1
|
Xie Y, Wang J, Wang Y, Wen Y, Pu Y, Wang B. Parasite-enhanced immunotherapy: transforming the "cold" tumors to "hot" battlefields. Cell Commun Signal 2024; 22:448. [PMID: 39327550 PMCID: PMC11426008 DOI: 10.1186/s12964-024-01822-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/08/2024] [Indexed: 09/28/2024] Open
Abstract
Immunotherapy has emerged as a highly effective treatment for various tumors. However, the variable response rates associated with current immunotherapies often restrict their beneficial impact on a subset of patients. Therefore, more effective treatment approaches that can broaden the scope of therapeutic benefits to a larger patient population are urgently needed. Studies have shown that some parasites and their products, for example, Plasmodium, Toxoplasma, Trypanosoma, and Echinococcus, can effectively transform "cold" tumors into "hot" battlefields and reshape the tumor microenvironment, thereby stimulating innate and adaptive antitumor immune responses. These parasitic infections not only achieve the functional reversal of innate immune cells, such as neutrophils, macrophages, myeloid-derived suppressor cells, regulatory T cells, and dendritic cells, in tumors but also successfully activate CD4+/CD8+ T cells and even B cells to produce antibodies, ultimately resulting in an antitumor-specific immune response and antibody-dependent cellular cytotoxicity. Animal studies have confirmed these findings. This review discusses the abovementioned content and the challenges faced in the future clinical application of antitumor treatment strategies based on parasitic infections. With the potential of these parasites and their byproducts to function as anticancer agents, we anticipate that further investigations in this field could yield significant advancements in cancer treatment.
Collapse
Affiliation(s)
- Yujun Xie
- Laboratory of Tumor Immunobiology, Department of Public Health and Pathogen Biology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Jinyan Wang
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, 110122, China
| | - Yafei Wang
- Faculty of Arts and Science, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Yalin Wen
- Laboratory of Tumor Immunobiology, Department of Public Health and Pathogen Biology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Yanping Pu
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Benfan Wang
- Laboratory of Tumor Immunobiology, Department of Public Health and Pathogen Biology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
- Institute of Surgery, The First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China.
| |
Collapse
|
2
|
Lotfalizadeh N, Sadr S, Morovati S, Lotfalizadeh M, Hajjafari A, Borji H. A potential cure for tumor-associated immunosuppression by Toxoplasma gondii. Cancer Rep (Hoboken) 2024; 7:e1963. [PMID: 38109851 PMCID: PMC10850000 DOI: 10.1002/cnr2.1963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/06/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Recently, immunotherapy has become very hopeful for cancer therapy. Cancer treatment through immunotherapy has excellent specificity and less toxicity than conventional chemoradiotherapy. Pathogens have been used in cancer immunotherapy for a long time. The current study aims to evaluate the possibility of Toxoplasma gondii (T. gondii) as a probable treatment for cancers such as melanoma, breast, ovarian, lung, and pancreatic cancer. RECENT FINDINGS Nonreplicating type I uracil auxotrophic mutants of T. gondii can stimulate immune responses against tumors by reverse immunosuppression at the cellular level. T. gondii can be utilized to research T helper 1 (Th1) cell immunity in intracellular infections. Avirulent T. gondii uracil auxotroph vaccine can change the tumor's immunosuppression and improve the production of type 1 helper cell cytokines, i.e., Interferon-gamma (IFN-γ) and Interleukin-12 (IL-12) and activate tumor-related Cluster of Differentiation 8 (CD8+) T cells to identify and destroy cancer cells. The T. gondii profilin protein, along with T. gondii secreted proteins, have been found to exhibit promising properties in the treatment of various cancers. These proteins are being studied for their potential to inhibit tumor growth and enhance the effectiveness of cancer therapies. Their unique mechanisms of action make them valuable candidates for targeted interventions in ovarian cancer, breast cancer, pancreatic cancer, melanoma, and lung cancer treatments. CONCLUSION In summary, the study underscores the significant potential of harnessing T. gondii, including its diverse array of proteins and antigens, particularly in its avirulent form, as a groundbreaking approach in cancer immunotherapy.
Collapse
Affiliation(s)
- Narges Lotfalizadeh
- Department of Pathobiology, Faculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | - Soheil Sadr
- Department of Pathobiology, Faculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | - Solmaz Morovati
- Division of Biotechnology, Department of Pathobiology, School of Veterinary MedicineShiraz UniversityShirazIran
| | - Mohammadhassan Lotfalizadeh
- Board Certificate Oral and Maxillofacial RadiologistNorth Khorasan University of Medical Sciences (NKUMS)BojnurdIran
| | - Ashkan Hajjafari
- Department of Pathobiology, Faculty of Veterinary MedicineIslamic Azad University, Science and Research BranchTehranIran
| | - Hassan Borji
- Department of Pathobiology, Faculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| |
Collapse
|
3
|
Zheng Z, Lu X, Zhou D, Deng XF, Liu QX, Liu XB, Zhang J, Li YQ, Zheng H, Dai JG. A novel enemy of cancer: recent investigations into protozoan anti-tumor properties. Front Cell Infect Microbiol 2024; 13:1325144. [PMID: 38274735 PMCID: PMC10808745 DOI: 10.3389/fcimb.2023.1325144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
Cancer remains a significant global health issue, despite advances in screening and treatment. While existing tumor treatment protocols such as surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy have proven effective in enhancing the prognosis for some patients, these treatments do not benefit all patients. Consequently, certain types of cancer continue to exhibit a relatively low 5-year survival rate. Therefore, the pursuit of novel tumor intervention strategies may help improve the current effectiveness of tumor treatment. Over the past few decades, numerous species of protozoa and their components have exhibited anti-tumor potential via immune and non-immune mechanisms. This discovery introduces a new research direction for the development of new and effective cancer treatments. Through in vitro experiments and studies involving tumor-bearing mice, the anti-tumor ability of Toxoplasma gondii, Plasmodium, Trypanosoma cruzi, and other protozoa have unveiled diverse mechanisms by which protozoa combat cancer, demonstrating encouraging prospects for their application. In this review, we summarize the anti-tumor ability and anti-tumor mechanisms of various protozoa and explore the potential for their clinical development and application.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army (Third Military) Medical University, Chongqing, China
| | - Ji-gang Dai
- Department of Thoracic Surgery, Xinqiao Hospital, Army (Third Military) Medical University, Chongqing, China
| |
Collapse
|
4
|
Mani R, Martin CG, Balu KE, Wang Q, Rychahou P, Izumi T, Evers BM, Suzuki Y. A Novel Protozoa Parasite-Derived Protein Adjuvant Is Effective in Immunization with Cancer Cells to Activate the Cancer-Specific Protective Immunity and Inhibit the Cancer Growth in a Murine Model of Colorectal Cancer. Cells 2024; 13:111. [PMID: 38247803 PMCID: PMC10814441 DOI: 10.3390/cells13020111] [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: 12/07/2023] [Revised: 12/31/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Cancer-specific CD8+ cytotoxic T cells play important roles in preventing cancer growth, and IFN-γ, in addition to IL-12 and type I interferon, is critical for activating CD8+ cytotoxic T cells. We recently identified the capability of the amino-terminus region of dense granule protein 6 (GRA6Nt) of Toxoplasma gondii, an intracellular protozoan parasite, to activate IFN-γ production of microglia, a tissue-resident macrophage population. Therefore, in the present study, we examined whether recombinant GRA6Nt protein (rGRA6Nt) functions as an effective adjuvant to potently activate cancer-specific protective immunity using a murine model of MC38 colorectal cancer (CRC). When mice were immunized with non-replicable (either treated with mitomycin C or irradiated by X-ray) MC38 CRC cells in combination with rGRA6Nt adjuvant and received a challenge implantation of replication-capable MC38 tumor cells, those mice markedly inhibited the growth of the implanted tumors in association with a two-fold increase in CD8+ T cell density within the tumors. In addition, CD8+ T cells of the immunized mice secreted significantly increased amounts of granzyme B, a key mediator of the cytotoxic activity of CD8+ T cells, and IFN-γ in response to MC38 CRC cells in vitro when compared to the T cells from unimmunized mice. Notably, the protective effects of the immunization were specific to MC38 CRC cells, as the immunized mice did not exhibit a significantly inhibited growth of EL4 lymphoma tumors. These results indicate that rGRA6Nt is a novel and effective protein adjuvant when used in immunizations with non-replicable cancer cells to potently activate the protective immunity specifically against the cancer cells employed in the immunization.
Collapse
Affiliation(s)
- Rajesh Mani
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA; (R.M.)
| | - Chloe G. Martin
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA; (R.M.)
| | - Kanal E. Balu
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA; (R.M.)
| | - Qingding Wang
- Department of Surgery, University of Kentucky, Lexington, KY 40536, USA (P.R.)
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Piotr Rychahou
- Department of Surgery, University of Kentucky, Lexington, KY 40536, USA (P.R.)
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Tadahide Izumi
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - B. Mark Evers
- Department of Surgery, University of Kentucky, Lexington, KY 40536, USA (P.R.)
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Yasuhiro Suzuki
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA; (R.M.)
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
5
|
El Skhawy N, Eissa MM. Shedding light on a mysterious link between Toxoplasma Gondii and cancer: A review. Exp Parasitol 2023; 250:108544. [PMID: 37149210 DOI: 10.1016/j.exppara.2023.108544] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/05/2023] [Accepted: 05/04/2023] [Indexed: 05/08/2023]
Abstract
The ongoing conflict regarding the affiliation of Toxoplasma gondii to cancer; whether an inducer or a suppressor needs to be resolved. Human epidemiological studies oscillate without attaining a firm ground. Some studies confirmed the detection of high seroprevalence of anti-Toxoplasma antibodies in different cancer patients without further justification whether being causation, co-incidences, or part of opportunistic infections. Others reported a state of resistance to cancer accompanying low titer of anti-Toxoplasma antibody. Worthwhile, preclinical experimental work confirmed the antineoplastic potency of Toxoplasma. Thus, further investigational research is essential to validate the potential application of Toxoplasma as a promising cancer immunotherapeutic vaccine candidate. In this paper, we present a review of this issue by examining epidemiological and preclinical experimental studies that explored the linkage between Toxoplasma gondii and cancer<i.></i> We consider this review an important step towards shedding a light on this mysterious link and a stepping-stone for potential research work addressing Toxoplasma as a cancer suppressor rather than a cancer inducer.
Collapse
Affiliation(s)
- Nahla El Skhawy
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Maha M Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
6
|
Kaur T, Sharma D. Fundamentals of utilizing microbes in advanced cancer therapeutics: Current understanding and potential applications. ADVANCES IN APPLIED MICROBIOLOGY 2023. [PMID: 37400175 DOI: 10.1016/bs.aambs.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
One of the biggest health related issues in the twenty-first century is cancer. The current therapeutic platforms have not advanced enough to keep up with the number of rising cases. The traditional therapeutic approaches frequently fail to produce the desired outcomes. Therefore, developing new and more potent remedies is crucial. Recently, investigating microorganisms as potential anti-cancer treatments have garnered a lot of attention. Tumor-targeting microorganisms are more versatile at inhibiting cancer than the majority of standard therapies. Bacteria preferentially gather and thrive inside tumors, where they can trigger anti-cancer immune responses. They can be further trained to generate and distribute anticancer drugs based on clinical requirements using straightforward genetic engineering approaches. To improve clinical outcomes, therapeutic strategies utilizing live tumor-targeting bacteria can be used either alone or in combination with existing anticancer treatments. On the other hand, oncolytic viruses that target cancer cells, gene therapy via viral vectors, and viral immunotherapy are other popular areas of biotechnological investigation. Therefore, viruses serve as a unique candidate for anti-tumor therapy. This chapter describes the role of microbes, primarily bacteria and viruses in anti-cancer therapeutics. The various approaches to utilizing microbes in cancer therapy are discussed and examples of microorganisms that are now in use or that are undergoing experimental research are briefly discussed. We further point out the hurdles and the prospects of microbes-based remedies for cancer treatment.
Collapse
|
7
|
Bahwal SA, Chen JJ, E L, Hao T, Chen J, Carruthers VB, Lai J, Zhou X. Attenuated Toxoplasma gondii enhances the antitumor efficacy of anti-PD1 antibody by altering the tumor microenvironment in a pancreatic cancer mouse model. J Cancer Res Clin Oncol 2022; 148:2743-2757. [PMID: 35556163 DOI: 10.1007/s00432-022-04036-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/19/2022] [Indexed: 01/05/2023]
Abstract
PURPOSE To investigate whether attenuated Toxoplasma is efficacious against solid tumors of pancreatic cancer and whether attenuated Toxoplasma improves the antitumor activity of αPD-1 antibody on pancreatic cancer. METHODS The therapeutic effects of attenuated Toxoplasma NRTUA strain monotherapy and combination therapy of NRTUA with anti-PD-1 antibody on PDAC tumor volume and tumor weight of Pan02 tumor-bearing mice were investigated. We characterized the effects of combination therapy of NRTUA with anti-PD-1 antibody on tumor-infiltrating lymphocytes and tumor-specific IFN-γ by using immunohistochemistry, flow cytometry and ELISA. The antitumor mechanisms of combination therapy of NRTUA with anti-PD-1 antibody were investigated via depletion of CD8+ T cells and IL-12. RESULTS NRTUA strain treatment inhibited tumor growth in a subcutaneous mouse model of PDAC through activating dendritic cells and increasing CD8+ T cell infiltration in the tumor microenvironment. More importantly, combination therapy of NRTUA with anti-PD-1 antibody elicited a significant antitumor immune response and synergistically controlled tumor growth in Pan02 tumor-bearing mice. Specifically, the combination treatment led to elevation of CD8+ T cell infiltration mediated by dendritic cell-secreted IL-12 and to tumor-specific IFN-γ production in the PDAC tumor microenvironment. Also, the combination treatment markedly reduced the immunosuppressive myeloid-derived suppressor cell population in PDAC mice. CONCLUSION These findings could provide a novel immunotherapy approach to treating solid tumors of PDAC and overcoming resistance to anti-PD-1 agents in PDAC tumors.
Collapse
Affiliation(s)
- Said Ahmed Bahwal
- Department of Biochemistry and Molecular Biology, Sun Yat-Sen University Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jane J Chen
- Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Lilin E
- Department of Biochemistry and Molecular Biology, Sun Yat-Sen University Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Taofang Hao
- Department of Biochemistry and Molecular Biology, Sun Yat-Sen University Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jiancong Chen
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Vern B Carruthers
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, MI, 48109-5620, USA.
| | - Jiaming Lai
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Xingwang Zhou
- Department of Biochemistry and Molecular Biology, Sun Yat-Sen University Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
8
|
Neospora caninum inhibits tumor development by activating the immune response and destroying tumor cells in a B16F10 melanoma model. Parasit Vectors 2022; 15:332. [PMID: 36138417 PMCID: PMC9503190 DOI: 10.1186/s13071-022-05456-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
Background Melanoma is a malignant tumor with a high mortality rate. Some microorganisms have been shown to activate the immune system and limit cancer progression. The objective of this study is to evaluate the anti-melanoma effect of Neospora caninum, a livestock pathogen with no pathogenic activity in humans. Methods Neospora caninum tachyzoites were inoculated into a C57BL/6 mouse melanoma model by intratumoral and distal subcutaneous injections. Tumor volumes were measured, and cell death areas were visualized by hematoxylin and eosin staining and quantified. Apoptosis in cell cultures and whole tumors was detected by propidium iodide (PI) and TUNEL staining, respectively. Cytokine and tumor-associated factor levels in tumors and spleens were detected by real-time quantitative polymerase chain reaction. Infiltration of macrophages and CD8+ T cells in the tumor microenvironment (TME) were detected by immunohistochemistry with anti-CD68 and anti-CD8 antibodies, respectively. Finally, 16S rRNA sequencing of mice cecal contents was performed to evaluate the effect of N. caninum on gut microbial diversity. Results Intratumoral and distal subcutaneous injections of N. caninum resulted in significant inhibition of tumor growth (P < 0.001), and more than 50% of tumor cells were dead without signs of apoptosis. Neospora caninum treatment significantly increased the mRNA expression levels of IL-12, IFN-γ, IL-2, IL-10, TNF-α, and PD-L1 in the TME, and IL-12 and IFN-γ in the spleen of tumor-bearing mice (P < 0.05). An increase in the infiltration of CD8+ T cells and macrophages in the TME was observed with these cytokine changes. Neospora caninum also restored the abundance of gut microbiota Lactobacillus, Lachnospiraceae, Adlercreutzia, and Prevotellaceae associated with tumor growth, but the changes were not significant. Conclusion Neospora caninum inhibits B16F10 melanoma by activating potent immune responses and directly destroying the cancer cells. The stable, non-toxic, and efficacious properties of N. caninum demonstrate the potential for its use as a cancer treatment. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05456-8.
Collapse
|
9
|
Chen J, Liao W, Peng H. Toxoplasma gondii infection possibly reverses host immunosuppression to restrain tumor growth. Front Cell Infect Microbiol 2022; 12:959300. [PMID: 36118042 PMCID: PMC9470863 DOI: 10.3389/fcimb.2022.959300] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor cells can successfully escape the host immune attack by inducing the production of immunosuppressive cells and molecules, leading to an ineffective tumor treatment and poor prognosis. Although immunotherapies have improved the survival rate of cancer patients in recent years, more effective drugs and therapies still need to be developed. As an intracellular parasite, Toxoplasma gondii can trigger a strong Th1 immune response in host cells, including upregulating the expression of interleukin-12 (IL-12) and interferon-γ (IFN-γ). Non-replicating uracil auxotrophic strains of T. gondii were used to safely reverse the immunosuppression manipulated by the tumor microenvironment. In addition to the whole lysate antigens, T. gondii-secreted effectors, including Toxoplasma profilin, rhoptry proteins (ROPs), and dense granule antigens (GRAs), are involved in arousing the host’s antigen presentation system to suppress tumors. When T. gondii infection relieves immunosuppression, tumor-related myeloid cells, including macrophages and dendritic cells (DCs), are transformed into immunostimulatory phenotypes, showing a powerful Th1 immune response mediated by CD8+ T cells. Afterwards, they target and kill the tumor cells, and ultimately reduce the size and weight of tumor tissues. This article reviews the latest applications of T. gondii in tumor therapy, including the activation of cellular immunity and the related signal pathways, which will help us understand why T. gondii infection can restrain tumor growth.
Collapse
Affiliation(s)
- Jiating Chen
- Department of Pathogen Biology, School of Public Health, Guangdong Provincial Key laboratory of Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Wenzhong Liao
- Department of Pathogen Biology, School of Public Health, Guangdong Provincial Key laboratory of Tropical Medicine, Southern Medical University, Guangzhou, China
| | - HongJuan Peng
- Department of Pathogen Biology, School of Public Health, Guangdong Provincial Key laboratory of Tropical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Anti-Tumor Effect of Parasitic Protozoans. Bioengineering (Basel) 2022; 9:bioengineering9080395. [PMID: 36004920 PMCID: PMC9405343 DOI: 10.3390/bioengineering9080395] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/12/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022] Open
Abstract
The immune system may aberrantly silence when against “altered self”, which consequently may develop into malignancies. With the development of tumor immunology and molecular biology, the deepened understanding of the relationship between parasites and tumors shifts the attitude towards parasitic pathogens from elimination to utilization. In recent years, the antitumor impact implemented by protozoan parasites and the derived products has been confirmed. The immune system is activated and enhanced by some protozoan parasites, thereby inhibiting tumor growth, angiogenesis, and metastasis in many animal models. In this work, we reviewed the available information on the antitumor effect of parasitic infection or induced by parasitic antigen, as well as the involved immune mechanisms that modulate cancer progression. Despite the fact that clinical trials of the protozoan parasites against tumors are limited and the specific mechanisms of the effect on tumors are not totally clear, the use of genetically modified protozoan parasites and derived molecules combined with chemotherapy could be an important element for promoting antitumor treatment in the future.
Collapse
|
11
|
Evaluation of cytotoxic activity of live toxoplasma gondii tachyzoites and toxoplasma antigen on MCF-7 human breast cancer cell line. EUREKA: LIFE SCIENCES 2022. [DOI: 10.21303/2504-5695.2022.002409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to investigate the cytotoxic potency of live Toxoplasma gondii tachyzoites as well as Toxoplasma antigen on MCF-7 human breast cancer cell line. Cancer cell lines are considered an essential preliminary step towards in-vitro investigation of the potential antineoplastic impact of novel chemotherapeutic agents. Pathogens, including viruses, bacteria, and parasites are noticeably under investigation, considering their potential antineoplastic activity. Some have attained a steady position in the clinical field as hepatitis B virus, human papilloma virus and BCG immunization. Toxoplasma gondii is an apicomplexan parasite with promising antineoplastic activity. In this study, live Toxoplasma tachyzoites provoked a direct cytotoxic effect on MCF-7 in a dose dependent manner, while Toxoplasma antigen didn’t induce such impact.
Skipping the direct cytotoxic effect of Toxoplasma antigen doesn’t totally divert the possible antineoplastic activity of Toxoplasma antigen. Potential alternative immune mediated mechanisms could be an alternative. Further in-vivo studies in different cancer models are mandatory to investigate the underlying mechanisms of antineoplastic activity of Toxoplasma gondii
Collapse
|
12
|
Wang L, Wang N, Zhao Y, Lu G. Toxoplasma gondii causes changes in the host's expression of cancer‑associated miRNAs. Oncol Lett 2022; 23:149. [PMID: 35350589 PMCID: PMC8941548 DOI: 10.3892/ol.2022.13267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/21/2022] [Indexed: 11/16/2022] Open
Abstract
Throughout the world, numerous individuals are infected with Toxoplasma gondii, which may improve immunity against cancer. Furthermore, microRNAs (miRs) may be differentially expressed in the host upon infection with T. gondii. In the present study, RNA-sequencing analysis and reverse transcription-quantitative PCR revealed that miR-429-3p, miR-145a-5p, miR-211-5p, miR-31-3p and miR-135a-5p were determined to be downregulated, while miR-21a-3p, miR-135b-5p, miR-210-5p and miR-146-3p were upregulated in mice post-infection with T. gondii. Antitumor genes [TNF receptor superfamily member 11b, large tumor suppressor kinase (Lats)2 and Lats1] were identified as targets of miR-429-3p, miR-145a-5p, miR-211-5p, miR-31-3p and miR-135a-5p with a luciferase reporter assay. In addition, the protein levels of Lats2 and Lats1 were detected to be higher in T. gondii-infected mice than in the control group. Therefore, these results provide favorable evidence for the suppression of cancer upon T. gondii infection and may give novel ideas for the treatment of tumors.
Collapse
Affiliation(s)
- Lin Wang
- Department of Epilepsy Center, Ji Nan Children's Hospital, Jinan, Shandong 250022, P.R. China
| | - Ning Wang
- Department of Clinical Laboratory, Qingdao Third People's Hospital, Qingdao, Shandong 266041, P.R. China
| | - Ying Zhao
- Department of Pathogen Biology, School of Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Gang Lu
- Department of Pathogen Biology, School of Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| |
Collapse
|
13
|
Divyashree M, Prakash SK, Aditya V, Aljabali AA, Alzahrani KJ, Azevedo V, Góes-Neto A, Tambuwala MM, Barh D. Bugs as drugs: neglected but a promising future therapeutic strategy in cancer. Future Oncol 2022; 18:1609-1626. [PMID: 35137604 DOI: 10.2217/fon-2021-1137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Effective cancer treatment is an urgent need due to the rising incidence of cancer. One of the most promising future strategies in cancer treatment is using microorganisms as cancer indicators, prophylactic agents, immune activators, vaccines or vectors in antitumor therapy. The success of bacteria-mediated chemotherapy will be dependent on the balance of therapeutic benefit and the control of bacterial infection in the body. Additionally, protozoans and viruses have the potential to be used in cancer therapy. This review summarizes how these microorganisms interact with tumor microenvironments and the challenges of a 'bugs as drugs' approach in cancer therapy. Several standpoints are discussed, such as bacteria as vectors for gene therapy that shuttle therapeutic compounds into tumor tissues, their intrinsic antitumor activities and their combination with chemotherapy or radiotherapy. Bug-based cancer therapy is a two-edged sword and we need to find the opportunities by overcoming the challenges.
Collapse
Affiliation(s)
- Mithoor Divyashree
- Nitte University Centre for Science Education & Research (NUCSER), NITTE (Deemed to be University), Paneer Campus, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Shama K Prakash
- K. S. Hegde Medical Academy, NITTE (Deemed to be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Vankadari Aditya
- Nitte University Centre for Science Education & Research (NUCSER), NITTE (Deemed to be University), Paneer Campus, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Alaa Aa Aljabali
- Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University-Faculty of Pharmacy, Irbid, 566, Jordan
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, 21944, Saudi Arabia
| | - Vasco Azevedo
- Department of Genetics, Laboratory of Cellular & Molecular Genetics, Ecology & Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP, 31270-901, Brazil
| | - Aristóteles Góes-Neto
- Department of Microbiology, Molecular & Computational Biology of Fungi Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP, 31270-901, Brazil
| | - Murtaza M Tambuwala
- School of Pharmacy & Pharmaceutical Science, Ulster University, Coleraine, Northern Ireland, BT52 1SA, UK
| | - Debmalya Barh
- Department of Genetics, Laboratory of Cellular & Molecular Genetics, Ecology & Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP, 31270-901, Brazil.,Institute of Integrative Omics & Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur WB, 721172, India
| |
Collapse
|
14
|
Lu X, Zhang J, Li YQ, Liu QX, Zhou D, Deng XF, Qiu Y, Chen Q, Li MY, Liu XQ, Dai JG, Zheng H. Plasmodium Circumsporozoite Protein Enhances the Efficacy of Gefitinib in Lung Adenocarcinoma Cells by Inhibiting Autophagy via Proteasomal Degradation of LC3B. Front Cell Dev Biol 2022; 10:830046. [PMID: 35186935 PMCID: PMC8851824 DOI: 10.3389/fcell.2022.830046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/13/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Almost all lung adenocarcinoma (LUAD) patients with EGFR mutant will develop resistance to EGFR-TKIs, which limit the long-term clinical application of these agents. Accumulating evidence shows one of the main reasons for resistance to EGFR-TKIs is induction of autophagy in tumor cells. Our previous study found that circumsporozoite protein (CSP) in Plasmodium can suppress autophagy in host hepatocytes. However, it is unknown whether CSP-mediated inhibition of autophagy could improve the anti-tumor effect of EGFR-TKIs. Methods: We constructed A549 and H1975 cell lines with stable overexpression of CSP (OE-CSP cells). CCK-8, Lactate Dehydrogenase (LDH), flow cytometry, and colony analysis were performed to observe the effect of CSP overexpression on cell viability, apoptosis rate, and colony formation ratio. The sensitizing effect of CSP on gefitinib was evaluated in vivo using a subcutaneous tumor model in nude mice and immunohistochemical assay. The role of CSP in regulation of autophagy was investigated by laser confocal microscopy assay and western blotting. A transcriptome sequencing assay and real-time polymerase chain reaction were used to determine the levels of mRNA for autophagy-related proteins. Cycloheximide (CHX), MG132, TAK-243, and immunoprecipitation assays were used to detect and confirm proteasomal degradation of LC3B. Results: OE-CSP A549 and H1975 cells were more sensitive to gefitinib, demonstrating significant amounts of apoptosis and decreased viability. In the OE-CSP group, autophagy was significantly inhibited, and there was a decrease in LC3B protein after exposure to gefitinib. Cell viability and colony formed ability were recovered when OE-CSP cells were exposed to rapamycin. In nude mice with xenografts of LUAD cells, inhibition of autophagy by CSP resulted in suppression of cell growth, and more marked apoptosis during exposure to gefitinib. CSP promoted ubiquitin-proteasome degradation of LC3B, leading to inhibition of autophagy in LUAD cells after treatment with gefitinib. When LUAD cells were treated with ubiquitin activating enzyme inhibitor TAK-243, cell viability, apoptosis, and growth were comparable between the OE-CSP group and a control group both in vivo and in vitro. Conclusion: CSP can inhibit gefitinib-induced autophagy via proteasomal degradation of LC3B, which suggests that CSP could be used as an autophagy inhibitor to sensitize EGFR-TKIs.
Collapse
Affiliation(s)
- Xiao Lu
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jiao Zhang
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yan-Qi Li
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Quan-Xing Liu
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Dong Zhou
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xu-Feng Deng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qian Chen
- Cancer Center of Daping Hospital, Army Medical University, Chongqing, China
| | - Man-Yuan Li
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiao-Qing Liu
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ji-Gang Dai
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
15
|
Xu LQ, Yao LJ, Jiang D, Zhou LJ, Chen M, Liao WZ, Zou WH, Peng HJ. A uracil auxotroph Toxoplasma gondii exerting immunomodulation to inhibit breast cancer growth and metastasis. Parasit Vectors 2021; 14:601. [PMID: 34895326 PMCID: PMC8665513 DOI: 10.1186/s13071-021-05032-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Breast cancer is the most common cause of cancer-related death among women, and prognosis is especially poor for patients with triple-negative breast cancer (TNBC); therefore, there is an urgent need for new effective therapies. Recent studies have demonstrated that the uracil auxotroph Toxoplasma gondii vaccine displays anti-tumor effects. Here, we examined the immunotherapy effects of an attenuated uracil auxotroph strain of T. gondii against 4T1 murine breast cancer. METHODS We constructed a uracil auxotroph T. gondii RH strain via orotidine 5'-monophosphate decarboxylase gene deletion (RH-Δompdc) with CRISPR/Cas9 technology. The strain's virulence in the T. gondii-infected mice was determined in vitro and in vivo by parasite replication assay, plaque assay, parasite burden detection in mice peritoneal fluids and survival analysis. The immunomodulation ability of the strain was evaluated by cytokine detection. Its anti-tumor effect was evaluated after its in situ inoculation into 4T1 tumors in a mouse model; the tumor volume was measured, and the 4T1 lung metastasis was detected by hematoxylin and eosin and Ki67 antibody staining, and the cytokine levels were measured by an enzyme-linked immunosorbent assay. RESULTS The RH-Δompdc strain proliferated normally when supplemented with uracil, but it was unable to propagate without the addition of uracil and in vivo, which suggested that it was avirulent to the hosts. This mutant showed vaccine characteristics that could induce intense immune responses both in vitro and in vivo by significantly boosting the expression of inflammatory cytokines. Inoculation of RH-Δompdc in situ into the 4T1 tumor inhibited tumor growth, reduced lung metastasis, promoted the survival of the tumor-bearing mice and increased the secretion of Th1 cytokines, including interleukin-12 (IL-12) and interferon-γ (INF-δ), in both the serum and tumor microenvironment (TME). CONCLUSION Inoculation of the uracil auxotroph RH-Δompdc directly into the 4T1 tumor stimulated anti-infection and anti-tumor immunity in mice, and resulted in inhibition of tumor growth and metastasis, promotion of the survival of the tumor-bearing mice and increased secretion of IL-12 and IFN-γ in both the serum and TME. Our findings suggest that the immunomodulation caused by RH-Δompdc could be a potential anti-tumor strategy.
Collapse
Affiliation(s)
- Li-Qing Xu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Li-Jie Yao
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Dan Jiang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Li-Juan Zhou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Min Chen
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Wen-Zhong Liao
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Wei-Hao Zou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Hong-Juan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China.
| |
Collapse
|
16
|
Novel Murine Pancreatic Tumor Model Demonstrates Immunotherapeutic Control of Tumor Progression by a Toxoplasma gondii Protein. Infect Immun 2021; 89:e0050821. [PMID: 34543124 DOI: 10.1128/iai.00508-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is the fourth leading cause of cancer-related death in the United States, with few effective treatments available and only 10% of those diagnosed surviving 5 years. Although immunotherapeutics is a growing field of study in cancer biology, there has been little progress in its use for the treatment of pancreatic cancer. Pancreatic cancer is considered a nonimmunogenic tumor because the tumor microenvironment does not easily allow for the immune system, even when stimulated, to attack the cancer. Infection with the protozoan parasite Toxoplasma gondii has been shown to enhance the immune response to clear cancer tumors. A subset of T. gondii proteins called soluble Toxoplasma antigen (STAg) contains an immunodominant protein called profilin. Both STAg and profilin have been shown to stimulate an immune response that reduces viral, bacterial, and parasitic burdens. Here, we use STAg and profilin to treat pancreatic cancer in a KPC mouse-derived allograft murine model. These mice exhibit pancreatic cancer with both Kras and P53 mutations as subcutaneous tumors. Pancreatic cancer tumors in C57BL/6J mice with a wild-type background showed a significant response to treatment with either profilin or STAg, exhibiting a decrease in tumor volume accompanied by an influx of CD4+ and CD8+ T cells into the tumors. Both IFN-γ-/- mice and Batf3-/- mice, which lack conventional dendritic cells, failed to show significant decreases in tumor volumes when treated. These results indicate that gamma interferon (IFN-γ) and dendritic cells may play critical roles in the immune response necessary to treat pancreatic cancer.
Collapse
|
17
|
Zhu YC, Elsheikha HM, Wang JH, Fang S, He JJ, Zhu XQ, Chen J. Synergy between Toxoplasma gondii type I Δ GRA17 immunotherapy and PD-L1 checkpoint inhibition triggers the regression of targeted and distal tumors. J Immunother Cancer 2021; 9:jitc-2021-002970. [PMID: 34725213 PMCID: PMC8562526 DOI: 10.1136/jitc-2021-002970] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 12/28/2022] Open
Abstract
Background In this study, we hypothesize that the ability of the protozoan Toxoplasma gondii to modulate immune response within the tumor might improve the therapeutic effect of immune checkpoint blockade. We examined the synergetic therapeutic activity of attenuated T. gondii RH ΔGRA17 strain and programmed death ligand-1 (PD-L1) treatment on both targeted and distal tumors in mice. Methods The effects of administration of T. gondii RH ΔGRA17 strain on the tumor volume and survival rate of mice bearing flank B16-F10, MC38, or LLC tumors were studied. We characterized the effects of ΔGRA17 on tumor biomarkers’ expression, PD-L1 expression, immune cells infiltrating the tumors, and expression of immune-related genes by using immunohistochemistry, immunofluorescence, flow cytometry, NanoString platform, and real-time quantitative PCR, respectively. The role of immune cells in the efficacy of ΔGRA17 plus PD-L1 blockade therapy was determined via depletion of immune cell subtypes. Results Treatment with T. gondii ΔGRA17 tachyzoites and anti-PD-L1 therapy significantly extended the survival of mice and suppressed tumor growth in preclinical mouse models of melanoma, Lewis lung carcinoma, and colon adenocarcinoma. Attenuation of the tumor growth was detected in the injected and distant tumors, which was associated with upregulation of innate and adaptive immune pathways. Complete regression of tumors was underpinned by late interferon-gamma-producing CD8+ cytotoxic T cells. Conclusion The results from these models indicate that intratumoral injection of ΔGRA17 induced a systemic effect, improved mouse immune response, and sensitized immunologically ‘cold’ tumors and rendered them sensitive to immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Yu-Chao Zhu
- Department of Radiology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang, China.,Immunology Innovation Team, Ningbo University School of Medicine, Ningbo, Zhejiang, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Jian-Hua Wang
- Department of Radiology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang, China.,Immunology Innovation Team, Ningbo University School of Medicine, Ningbo, Zhejiang, China
| | - Shuai Fang
- Department of Radiology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang, China.,Immunology Innovation Team, Ningbo University School of Medicine, Ningbo, Zhejiang, China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China .,Key Laboratory of Veterinary Public Health of Higher Education of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Jia Chen
- Department of Radiology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang, China .,Immunology Innovation Team, Ningbo University School of Medicine, Ningbo, Zhejiang, China
| |
Collapse
|
18
|
Saraav I, Cervantes-Barragan L, Olias P, Fu Y, Wang Q, Wang L, Wang Y, Mack M, Baldridge MT, Stappenbeck T, Colonna M, Sibley LD. Chronic Toxoplasma gondii infection enhances susceptibility to colitis. Proc Natl Acad Sci U S A 2021; 118:e2106730118. [PMID: 34462359 PMCID: PMC8433586 DOI: 10.1073/pnas.2106730118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Oral infection with Toxoplasma gondii results in dysbiosis and enteritis, both of which revert to normal during chronic infection. However, whether infection leaves a lasting impact on mucosal responses remains uncertain. Here we examined the effect of the chemical irritant dextran sodium sulfate (DSS) on intestinal damage and wound healing in chronically infected mice. Our findings indicate that prior infection with T. gondii exacerbates damage to the colon caused by DSS and impairs wound healing by suppressing stem cell regeneration of the epithelium. Enhanced tissue damage was attributable to inflammatory monocytes that emerge preactivated from bone marrow, migrate to the intestine, and release inflammatory mediators, including nitric oxide. Tissue damage was reversed by neutralization of inflammatory monocytes or nitric oxide, revealing a causal mechanism for tissue damage. Our findings suggest that chronic infection with T. gondii enhances monocyte activation to increase inflammation associated with a secondary environmental insult.
Collapse
Affiliation(s)
- Iti Saraav
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Luisa Cervantes-Barragan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Philipp Olias
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Yong Fu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Qiuling Wang
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Leran Wang
- Department of Medicine, Division of Infectious Diseases, Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Yi Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Matthias Mack
- Department of Nephrology, University of Regensburg, 93042 Regensburg, Germany
| | - Megan T Baldridge
- Department of Medicine, Division of Infectious Diseases, Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Thaddeus Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110;
| |
Collapse
|
19
|
Toxoplasma gondii could have a possible role in the cancer mechanism by modulating the host's cell response. Acta Trop 2021; 220:105966. [PMID: 34023305 DOI: 10.1016/j.actatropica.2021.105966] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 12/15/2022]
Abstract
Toxoplasma gondii, which manipulates many signaling pathways to achieve persistence in host cells, is intimately linked to immune and inflammation responses. However, there is still lack of information about the impact of T. gondii on cellular and immune responses. This study was designed to seek the impact of T. gondii infection causing life-long inflammation in brain, on cancer mechanism. To identify molecular effects of the T. gondii and understand the association between the functional perturbations occurring during infection and cancer development, the transcriptomic datasets obtained mice infected with T. gondii were downloaded from GEO. The differentially expressed genes (DEGs) were identified and functional enrichment analysis was performed using IPA platform, then all results were evaluated with comparison analyses. Subsequently, a T. gondii infection model with human neuroepithelioma cell culture was performed in order to validate top DEGs participated in common networks/pathways in cancer mechanism. Transcriptomic analyses of infected mice and in vitro cell culture model revealed a strong immune response and inflammation occurred by parasite-induced damage and parasite-associated immunopathology in host cell and tissue. T. gondii infection could modulate certain signaling pathways of host, which were also common to those perturbed in carcinogenesis. Interestingly, the network analysis of the data sets predicted an activation in development of solid cancer vice versa inhibition in hematological cancer during T. gondii infection. Parasite might also control the tumor growth due to its potent immune-stimulant effects. As result, T. gondii infection generating a continual inflammation in tissues might potentially contribute to cancer development by regulating critical host signaling pathways or reveal an anti-tumoral activity.
Collapse
|
20
|
Hafez EN, Moawed FSM, Abdel-Hamid GR, Elbakary NM. Gamma Radiation-Attenuated Toxoplasma gondii Provokes Apoptosis in Ehrlich Ascites Carcinoma-Bearing Mice Generating Long-Lasting Immunity. Technol Cancer Res Treat 2021; 19:1533033820926593. [PMID: 32567499 PMCID: PMC7309383 DOI: 10.1177/1533033820926593] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose: Pathological angiogenesis and apoptosis evasions are common hallmarks of cancer. A different approach to the antitumor effect of parasitic diseases caused by certain protozoans and helminthes had been adopted in recent years as they can affect many cancer characteristics. The present work is an attempt to assess the effect of gamma radiation-attenuated Toxoplasma gondii ME49 as an antiapoptotic and angiogenic regulator modifier on tumor growth aimed at improving cancer protective protocols. Methods: Attenuated Toxoplasma gondii ME49 was administered orally to mice 2 weeks before inoculation with Ehrlich ascites carcinoma to allow stimulation of the immune response. Hepatic histopathology and immune responses were determined for each group. Results: Marked suppression of the tumor proliferation with induction of long-lasting immunity by stimulating interferon γ and downregulating transforming growth factor β. The level of tumor promoting inflammatory markers (STAT-3 and tumor necrosis factor α), the angiogenic factors (vascular endothelial growth factor A, integrin, and matrix metallopeptidase 2 and matrix metallopeptidase 9), as well as nitric oxide concentration were significantly decreased. This was collimated with an improvement in apoptotic regulators (cytochrome-c, Bax, Bak, and caspase 3) in liver tissues of vaccinated mice group compared to Ehrlich ascites carcinoma-bearing one. Moreover, the histopathological investigations confirmed this improvement. Conclusion: Hence, there is an evidence of potency of radiation attenuated Toxoplasma vaccine in immune activation and targeting tumor cell that can be used as a prophylactic or an adjuvant in combination with chemotherapeutic drugs.
Collapse
Affiliation(s)
- Eman N Hafez
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT)-Atomic Energy Authority (AEA), Cairo, Egypt
| | - Fatma S M Moawed
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT)-Atomic Energy Authority (AEA), Cairo, Egypt
| | - Gehan R Abdel-Hamid
- Radiation Biology Department, National Center for Radiation Research and Technology, Atomic Energy authority, Cairo, Egypt
| | - Nermeen M Elbakary
- Radiation Biology Department, National Center for Radiation Research and Technology, Atomic Energy authority, Cairo, Egypt
| |
Collapse
|
21
|
Caner A, Sadıqova A, Erdoğan A, Namlıses D, Nalbantsoy A, Oltulu F, Toz S, Yiğittürk G, Ozkök E, Gunduz C, Ozbel Y, Haydaroğlu A. Targeting of antitumor ımmune responses with live-attenuated Leishmania strains in breast cancer model. Breast Cancer 2020; 27:1082-1095. [PMID: 32472473 DOI: 10.1007/s12282-020-01112-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/16/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cancer is a major cause of death worldwide and most of the therapeutic approaches are relatively ineffective in eliminating cancer especially due to drug resistance. As an alternative, therapy with live microorganisms can induce a robust proinflammatory and anti-cancer immune response in the microenvironment of the tumor. In the present study, we aimed to establish a model for taking the advantages of immune responses against intracellular protozoan parasites for cancer treatment. METHODS Leishmania infantum and L. tropica were used in our study as agents of visceral and cutaneous forms of the infection, respectively. After establishing 4T1 breast cancer in mice groups, live-attenuated L. infantum (At-Li) and live-attenuated L. tropica (At-Lt) treatments were performed and results were evaluated according to tumor volume, immune markers and histological examination. RESULTS Live-attenuated Leishmania strains regressed 4T1-breast cancer in mice and are nonpathogenic, and these strains induce an immune response against 4T1 breast cancer. It is shown that At-Lt is found to be more effective than At-Li in breast cancer treatment using different methods included in the study as analyses of immune parameters, and histopathological examination in tumor tissue besides spleen cells. The tumor grew more slowly by the immune-stimulant effect of live-attenuated Leishmania parasites. CONCLUSION This promising therapy should be investigated for optimization in further studies with different cancer types and L. tropica may be designed to express antigens to enhance tumor antigen-specific responses, which may further improve efficacy and immune memory development.
Collapse
Affiliation(s)
- Ayse Caner
- Departments of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey. .,Department of Parasitology, Ege University Medical School, Izmir, Turkey.
| | - Aygül Sadıqova
- Infectious Disease Division, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Alper Erdoğan
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Dünya Namlıses
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Ayse Nalbantsoy
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Turkey
| | - Fatih Oltulu
- Department of Histology and Embryology, Ege University Medical School, Izmir, Turkey
| | - Seray Toz
- Department of Parasitology, Ege University Medical School, Izmir, Turkey
| | - Gürkan Yiğittürk
- Department of Histology and Embryology, Ege University Medical School, Izmir, Turkey
| | - Emel Ozkök
- Department of Pathology, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Cumhur Gunduz
- Department of Medical Biology, Ege University Medical School, Izmir, Turkey
| | - Yusuf Ozbel
- Department of Parasitology, Ege University Medical School, Izmir, Turkey
| | - Ayfer Haydaroğlu
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| |
Collapse
|
22
|
Hafez EN, Youssef HMG, El-Kabany HA. Vaccination with gamma radiation-attenuated Toxoplasma gondii protects against ovarian infiltration in mice-bearing Ehrlich ascites carcinoma. Int J Radiat Biol 2020; 96:814-822. [PMID: 32149560 DOI: 10.1080/09553002.2020.1739772] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Purpose: Cancer is one of the most common causes of mortality and morbidity worldwide. Vaccines have been emerged as an attractive approach for their capacity of eliciting long-term immune response targeting cancer cells. Attenuated avirulent Toxoplasma gondii stimulate immunity and activate antitumor cells thereby eliciting rejection of some established cancer. The purpose of this study was to evaluate the antitumor-protective capacity of vaccination with gamma radiation-attenuated T. gondii against ovarian penetration in Ehrlich ascites carcinoma (EAC)-bearing mice.Materials and methods: Forty-five mice were randomly divided into three groups as follows: nontumor-bearing (normal control); EAC-bearing group (EAC); and mice vaccinated orally with gamma radiation-attenuated T. gondii then inoculated 2 weeks later with EAC (TG + EAC). Survival rate, serum interleukin-12 (IL-12), and levels of IFN-γ mRNA, CD4, and CD8 in ovarian tissues homogenate were assessed. Also, ovarian histopathology and immunohistochemical expressions of metalloproteinase-2, CD34, and vimentin were determined.Results: The group vaccinated with attenuated T. gondii showed significantly increased survival rates, serum IL-12, and levels of IFN-γ, CD4, and CD8 in ovarian tissue homogenates as well as an enhancement of histopathological and immunohistochemical changes compared to EAC-bearing group.Conclusion: Vaccination with gamma radiation-attenuated T. gondii has the capacity to supply immunoprotective impact against ovarian invasion in EAC-bearing mice.
Collapse
Affiliation(s)
- Eman N Hafez
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Hanan M G Youssef
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Hanan A El-Kabany
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
23
|
Rommereim LM, Fox BA, Butler KL, Cantillana V, Taylor GA, Bzik DJ. Rhoptry and Dense Granule Secreted Effectors Regulate CD8 + T Cell Recognition of Toxoplasma gondii Infected Host Cells. Front Immunol 2019; 10:2104. [PMID: 31555296 PMCID: PMC6742963 DOI: 10.3389/fimmu.2019.02104] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/21/2019] [Indexed: 12/21/2022] Open
Abstract
Toxoplasma gondii secretes rhoptry (ROP) and dense granule (GRA) effector proteins to evade host immune clearance mediated by interferon gamma (IFN-γ), immunity-related GTPase (IRG) effectors, and CD8+ T cells. Here, we investigated the role of parasite-secreted effectors in regulating host access to parasitophorous vacuole (PV) localized parasite antigens and their presentation to CD8+ T cells by the major histocompatibility class I (MHC-I) pathway. Antigen presentation of PV localized parasite antigens by MHC-I was significantly increased in macrophages and/or dendritic cells infected with mutant parasites that lacked expression of secreted GRA (GRA2, GRA3, GRA4, GRA5, GRA7, GRA12) or ROP (ROP5, ROP18) effectors. The ability of various secreted GRA or ROP effectors to suppress antigen presentation by MHC-I was dependent on cell type, expression of IFN-γ, or host IRG effectors. The suppression of antigen presentation by ROP5, ROP18, and GRA7 correlated with a role for these molecules in preventing PV disruption by IFN-γ-activated host IRG effectors. However, GRA2 mediated suppression of antigen presentation was not correlated with PV disruption. In addition, the GRA2 antigen presentation phenotypes were strictly co-dependent on the expression of the GRA6 protein. These results show that MHC-I antigen presentation of PV localized parasite antigens was controlled by mechanisms that were dependent or independent of IRG effector mediated PV disruption. Our findings suggest that the GRA6 protein underpins an important mechanism that enhances CD8+ T cell recognition of parasite-infected cells with damaged or ruptured PV membranes. However, in intact PVs, parasite secreted effector proteins that associate with the PV membrane or the intravacuolar network membranes play important roles to actively suppress antigen presentation by MHC-I to reduce CD8+ T cell recognition and clearance of Toxoplasma gondii infected host cells.
Collapse
Affiliation(s)
- Leah M Rommereim
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Barbara A Fox
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Kiah L Butler
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Viviana Cantillana
- Division of Geriatrics, Departments of Medicine, Molecular Genetics and Microbiology, and Immunology, Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC, United States
| | - Gregory A Taylor
- Division of Geriatrics, Departments of Medicine, Molecular Genetics and Microbiology, and Immunology, Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC, United States.,Geriatric Research, Education and Clinical Center, VA Medical Center, Durham, NC, United States
| | - David J Bzik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
24
|
Jazeela K, Chakraborty A, Karunasagar I, Deekshit VK. Nontyphoidal Salmonella: a potential anticancer agent. J Appl Microbiol 2019; 128:2-14. [PMID: 31038778 DOI: 10.1111/jam.14297] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 02/06/2023]
Abstract
Use of bacteria in cancer therapy, despite being considered as a potent strategy, has not really picked up the way other methods of cancer therapies have evolved. However, in recent years, the interest on use of bacteria to kill cancer cells has renewed considerably. The standard and widely followed strategies of cancer treatment often fail either due to the complexity of tumour biology or because of the accompanying side effects. In contrast, these limitations can be easily overcome in a bacteria-mediated approach. Salmonella is a bacterium, which is known for its ability to colonize solid or semisolid tumours more efficiently than any other bacteria. Among more than 2500 serovars of Salmonella, S. Typhimurium has been widely studied for its antagonistic effects on cancer cells. Here in, we review the current status of the preclinical and the clinical studies with a focus on the mechanisms that attribute the anticancer properties to nontyphoidal Salmonella.
Collapse
Affiliation(s)
- K Jazeela
- Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| | - A Chakraborty
- Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| | - I Karunasagar
- Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| | - V K Deekshit
- Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Deralakatte, Mangaluru, Karnataka, India
| |
Collapse
|
25
|
Lu G, Zhou J, Zhao YH, Li QL, Gao YY, Wang L. Transcriptome Sequencing Investigated the Tumor-Related Factors Changes After T. gondii Infection. Front Microbiol 2019; 10:181. [PMID: 30792708 PMCID: PMC6374557 DOI: 10.3389/fmicb.2019.00181] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 01/23/2019] [Indexed: 12/13/2022] Open
Abstract
Toxoplasma gondii is an intracellular parasite and causes a global epidemic parasitic disease. T. gondii-infection could inhibit the growth of tumor. In this study, the transcriptomes of samples were detected by deep sequencing analysis. The transcriptome data was compared with reference genome to perform sequence alignment and the further analysis. The analyses of differential expression and the differentially expressed genes were performed in the present study. Genes involved in P53 signaling pathway, COLORECTAL cancer pathway, NON-SMALL CELL LUNG cancer signaling pathway, and BREAST cancer signaling pathway were up-regulated or down-regulated among the samples. The KEGG analysis indicated that the cancer pathways changed after infection of T. gondii. Furthermore, tumor-related mRNAs from different samples had a large difference, which suggested that the difference might provide important information in resisting cancer. The protein results indicated that tumor-related protein changes occurred after infection of T. gondii. In conclusion, the infection changed the cancer pathways, which could possibly inhibit the growth of tumor.
Collapse
Affiliation(s)
- Gang Lu
- Institute of Pathogen Biology, Taishan Medical College, Tai'an, China
| | - Jian Zhou
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Sports Medicine Research Center, Central South University, Changsha, China
| | - Ying Hui Zhao
- Institute of Pathogen Biology, Taishan Medical College, Tai'an, China
| | - Qiao Ling Li
- Institute of Pathogen Biology, Taishan Medical College, Tai'an, China
| | - Yun Yun Gao
- Institute of Pathogen Biology, Taishan Medical College, Tai'an, China
| | - Lin Wang
- Department of Ji Nan Children's Hospital, Jinan, China
| |
Collapse
|
26
|
Microorganisms in the Treatment of Cancer: Advantages and Limitations. J Immunol Res 2018; 2018:2397808. [PMID: 29682586 PMCID: PMC5848056 DOI: 10.1155/2018/2397808] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 01/09/2018] [Indexed: 12/22/2022] Open
Abstract
Cancer remains one of the major challenges of the 21st century. The increasing numbers of cases are not accompanied by adequate progress in therapy. The standard methods of treatment often do not lead to the expected effects. Therefore, it is extremely important to find new, more effective treatments. One of the most promising research directions is immunotherapy, including the use of specific types of microorganisms. This type of treatment is expected to stimulate the immune system for the selective elimination of cancer cells. The research results seem to be promising and show the intensive activation of the immune response as a result of bacterial stimulation. In addition, it is possible to use microorganisms in many different ways, based on their specific properties, that is, toxin production, anaerobic lifestyle, or binding substances that can be delivered to a specific location (vectors). This paper provides an overview of selected microorganisms which are already in use or that are in the experimental phase. Just like any other therapy, the use of microbes for cancer treatment also has some disadvantages. Nevertheless, this kind of treatment can supplement conventional anticancer therapy, giving cancer patients a chance and hope of recovery.
Collapse
|
27
|
Cancer therapy in a microbial bottle: Uncorking the novel biology of the protozoan Toxoplasma gondii. PLoS Pathog 2017; 13:e1006523. [PMID: 28910406 PMCID: PMC5599061 DOI: 10.1371/journal.ppat.1006523] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
28
|
Ngô HM, Zhou Y, Lorenzi H, Wang K, Kim TK, Zhou Y, El Bissati K, Mui E, Fraczek L, Rajagopala SV, Roberts CW, Henriquez FL, Montpetit A, Blackwell JM, Jamieson SE, Wheeler K, Begeman IJ, Naranjo-Galvis C, Alliey-Rodriguez N, Davis RG, Soroceanu L, Cobbs C, Steindler DA, Boyer K, Noble AG, Swisher CN, Heydemann PT, Rabiah P, Withers S, Soteropoulos P, Hood L, McLeod R. Toxoplasma Modulates Signature Pathways of Human Epilepsy, Neurodegeneration & Cancer. Sci Rep 2017; 7:11496. [PMID: 28904337 PMCID: PMC5597608 DOI: 10.1038/s41598-017-10675-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 08/14/2017] [Indexed: 12/27/2022] Open
Abstract
One third of humans are infected lifelong with the brain-dwelling, protozoan parasite, Toxoplasma gondii. Approximately fifteen million of these have congenital toxoplasmosis. Although neurobehavioral disease is associated with seropositivity, causality is unproven. To better understand what this parasite does to human brains, we performed a comprehensive systems analysis of the infected brain: We identified susceptibility genes for congenital toxoplasmosis in our cohort of infected humans and found these genes are expressed in human brain. Transcriptomic and quantitative proteomic analyses of infected human, primary, neuronal stem and monocytic cells revealed effects on neurodevelopment and plasticity in neural, immune, and endocrine networks. These findings were supported by identification of protein and miRNA biomarkers in sera of ill children reflecting brain damage and T. gondii infection. These data were deconvoluted using three systems biology approaches: "Orbital-deconvolution" elucidated upstream, regulatory pathways interconnecting human susceptibility genes, biomarkers, proteomes, and transcriptomes. "Cluster-deconvolution" revealed visual protein-protein interaction clusters involved in processes affecting brain functions and circuitry, including lipid metabolism, leukocyte migration and olfaction. Finally, "disease-deconvolution" identified associations between the parasite-brain interactions and epilepsy, movement disorders, Alzheimer's disease, and cancer. This "reconstruction-deconvolution" logic provides templates of progenitor cells' potentiating effects, and components affecting human brain parasitism and diseases.
Collapse
Affiliation(s)
- Huân M Ngô
- The University of Chicago, Chicago, IL, 60637, USA.,Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA.,BrainMicro LLC, New Haven, CT, 06511, USA
| | - Ying Zhou
- The University of Chicago, Chicago, IL, 60637, USA
| | | | - Kai Wang
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Taek-Kyun Kim
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Yong Zhou
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | | | - Ernest Mui
- The University of Chicago, Chicago, IL, 60637, USA
| | | | | | | | - Fiona L Henriquez
- The University of Chicago, Chicago, IL, 60637, USA.,FLH, IBEHR School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - Alexandre Montpetit
- Genome Quebec, Montréal, QC H3B 1S6, Canada; McGill University, Montréal, QC H3A 0G4, Canada
| | - Jenefer M Blackwell
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, United Kingdom.,Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Sarra E Jamieson
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | | | | | | | | | | | | | - Charles Cobbs
- California Pacific Medical Center, San Francisco, CA, 94114, USA
| | - Dennis A Steindler
- JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| | - Kenneth Boyer
- Rush University Medical Center, Chicago, IL, 60612, USA
| | - A Gwendolyn Noble
- Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Charles N Swisher
- Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | | | - Peter Rabiah
- Northshore University Health System, Evanston, IL, 60201, USA
| | | | | | - Leroy Hood
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Rima McLeod
- The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
29
|
Fox BA, Sanders KL, Rommereim LM, Guevara RB, Bzik DJ. Secretion of Rhoptry and Dense Granule Effector Proteins by Nonreplicating Toxoplasma gondii Uracil Auxotrophs Controls the Development of Antitumor Immunity. PLoS Genet 2016; 12:e1006189. [PMID: 27447180 PMCID: PMC4957766 DOI: 10.1371/journal.pgen.1006189] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 06/22/2016] [Indexed: 12/19/2022] Open
Abstract
Nonreplicating type I uracil auxotrophic mutants of Toxoplasma gondii possess a potent ability to activate therapeutic immunity to established solid tumors by reversing immune suppression in the tumor microenvironment. Here we engineered targeted deletions of parasite secreted effector proteins using a genetically tractable Δku80 vaccine strain to show that the secretion of specific rhoptry (ROP) and dense granule (GRA) proteins by uracil auxotrophic mutants of T. gondii in conjunction with host cell invasion activates antitumor immunity through host responses involving CD8α+ dendritic cells, the IL-12/interferon-gamma (IFN-γ) TH1 axis, as well as CD4+ and CD8+ T cells. Deletion of parasitophorous vacuole membrane (PVM) associated proteins ROP5, ROP17, ROP18, ROP35 or ROP38, intravacuolar network associated dense granule proteins GRA2 or GRA12, and GRA24 which traffics past the PVM to the host cell nucleus severely abrogated the antitumor response. In contrast, deletion of other secreted effector molecules such as GRA15, GRA16, or ROP16 that manipulate host cell signaling and transcriptional pathways, or deletion of PVM associated ROP21 or GRA3 molecules did not affect the antitumor activity. Association of ROP18 with the PVM was found to be essential for the development of the antitumor responses. Surprisingly, the ROP18 kinase activity required for resistance to IFN-γ activated host innate immunity related GTPases and virulence was not essential for the antitumor response. These data show that PVM functions of parasite secreted effector molecules, including ROP18, manipulate host cell responses through ROP18 kinase virulence independent mechanisms to activate potent antitumor responses. Our results demonstrate that PVM associated rhoptry effector proteins secreted prior to host cell invasion and dense granule effector proteins localized to the intravacuolar network and host nucleus that are secreted after host cell invasion coordinately control the development of host immune responses that provide effective antitumor immunity against established ovarian cancer.
Collapse
Affiliation(s)
- Barbara A. Fox
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Kiah L. Sanders
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Leah M. Rommereim
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Rebekah B. Guevara
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - David J. Bzik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|