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
|
Jafari H, Mahami-Oskouei M, Spotin A, Baradaran B, Shanehbandi D, Baghbanzadeh A, Alizadeh Z. MicroRNA-1 Inhibits the Growth of Breast Cancer Cells MDA-MB-231 and MCF-7 Treated with Hydatid Cyst Fluid. J Trop Med 2024; 2024:7474039. [PMID: 38504949 PMCID: PMC10950417 DOI: 10.1155/2024/7474039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/21/2024] Open
Abstract
Antigens in hydatid cyst fluid (HCF) have been discovered to bear a significant resemblance to antigens present in cancer cells. MicroRNA-1 (miR-1) is a well-known member of the tumor inhibitor miRNA family and has been shown to have pro-apoptotic and tumor-inhibitory functions. This study aimed to evaluate the ability of HCF to prevent breast cancer and to explore the underlying mechanisms that affect cancer cells. For this study, MDA-MB-231 and MCF-7 breast cancer cells were cultured and divided into two groups: one group received HCF treatment and the other group was untreated and served as the control group. The cytotoxicity and cell viability of various HCF concentrations on breast cancer cells were evaluated using the MTT assay. In addition, the expression level of miR-1 in HCF-treated and untreated breast cancer cells was analyzed using qRT-PCR. The study found that HCF treatment reduced the growth of MDA-MB-231 and MCF-7 breast cancer cells, indicating that it was cytotoxic to the cells. Specifically, the IC50 concentration of HCF after 24 hours of treatment was 7.32 µg/mL for MDA-MB-231 cells and 13.63 µg/mL for MCF-7 cells. In addition, qRT-PCR analysis revealed that the expression level of miR-1 was significantly increased in HCF-treated MDA-MB-231 (P=0.0203) and MCF-7 (P=0.0394) cell lines compared to untreated controls. Although HCF has been shown to inhibit the growth of breast cancer cells and to upregulate miR-1, a key tumor suppressor in cancer cells, the specific mechanisms responsible for this effect remain unclear. Further studies are needed to fully understand the molecular pathways underlying HCF's antitumor activity and its potential as a therapeutic agent in cancer therapy.
Collapse
Affiliation(s)
- Hadis Jafari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahmoud Mahami-Oskouei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Adel Spotin
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Alizadeh
- Department of Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Baysal İ, Örsten S, Cengiz G, Ünal E, Doğrul AB, Çiftçi T, Çiftçi SY, Akinci D, Akhan O. Assessing the Potential Apoptotic Effects of Different Hydatid Cyst Fluids on Human Healthy Hepatocytes and Hepatocellular Carcinoma Cells. Acta Parasitol 2024; 69:700-709. [PMID: 38372909 PMCID: PMC11001659 DOI: 10.1007/s11686-024-00797-z] [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: 06/07/2023] [Accepted: 01/09/2024] [Indexed: 02/20/2024]
Abstract
Cystic Echinococcosis (CE) is a zoonotic infection caused by the larval form of Echinococcus granulosus in humans. Emerging evidence suggests an intriguing inverse association between E. granulosus infection and the occurrence of cancer. This study aimed to investigate the influence of diverse host-derived hydatid cyst fluids (HCF) with distinct genotypes on human liver hepatocytes (HC) and hepatocellular carcinoma cells (HepG2). Specifically, we examined their effects on cell proliferation, apoptosis sensitivity (BAX/BCL-2), apoptosis-related p53 expression, and the expression of cancer-related microRNA (hsa-miR-181b-3p). Cell proliferation assays, real-time PCR, and ELISA studies were conducted to evaluate potential anti-cancer properties. The findings revealed that animal-origin HCF (G1(A)) induced direct cell death by augmenting the susceptibility of HepG2 cells to apoptosis. Treatment with both G1(A) and G1(H) HCF sensitized HepG2 and HC cell lines to apoptosis by modulating the BAX/BCL-2 ratio, accompanied by upregulation of the p53 gene. Additionally, G1(A) HCF and human-derived HCFs (G1(H), G7(H)) reduced the expression of miR-181b-3p in HepG2 cells. Consequently, this study demonstrates the potential anti-cancer effect of HCF in HepG2 cells and provides the first comparative assessment of HCFs from human and animal sources with diverse genotypes, offering novel insights into this field.
Collapse
Affiliation(s)
- İpek Baysal
- Vocational School of Health Services, Hacettepe University, 06100, Ankara, Turkey.
| | - Serra Örsten
- Vocational School of Health Services, Hacettepe University, 06100, Ankara, Turkey
| | - Görkem Cengiz
- Vocational School of Health Services, Yüksek İhtisas University, 06291, Ankara, Turkey
| | - Emre Ünal
- Faculty of Medicine, Department of Radiology, Hacettepe University, 06100, Ankara, Turkey
| | - Ahmet Bülent Doğrul
- Faculty of Medicine, Department of General Surgery, Hacettepe University, 06100, Ankara, Turkey
| | - Türkmen Çiftçi
- Faculty of Medicine, Department of Radiology, Hacettepe University, 06100, Ankara, Turkey
| | - Samiye Yabanoğlu Çiftçi
- Faculty of Pharmacy, Department of Biochemistry, Hacettepe University, 06100, Ankara, Turkey
| | - Devrim Akinci
- Faculty of Medicine, Department of Radiology, Hacettepe University, 06100, Ankara, Turkey
| | - Okan Akhan
- Faculty of Medicine, Department of Radiology, Hacettepe University, 06100, Ankara, Turkey
| |
Collapse
|
4
|
Esperante D, Gutiérrez MIM, Issa ME, Schcolnik-Cabrera A, Mendlovic F. Similarities and divergences in the metabolism of immune cells in cancer and helminthic infections. Front Oncol 2023; 13:1251355. [PMID: 38044996 PMCID: PMC10690632 DOI: 10.3389/fonc.2023.1251355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/16/2023] [Indexed: 12/05/2023] Open
Abstract
Energetic and nutritional requirements play a crucial role in shaping the immune cells that infiltrate tumor and parasite infection sites. The dynamic interaction between immune cells and the microenvironment, whether in the context of tumor or helminth infection, is essential for understanding the mechanisms of immunological polarization and developing strategies to manipulate them in order to promote a functional and efficient immune response that could aid in the treatment of these conditions. In this review, we present an overview of the immune response triggered during tumorigenesis and establishment of helminth infections, highlighting the transition to chronicity in both cases. We discuss the energetic demands of immune cells under normal conditions and in the presence of tumors and helminths. Additionally, we compare the metabolic changes that occur in the tumor microenvironment and the infection site, emphasizing the alterations that are induced to redirect the immune response, thereby promoting the survival of cancer cells or helminths. This emerging discipline provides valuable insights into disease pathogenesis. We also provide examples of novel strategies to enhance immune activity by targeting metabolic pathways that shape immune phenotypes, with the aim of achieving positive outcomes in cancer and helminth infections.
Collapse
Affiliation(s)
- Diego Esperante
- Plan de Estudios Combinados en Medicina (PECEM), Facultad de Medicina, Universidad Nacional Autonóma de México (UNAM), Mexico City, Mexico
| | - Mónica Itzel Martínez Gutiérrez
- Plan de Estudios Combinados en Medicina (PECEM), Facultad de Medicina, Universidad Nacional Autonóma de México (UNAM), Mexico City, Mexico
| | - Mark E. Issa
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Alejandro Schcolnik-Cabrera
- Département de Biochimie et Médicine Moléculaire, Université de Montréal, Succursale Centre-Ville, Montréal, QC, Canada
- Department of Immunology-Oncology, Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC, Canada
| | - Fela Mendlovic
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Facultad de Ciencias de la Salud, Universidad Anáhuac México Norte, Huixquilucan, Mexico
| |
Collapse
|
5
|
Sadr S, Borji H. Echinococcus granulosus as a Promising Therapeutic Agent against Triplenegative Breast Cancer. CURRENT CANCER THERAPY REVIEWS 2023; 19:292-297. [DOI: 10.2174/1573394719666230427094247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/06/2022] [Accepted: 01/19/2023] [Indexed: 08/19/2024]
Abstract
Abstract:Breast cancer is a major cause of cancer deaths in women, with approximately 1.2 million new cases per year. Current treatment options for breast cancer include surgery, radiation, hormone therapy, and chemotherapy. However, the non-selective cytotoxicity of chemotherapeutic agents often leads to severe side effects, while drug resistance can worsen patient outcomes. Therefore, the development of more effective and less toxic anticancer drugs is a critical need. This study aimed to review the literature on Echinococcus granulosus antigens with anticancer potential against triple-negative breast cancer. Recent studies have suggested that certain parasite antigens may have potential anticancer effects. Specifically, research has shown that echinococcosis, a disease caused by the parasitic cestode Echinococcus granulosus, may have a protective effect against cancer. These findings offer new insights into the potential use of E. granulosus antigens in the development of novel cancer therapies and tumor cell vaccines. The findings of recent studies suggested that E. granulosus antigens may have the potential to be used in effective and less toxic cancer treatments. However, further research is needed to fully understand the mechanisms behind the anticancer effects of these antigens and develop new cancer therapies and vaccines
Collapse
Affiliation(s)
- Soheil Sadr
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hassan Borji
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
6
|
Chakraborty P, Aravindhan V, Mukherjee S. Helminth-derived biomacromolecules as therapeutic agents for treating inflammatory and infectious diseases: What lessons do we get from recent findings? Int J Biol Macromol 2023; 241:124649. [PMID: 37119907 DOI: 10.1016/j.ijbiomac.2023.124649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Despite the tremendous progress in healthcare sectors, a number of life-threatening infectious, inflammatory, and autoimmune diseases are continuously challenging mankind throughout the globe. In this context, recent successes in utilizing helminth parasite-derived bioactive macromolecules viz. glycoproteins, enzymes, polysaccharides, lipids/lipoproteins, nucleic acids/nucleotides, and small organic molecules for treating various disorders primarily resulted from inflammation. Among the several parasites that infect humans, helminths (cestodes, nematodes, and trematodes) are known as efficient immune manipulators owing to their explicit ability to modulate and modify the innate and adaptive immune responses of humans. These molecules selectively bind to immune receptors on innate and adaptive immune cells and trigger multiple signaling pathways to elicit anti-inflammatory cytokines, expansion of alternatively activated macrophages, T-helper 2, and immunoregulatory T regulatory cell types to induce an anti-inflammatory milieu. Reduction of pro-inflammatory responses and repair of tissue damage by these anti-inflammatory mediators have been exploited for treating a number of autoimmune, allergic, and metabolic diseases. Herein, the potential and promises of different helminths/helminth-derived products as therapeutic agents in ameliorating immunopathology of different human diseases and their mechanistic insights of function at cell and molecular level alongside the molecular signaling cross-talks have been reviewed by incorporating up-to-date findings achieved in the field.
Collapse
Affiliation(s)
- Pritha Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India
| | | | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India.
| |
Collapse
|
7
|
Raisnia R, Mohebalian H, Shahrokhi Sabzevar A, Borji H. Anti-Tumor Effect of Marshallagia marshalli Somatic Antigen on Inhibition Cell Growth of K562. IRANIAN JOURNAL OF PARASITOLOGY 2022; 17:28-35. [PMID: 36046565 PMCID: PMC9375725 DOI: 10.18502/ijpa.v17i1.9013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/02/2021] [Indexed: 06/15/2023]
Abstract
BACKGROUND Today, the inhibitory effect of helminths on tumor cell growth has been proven. We investigated the anti-tumor activity of Marshallagia marshalli somatic against K562 cells. METHODS Different concentrations of M. marshalli somatic antigen were incorporated in the culture medium of K652 cells, and the proliferation and apoptosis were measured after 24, 48, and 72 h, using MTT and Annexin V/PI staining assay. RESULTS Treatment of cells with 1-2 mg/ml antigen for 24-72 h could suppress cell proliferation and increase apoptosis. While treating cells with 0.1 mg/ml antigen for 72 h could inhibit cell growth. There was no meaningful effect on treated cells in comparison with the control group (P< 0.05) after incubating cells for 24- 48 h with 0.1 mg/ml antigen. CONCLUSION M. marshalli somatic antigen had an anti-cancer property, and its role in cancer treatment could be considered as an effective therapeutic method.
Collapse
|
8
|
Mohammadi M, Spotin A, Mahami-Oskouei M, Shanehbandi D, Ahmadpour E, Casulli A, Rostami A, Baghbanzadeh A, Asadi M. MicroRNA-365 promotes apoptosis in human melanoma cell A375 treated with hydatid cyst fluid of Echinococcus granulosus sensu stricto. Microb Pathog 2021; 153:104804. [PMID: 33609644 DOI: 10.1016/j.micpath.2021.104804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 11/18/2022]
Abstract
Hydatid cyst fluid (HCF)-based therapeutics has experimentally targeted approaches for treating human cancer cell lines. MicroRNA-365 (miR-365) has been reported to be an important tumor suppressor miRNA in cancers. However, it remains unknown, how miR-365 plays a pivotal role in inducing apoptosis in HCF-treated cancer cells in vitro. The fertile/infertile HCF was aspirated from liver of infected sheep and in terms of molecular taxonomy was identified as G1 genotype of Echinococcus granulosus sensu stricto. A375 human melanoma cancer cells were cultured into two groups: fertile and infertile HCF-treated A375 cells. To assess the cytotoxicity of various concentrations of HCF on melanoma cells, cell viability was determined by using MTT assay. The IC50 value of HCF on A375 cells was determined 85 μg/mL. Caspase-3 enzymatic activity was evaluated by fluorometric assay in the HCF-treated melanoma cells. In addition, the mRNA expression of Bax, Bcl-2, Caspase-9 and miR-365 were determined by qRT-PCR. Findings of MTT assay showed that concentrations 85 μg/mL to 100 μg/mL of fertile HCF have the highest mortality (50%-52%) on A375 cells during 24 h. The fold change of Bax/Bcl-2 ratio, Caspase-9, miR-365 and Caspase-3 activity was higher in the fertile HCF-treated melanoma cells compared to infertile fluid treated A375 cells and human normal epithelial cell (as control cell). In conclusion, we over-expressed the miR-365 in melanoma A375 cells, via treatment of fertile HCF. Our findings suggested that inducing high expression of miR-365 might be a negative regulator of melanoma growth through activation of pro-apoptotic Bax, Caspase-9 and Caspase-3 that are essential to intrinsic apoptotic pathway. These findings provide new insights into the use of Echinococcus HCF-derived metabolites in the design of drug therapies and in vivo tumor cell vaccine to combat melanoma progression.
Collapse
Affiliation(s)
- Mahnaz Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Adel Spotin
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mahmoud Mahami-Oskouei
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsan Ahmadpour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Adriano Casulli
- WHO Collaborating Centre for the Epidemiology, Detection and Control of Cystic and Alveolar Echinococcosis, Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy; European Union Reference Laboratory for Parasites (EURLP), Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Ali Rostami
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
Porras-Silesky C, Mejías-Alpízar MJ, Mora J, Baneth G, Rojas A. Spirocerca lupi Proteomics and Its Role in Cancer Development: An Overview of Spirocercosis-Induced Sarcomas and Revision of Helminth-Induced Carcinomas. Pathogens 2021; 10:pathogens10020124. [PMID: 33530324 PMCID: PMC7911836 DOI: 10.3390/pathogens10020124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/19/2022] Open
Abstract
Spirocerca lupi is a parasitic nematode of canids that induces a myriad of clinical manifestations in its host and, in 25% of infections, leads to the formation of sarcomas. The description of the protein composition of the excretory and secretory products (Sl-ESP) of S. lupi has shed light on its possible interactions with the host environment, including migration within the host and mechanisms of immunomodulation. Despite this, the process by which S. lupi induces cancer in the dog remains poorly understood, and some hypotheses have arisen regarding these possible mechanisms. In this review, we discuss the role of specific ESP from the carcinogenic helminths Clonorchis sinensis, Opisthorchis viverrini and Schistosoma haematobium in inducing chronic inflammation and cancer in their host’s tissues. The parasitic worms Taenia solium, Echinococcus granulosus, Heterakis gallinarum, Trichuris muris and Strongyloides stercoralis, which have less-characterized mechanisms of cancer induction, are also analyzed. Based on the pathological findings in spirocercosis and the mechanisms by which other parasitic helminths induce cancer, we propose that the sustained inflammatory response in the dog´s tissues produced in response to the release of Sl-ESP homologous to those of other carcinogenic worms may lead to the malignant process in infected dogs.
Collapse
Affiliation(s)
- Catalina Porras-Silesky
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
| | - María José Mejías-Alpízar
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
| | - Javier Mora
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
| | - Gad Baneth
- Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot 7610001, Israel;
| | - Alicia Rojas
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
- Correspondence: ; Tel.: +506-2511-8644
| |
Collapse
|
10
|
Moghaddam SM, Picot S, Ahmadpour E. Interactions between hydatid cyst and regulated cell death may provide new therapeutic opportunities. ACTA ACUST UNITED AC 2019; 26:70. [PMID: 31782727 PMCID: PMC6884020 DOI: 10.1051/parasite/2019070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022]
Abstract
Cystic echinococcosis and alveolar echinococcosis are chronic zoonotic infections, transmitted throughout the world. Development of the cestode larval stages in the liver and lungs causes damage to intermediate hosts, including humans. Several pathways leading to the suppression of host immune response and the survival of the cysts in various hosts are known. Immune response modulation and regulated cell death (RCD) play a fundamental role in cyst formation, development and pathogenesis. RCD, referring to apoptosis, necrosis and autophagy, can be triggered either via intrinsic or extrinsic cell stimuli. In this review, we provide a general overview of current knowledge on the process of RCD during echinococcosis. The study of interactions between RCD and Echinococcus spp. metacestodes may provide in-depth understanding of echinococcosis pathogenesis and open new horizons for human intervention and treatment of the disease.
Collapse
Affiliation(s)
- Sirous Mehrani Moghaddam
- Immunology Research Center, Tabriz University of Medical Sciences, 5166/15731 Tabriz, Iran - Student Research Committee, Tabriz University of Medical Sciences, 5166/15731 Tabriz, Iran
| | - Stephane Picot
- Malaria Research Unit, SMITh, ICBMS, UMR 5246 CNRS INSA CPE University Lyon, 69100 Lyon, France - Institute of Parasitology and Medical Mycology, Croix-Rousse Hospital, Hospices Civils de Lyon, 69004 Lyon, France
| | - Ehsan Ahmadpour
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, 5166/15731 Tabriz, Iran - Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, 5166/15731 Tabriz, Iran
| |
Collapse
|
11
|
Fernandes R, Alves H, Botelho MC. The Cancer Hygiene Hypothesis: From Theory to Therapeutic Helminths. CURRENT CANCER THERAPY REVIEWS 2019. [DOI: 10.2174/1573394714666181003143717] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background::
The “Hygiene Hypothesis” was postulated by David Strachan in 1989 to
explain for the rapid 20th century increase in allergic diseases such as asthma. In this hypothesis,
children from developed countries living in ultra-hygienic environments and avoiding exposure to
viruses, bacteria, and parasites, especially helminthes, have an increased risk of atopic disorders.
Materials & Methods::
Research and online content related to cancer hygiene hypothesis is highlighted
and to illustrate key themes. Guidelines for meaningful participation in DOC activities for
people with diabetes, families, health care providers, and industry are provided.
Results::
The lack of worms leads to failure of stimulation of mucosal Th2 responses. These infections
exert their effect through critically altering T-helper (Th)1/Th2 regulation, and it is postulated
that, thus, they protect against atopy and asthma, through the induction of Th1 regulatory response.
It is now also recognized that this “hygiene hypothesis” concept applies to a wider range
of chronic inflammatory diseases than atopy and asthma, such as diabetes, multiple sclerosis and
cancer.
Conclusion::
Here we discuss the major implications of these findings for the association between
microorganisms and cancer, and also between some immune pathologies, like autoimmune diseases,
and cancer.
Collapse
Affiliation(s)
- Ruben Fernandes
- I3S, Instituto de Investigação e Inovação da Universidade do Porto, Porto, Portugal
| | - Helena Alves
- Fundação Professor Ernesto Morais, Porto, Portugal
| | - Monica C. Botelho
- I3S, Instituto de Investigação e Inovação da Universidade do Porto, Porto, Portugal
| |
Collapse
|
12
|
Wang H, Zhang CS, Fang BB, Li ZD, Li L, Bi XJ, Li WD, Zhang N, Lin RY, Wen H. Thioredoxin peroxidase secreted by Echinococcus granulosus (sensu stricto) promotes the alternative activation of macrophages via PI3K/AKT/mTOR pathway. Parasit Vectors 2019; 12:542. [PMID: 31727141 PMCID: PMC6857240 DOI: 10.1186/s13071-019-3786-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/04/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Larvae of Echinococcus granulosus (sensu lato) dwell in host organs for a long time but elicit only a mild inflammatory response, which indicates that the resolution of host inflammation is necessary for parasite survival. The recruitment of alternatively activated macrophages (AAMs) has been observed in a variety of helminth infections, and emerging evidence indicates that AAMs are critical for the resolution of inflammation. However, whether AAMs can be induced by E. granulosus (s.l.) infection or thioredoxin peroxidase (TPx), one of the important molecules secreted by the parasite, remains unclear. METHODS The activation status of peritoneal macrophages (PMs) derived from mice infected with E. granulosus (sensu stricto) was analyzed by evaluating the expression of phenotypic markers. PMs were then treated in vivo and in vitro with recombinant EgTPx (rEgTPx) and its variant (rvEgTPx) in combination with parasite excretory-secretory (ES) products, and the resulting activation of the PMs was evaluated by flow cytometry and real-time PCR. The phosphorylation levels of various molecules in the PI3K/AKT/mTOR pathway after parasite infection and antigen stimulation were also detected. RESULTS The expression of AAM-related genes in PMs was preferentially induced after E. granulosus (s.s.) infection, and phenotypic differences in cell morphology were detected between PMs isolated from E. granulosus (s.s.)-infected mice and control mice. The administration of parasite ES products or rEgTPx induced the recruitment of AAMs to the peritoneum and a notable skewing of the ratio of PM subsets, and these effects are consistent with those obtained after E. granulosus (s.s.) infection. ES products or rEgTPx also induced PMs toward an AAM phenotype in vitro. Interestingly, this immunomodulatory property of rEgTPx was dependent on its antioxidant activity. In addition, the PI3K/AKT/mTOR pathway was activated after parasite infection and antigen stimulation, and the activation of this pathway was suppressed by pre-treatment with an AKT/mTOR inhibitor. CONCLUSIONS This study demonstrates that E. granulosus (s.s.) infection and ES products, including EgTPx, can induce PM recruitment and alternative activation, at least in part, via the PI3K/AKT/mTOR pathway. These results suggest that EgTPx-induced AAMs might play a key role in the resolution of inflammation and thereby favour the establishment of hydatid cysts in the host.
Collapse
Affiliation(s)
- Hui Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, People's Republic of China.,Branch of The First Affiliated Hospital of Xinjiang Medical University, Changji, 831100, Xinjiang, People's Republic of China.,Basic Medical College, Xinjiang Medical University, Ürümqi, 830054, Xinjiang, China
| | - Chuan-Shan Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, People's Republic of China.,Xinjiang Key Laboratory of Echinococcosis, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, China
| | - Bin-Bin Fang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, People's Republic of China
| | - Zhi-De Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, People's Republic of China
| | - Liang Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, People's Republic of China
| | - Xiao-Juan Bi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, People's Republic of China
| | - Wen-Ding Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, People's Republic of China
| | - Ning Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, People's Republic of China
| | - Ren-Yong Lin
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, People's Republic of China. .,Xinjiang Key Laboratory of Echinococcosis, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, China.
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, People's Republic of China. .,Xinjiang Key Laboratory of Echinococcosis, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, China.
| |
Collapse
|
13
|
Guan W, Zhang X, Wang X, Lu S, Yin J, Zhang J. Employing Parasite Against Cancer: A Lesson From the Canine Tapeworm Echinococcus Granulocus. Front Pharmacol 2019; 10:1137. [PMID: 31607934 PMCID: PMC6774290 DOI: 10.3389/fphar.2019.01137] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/03/2019] [Indexed: 01/03/2023] Open
Abstract
Cystic echinococcosis (CE), a devastating zoonotic condition caused by the tapeworm Echinococcus granulosus, remain a significant public health problem worldwide. However, after a negative correlation between solid tumor and CE has been incidentally discovered, accumulating evidence have suggested that this parasite may induce anticancer effect through activating host immune response and secreting molecules with anticancer potential, which may provide some new understanding for immunotherapy. This article will review the evidence supporting the anticancer effect of E. granulosus and its underlying mechanisms and discuss the possible implications in immunotherapy.
Collapse
Affiliation(s)
- Wang Guan
- Department of Cancer Prevention and Treatment, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoqin Zhang
- Intensive Care Unit, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Wang
- Department of Cancer Prevention and Treatment, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shun Lu
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Yin
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinxin Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
The Untapped Pharmacopeic Potential of Helminths. Trends Parasitol 2018; 34:828-842. [PMID: 29954660 DOI: 10.1016/j.pt.2018.05.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 02/06/2023]
Abstract
The dramatic rise in immunological disorders that occurs with socioeconomic development is associated with alterations in microbial colonization and reduced exposure to helminths. Excretory-secretory (E/S) helminth products contain a mixture of proteins and low-molecular-weight molecules representing the primary interface between parasite and host. Research has shown great pharmacopeic potential for helminth-derived products in animal disease models and even in clinical trials. Although in its infancy, the translation of worm-derived products into therapeutics is highly promising. Here, we focus on important key aspects in the development of immunomodulatory drugs, also highlighting novel approaches that hold great promise for future development of innovative research strategies.
Collapse
|