles in Salmonella-mediated cancer therapy. A better understanding of host immune responses in Salmonella therapy may increase the success of a given drug, particularly when various strategies are combined with bacteriotherapy.
Collapse
|
46
|
Chen JQ, Zhan YF, Wang W, Jiang SN, Li XY. The engineered Salmonella typhimurium inhibits tumorigenesis in advanced glioma. Onco Targets Ther 2015; 8:2555-63. [PMID: 26451114 PMCID: PMC4592054 DOI: 10.2147/ott.s86899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Objective To explore the antitumor role of the attenuated Salmonella typhimurium ΔppGpp with inducible cytolysin A (ClyA) in advanced stage of glioma. Materials and methods The C6 rat glioma cells were orthotopically implanted by surgery into the caudate nucleus of rat brains. The rats were then randomly divided into the treatment group (SL + ClyA) (n=12), negative control group (SL) (n=12), and control group (phosphate-buffered saline [PBS]) (n=12). In the treatment group, the attenuated S. typhimurium were transformed with the plasmid-encoded antitumor gene ClyA. The expression of ClyA was controlled by the TetR-regulated promoter in response to extracellular doxycycline. The plasmid also contained an imaging gene lux to allow illumination of the tumor infected by the bacteria. The rat glioma C6 cells were implanted into the caudate nucleus of all rats. The engineered S. typhimurium and respective controls were injected intravenously into the rats 21 days after initial tumor implantation. The pathological analysis of the glioma tumor was performed at 21 days and 28 days (7 days after doxycycline treatment) postimplantation. All rats underwent MRI (magnetic resonance imaging) and bioluminescence study at 21 days and 28 days postimplantation to detect tumor volume. The differences between the three groups in tumor volume and survival time were analyzed. Results Advanced stage glioma was detected at 21 days postimplantation. Bioluminescence showed that the engineered S. typhimurium accumulated in glioma tumors and disappeared in the normal reticuloendothelial tissues 3 days after intravenous injection. MRI showed that the tumor volume in the S. typhimurium with ClyA group were significantly reduced compared to the bacteria alone and no bacteria groups 7 days post-doxycycline treatment (P<0.05), while the necrotic tumor volume in the S. typhimurium with ClyA group and S. typhimurium alone group increased significantly compared to the control group (P<0.01). In addition, the survival time was significantly prolonged in the bacteria-treated group compared to the PBS-treated control group (P<0.01). Conclusion The engineered S. typhimurium can significantly induce cancer cell apoptosis in the tumor center and inhibit cancer cell proliferation in the outer zone of advanced glioma tumor, leading to a prolonged survival time in rats. In addition, the engineered S. typhimurium that carried the antitumor and imaging genes controlled by the TetR-regulated promoter have high delivery efficiency with tolerable side effects in rats.
Collapse
Affiliation(s)
- Jian-Qiang Chen
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yue-Fu Zhan
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University, Haikou, Hainan, People's Republic of China
| | - Wei Wang
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Sheng-Nan Jiang
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University, Haikou, Hainan, People's Republic of China ; Department of Nuclear Medicine, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University, Haikou, Hainan, People's Republic of China
| | - Xiang-Ying Li
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University, Haikou, Hainan, People's Republic of China
| |
Collapse
|
47
|
Hong H, Lim D, Kim GJ, Park SH, Sik Kim H, Hong Y, Choy HE, Min JJ. Targeted deletion of the ara operon of Salmonella typhimurium enhances L-arabinose accumulation and drives PBAD-promoted expression of anti-cancer toxins and imaging agents. Cell Cycle 2015; 13:3112-20. [PMID: 25486570 DOI: 10.4161/15384101.2014.949527] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumor-specific expression of antitumor drugs can be achieved using attenuated Salmonella typhimurium harboring the PBAD promoter, which is induced by L-arabinose. However, L-arabinose does not accumulate because it is metabolized to D-xylulose-5-P by enzymes encoded by the ara operon in Salmonellae. To address this problem, we developed an engineered strain of S. typhimurium in which the ara operon is deleted. Linear DNA transformation was performed using λ red recombinase to exchange the ara operon with linear DNA carrying an antibiotic-resistance gene with homology to regions adjacent to the ara operon. The ara operon-deleted strain and its parental strain were transformed with a plasmid encoding Renilla luciferase variant 8 (RLuc8) or cytolysin A (clyA) under the control of the PBAD promoter. Luciferase assays demonstrated that RLuc8 expression was 49-fold higher in the ara operon-deleted S. typhimurium than in the parental strain after the addition of L-arabinose. In vivo bioluminescence imaging showed that the tumor tissue targeted by the ara operon-deleted Salmonella had a stronger imaging signal (~30-fold) than that targeted by the parental strain. Mice with murine colon cancer (CT26) that had been injected with the ara operon-deleted S. typhimurium expressing clyA showed significant tumor suppression. The present report demonstrates that deletion of the ara operon of S. typhimurium enhances L-arabinose accumulation and thereby drives PBAD-promoted expression of cytotoxic agents and imaging agents. This is a promising approach for tumor therapy and imaging.
Collapse
Affiliation(s)
- Hyun Hong
- a Department of Nuclear Medicine ; Chonnam National University Medical School and Hwasun Hospital ; Jeonnam , Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Tennant SM, Levine MM. Live attenuated vaccines for invasive Salmonella infections. Vaccine 2015; 33 Suppl 3:C36-41. [PMID: 25902362 PMCID: PMC4469493 DOI: 10.1016/j.vaccine.2015.04.029] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/06/2015] [Accepted: 04/08/2015] [Indexed: 02/04/2023]
Abstract
Salmonella enterica serovar Typhi produces significant morbidity and mortality worldwide despite the fact that there are licensed Salmonella Typhi vaccines available. This is primarily due to the fact that these vaccines are not used in the countries that most need them. There is growing recognition that an effective invasive Salmonella vaccine formulation must also prevent infection due to other Salmonella serovars. We anticipate that a multivalent vaccine that targets the following serovars will be needed to control invasive Salmonella infections worldwide: Salmonella Typhi, Salmonella Paratyphi A, Salmonella Paratyphi B (currently uncommon but may become dominant again), Salmonella Typhimurium, Salmonella Enteritidis and Salmonella Choleraesuis (as well as other Group C Salmonella). Live attenuated vaccines are an attractive vaccine formulation for use in developing as well as developed countries. Here, we describe the methods of attenuation that have been used to date to create live attenuated Salmonella vaccines and provide an update on the progress that has been made on these vaccines.
Collapse
Affiliation(s)
- Sharon M Tennant
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Myron M Levine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
49
|
L-Asparaginase delivered by Salmonella typhimurium suppresses solid tumors. MOLECULAR THERAPY-ONCOLYTICS 2015; 2:15007. [PMID: 27119104 PMCID: PMC4845971 DOI: 10.1038/mto.2015.7] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 03/02/2015] [Indexed: 01/16/2023]
Abstract
Bacteria can be engineered to deliver anticancer proteins to tumors via a controlled expression system that maximizes the concentration of the therapeutic agent in the tumor. L-asparaginase (L-ASNase), which primarily converts asparagine to aspartate, is an anticancer protein used to treat acute lymphoblastic leukemia. In this study, Salmonellae were engineered to express L-ASNase selectively within tumor tissues using the inducible araBAD promoter system of Escherichia coli. Antitumor efficacy of the engineered bacteria was demonstrated in vivo in solid malignancies. This result demonstrates the merit of bacteria as cancer drug delivery vehicles to administer cancer-starving proteins such as L-ASNase to be effective selectively within the microenvironment of cancer tissue.
Collapse
|
50
|
Rice CJ, Ramachandran VK, Shearer N, Thompson A. Transcriptional and Post-Transcriptional Modulation of SPI1 and SPI2 Expression by ppGpp, RpoS and DksA in Salmonella enterica sv Typhimurium. PLoS One 2015; 10:e0127523. [PMID: 26039089 PMCID: PMC4454661 DOI: 10.1371/journal.pone.0127523] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/16/2015] [Indexed: 12/02/2022] Open
Abstract
The expression of genes within Salmonella Pathogenicity Islands 1 and 2 (SPI1, SPI2) is required to facilitate invasion and intracellular replication respectively of S. Typhimurium in host cell lines. Control of their expression is complex and occurs via a variety of factors operating at transcriptional and post-transcriptional levels in response to the environmental stimuli found within the host. Several of the factors that modulate SPI1 and SPI2 expression are involved in the redistribution or modification of RNA polymerase (RNAP) specificity. These factors include the bacterial alarmone, ppGpp, the alternative sigma factor, RpoS, and the RNAP accessory protein, DksA. In this report we show not only how these three factors modulate SPI1 and SPI2 expression but also how they contribute to the 'phased' expression of SPI1 and SPI2 during progress through late-log and stationary phase in aerobic rich broth culture conditions. In addition, we demonstrate that the expression of at least one SPI1-encoded protein, SipC is subject to DksA-dependent post-transcriptional control.
Collapse
Affiliation(s)
| | | | - Neil Shearer
- Institute of Food Research, Norwich, NR4 7UA, United Kingdom
| | - Arthur Thompson
- Institute of Food Research, Norwich, NR4 7UA, United Kingdom
| |
Collapse
|