1
|
Serrano-Rodríguez M, Araya JE, Cortez M, Orrego PR. Cytotoxic Effect of Trypanosoma cruzi Calcineurin B Against Melanoma and Adenocarcinoma Cells In Vitro. Adv Pharmacol Pharm Sci 2024; 2024:5394494. [PMID: 39640496 PMCID: PMC11620811 DOI: 10.1155/adpp/5394494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 09/30/2024] [Accepted: 10/26/2024] [Indexed: 12/07/2024] Open
Abstract
Chagas disease caused by the obligate intracellular flagellate protozoan Trypanozoma cruzi infects about 6 million people. From the 1930s to the present, the antitumor capacity of T. cruzi has been studied; however, the identification of the responsible molecules for this effect remains undiscovered. Calcineurin, a calcium/calmodulin-dependent serine/threonine phosphatase, is a heterodimer consisting of a catalytic subunit (CaNA) and a regulatory subunit (CaNB). It has been described that T. cruzi CaN is involved in the cell invasion and proliferation of the parasite. Recently, extracellular human CaNB has been demonstrated to be capable of inhibiting tumor growth cells, conferring an antitumor effect; however, the extracellular role of T. cruzi CaNB (TcCaNB) is still unknown. The objective of this work was to investigate the antitumor potential of TcCaNB by interacting with membrane proteins and evaluating its effects on the viability, proliferation, and morphology of tumor cells in vitro. Additionally, the possible mechanism of action of TcCaNB was explored. Murine melanoma (B16-F10), human cervical adenocarcinoma (HeLa), and African green monkey kidney epithelial (Vero) cell lines were employed for in vitro assays. Far Western blot and immunofluorescence were performed to assess the interaction of TcCaNB with membrane proteins, and the effect of TcCaNB on cell viability and proliferation was evaluated using the MTS assay and the CyQUANT NF assay, respectively. The effect of the caspase inhibitor Z-VAD-FMK on TcCaNB-stimulated tumor cells was investigated to determine if TcCaNB-induced cell death was associated with apoptosis. To assess cell cycle progression, TcCaNB-treated cells were analyzed by flow cytometry. In this study, the results showed an interaction of TcCaNB with cell membrane proteins in B16-F10 and HeLa tumor lines, indicating that TcCaNB is capable of decreasing viability and proliferation of B16-F10 and HeLa cells, with no significant effect observed in Vero cells. Furthermore, morphological changes were observed in tumor cells treated with TcCaNB. DNA fragmentations and inhibition of caspases with Z-VAD-FMK partially counteracted the cytotoxic effects of TcCaNB on tumor cells, suggesting that TcCaNB-induced cell death might be associated with apoptosis. Additionally, TcCaNB caused S phase cell cycle arrest in HeLa cells, with an increase in the sub-G1 population indicative of apoptosis, while no significant effects were observed in Vero cells.
Collapse
Affiliation(s)
- Mayela Serrano-Rodríguez
- Biomedical Department, Faculty of Health Sciences, University of Antofagasta, Antofagasta 1240000, Chile
| | - Jorge E. Araya
- Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta 1240000, Chile
| | - Mauro Cortez
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Patricio R. Orrego
- Biomedical Department, Faculty of Health Sciences, University of Antofagasta, Antofagasta 1240000, Chile
| |
Collapse
|
2
|
Alizadeh G, Kheirandish A, Alipour M, Jafari M, Radfar M, Bybordi T, Rafiei-Sefiddashti R. The role of helminths and their antigens in cancer therapy: insights from cell line models. Infect Agent Cancer 2024; 19:52. [PMID: 39385244 PMCID: PMC11465614 DOI: 10.1186/s13027-024-00613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Recent articles have explored the effect of worms on cancer cells. This review focused on various cell cultures employed to understand which cells are more commonly and less utilized. METHODS The present review analyzed studies published between 2013 and 2023 to obtain information about different cell cultures used in cancer studies involving helminths. Databases such as PubMed, Google Scholar, HINARI, and the Cochrane Library were searched. RESULTS This search yielded 130 records, but 97 papers were excluded because they were either irrelevant to the research topic (n = 72) or contradicted the research idea (n = 25).The remaining twenty-one articles focused on different types of worms, such as Echinococcus granulosus, Clonorchis sinensis, Opisthorchis felineus, Opisthorchis viverrini, Trichinella spiralis, Toxocara canis, and Heligmosomoides polygyrus. CONCLUSION Due to the presence of numerous antigens, parasites at different growth stages can impact various cells through unknown mechanisms. Given the high diversity of antigens and their effects, artificial intelligence can assist in predicting initial outcomes for future studies.
Collapse
Affiliation(s)
- Gita Alizadeh
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Kheirandish
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Alipour
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahnaz Jafari
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdis Radfar
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tina Bybordi
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Raheleh Rafiei-Sefiddashti
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Eissa MM, Salem AE, El Skhawy N. Parasites revive hope for cancer therapy. Eur J Med Res 2024; 29:489. [PMID: 39367471 PMCID: PMC11453045 DOI: 10.1186/s40001-024-02057-2] [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: 06/10/2024] [Accepted: 09/10/2024] [Indexed: 10/06/2024] Open
Abstract
Parasites have attained a life-long stigma of being detrimental organisms with deleterious outcomes. Yet, recently, a creditable twist was verified that can dramatically change our perception of those parasites from being a source of misery to millions of people to a useful anti-cancerous tool. Various parasites have shown promise to combat cancer in different experimental models, including colorectal, lung, and breast cancers, among others. Helminths and protozoan parasites, as well as their derivatives such as Echinococcus granulosus protein KI-1, Toxoplasma gondii GRA15II, and Trypanosoma cruzi calreticulin, have demonstrated the ability to inhibit tumor growth, angiogenesis, and metastasis. This article provides an overview of the literature on various cancer types that have shown promising responses to parasite therapy in both in vitro and in vivo animal studies. Parasites have shown anti-neoplastic activity through a variety of mechanisms that collectively contribute to their anti-cancer properties. These include immunomodulation, inhibition of angiogenesis, and molecular mimicry with cancer cells. This review article sheds light on this intriguing emerging field and emphasizes the value of collaborative multidisciplinary research projects with funding agencies and pharmaceutical companies. Thus, these strategies would secure continuous exploration of this new avenue and accelerate the advancement of cancer therapy research. Although experimental studies are heavily conducted by leaps and bounds, further steps are definitely lagging. Upgrading research from the experimental level to the clinical trial would be a wise progression toward efficient exploitation of the anti-neoplastic capabilities of parasites, ultimately saving countless lives.
Collapse
Affiliation(s)
- Maha M Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Ahmed Ebada Salem
- Department of Radiology and Nuclear Medicine, School of Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 48123, USA
| | - Nahla El Skhawy
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
4
|
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
|
5
|
Maravelez Acosta VA, Crisóstomo Vázquez MDP, Eligio García L, Franco Sandoval LO, Castro Pérez D, Patiño López G, Medina Contreras O, Jiménez Cardoso E. Tumor-Suppressive Cross-Linking of Anti- T. cruzi Antibodies in Acute Lymphoblastic Leukemia. Int J Mol Sci 2024; 25:8307. [PMID: 39125875 PMCID: PMC11313589 DOI: 10.3390/ijms25158307] [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: 06/12/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
Parasites have been associated with possible anticancer activity, including Trypanosoma cruzi, which has been linked to inhibiting the growth of solid tumors. To better understand this antitumor effect, we investigated the association of anti-T. cruzi antibodies with B cells of the acute lymphoblastic leukemia (ALL) SUPB15 cell line. The antibodies were generated in rabbits. IgGs were purified by affinity chromatography. Two procedures (flow cytometry (CF) and Western blot(WB)) were employed to recognize anti-T. cruzi antibodies on SUPB15 cells. We also used CF to determine whether the anti-T. cruzi antibodies could suppress SUPB15 cells. The anti-T. cruzi antibodies recognized 35.5% of the surface antigens of SUPB15. The complement-dependent cytotoxicity (CDC) results demonstrate the cross-suppression of anti-T. cruzi antibodies on up to 8.4% of SUPB15 cells. For the WB analysis, a band at 100 kDa with high intensity was sequenced using mass spectrometry, identifying the protein as nucleolin. This protein may play a role in the antitumor effect on T. cruzi. The anti-T. cruzi antibodies represent promising polyclonal antibodies that have the effect of tumor-suppressive cross-linking on cancer cells, which should be further investigated.
Collapse
Affiliation(s)
- Víctor Alberto Maravelez Acosta
- Laboratorio de Investigación en Parasitología, Hospital Infantil de México Federico Gómez (HIMFG), Dr. Márquez 162. Col Doctores, Cuauhtémoc, México City 06720, Mexico; (V.A.M.A.); (M.d.P.C.V.); (L.E.G.); (L.O.F.S.); (D.C.P.)
| | - María del Pilar Crisóstomo Vázquez
- Laboratorio de Investigación en Parasitología, Hospital Infantil de México Federico Gómez (HIMFG), Dr. Márquez 162. Col Doctores, Cuauhtémoc, México City 06720, Mexico; (V.A.M.A.); (M.d.P.C.V.); (L.E.G.); (L.O.F.S.); (D.C.P.)
| | - Leticia Eligio García
- Laboratorio de Investigación en Parasitología, Hospital Infantil de México Federico Gómez (HIMFG), Dr. Márquez 162. Col Doctores, Cuauhtémoc, México City 06720, Mexico; (V.A.M.A.); (M.d.P.C.V.); (L.E.G.); (L.O.F.S.); (D.C.P.)
| | - Luz Ofelia Franco Sandoval
- Laboratorio de Investigación en Parasitología, Hospital Infantil de México Federico Gómez (HIMFG), Dr. Márquez 162. Col Doctores, Cuauhtémoc, México City 06720, Mexico; (V.A.M.A.); (M.d.P.C.V.); (L.E.G.); (L.O.F.S.); (D.C.P.)
| | - Denia Castro Pérez
- Laboratorio de Investigación en Parasitología, Hospital Infantil de México Federico Gómez (HIMFG), Dr. Márquez 162. Col Doctores, Cuauhtémoc, México City 06720, Mexico; (V.A.M.A.); (M.d.P.C.V.); (L.E.G.); (L.O.F.S.); (D.C.P.)
| | - Genaro Patiño López
- Unidad de Investigación en Inmunología y Proteomica, Hospital Infantil de México Federico Gómez (HIMFG), Dr. Márquez 162. Col Doctores, Cuauhtémoc, México City 06720, Mexico;
| | - Oscar Medina Contreras
- Unidad de Investigación Epidemiologica en Endocrinologia y Nutricion, Hospital Infantil de México Federico Gómez (HIMFG), Dr. Márquez 162. Col Doctores, Cuauhtémoc, México City 06720, Mexico;
| | - Enedina Jiménez Cardoso
- Laboratorio de Investigación en Parasitología, Hospital Infantil de México Federico Gómez (HIMFG), Dr. Márquez 162. Col Doctores, Cuauhtémoc, México City 06720, Mexico; (V.A.M.A.); (M.d.P.C.V.); (L.E.G.); (L.O.F.S.); (D.C.P.)
| |
Collapse
|
6
|
Walter NS, Bhattacharyya S. Mining parasites for their potential as novel therapeutic agents against cancer. Med Oncol 2024; 41:211. [PMID: 39073638 DOI: 10.1007/s12032-024-02458-7] [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: 06/07/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Despite recent advances in the management and therapeutic of cancer, the treatment of the disease is limited by its high cost and severe side effects. In this scenario, there is an unmet need to identify novel treatment alternatives for this dreaded disease. Recently there is growing evidence that parasites may cause anticancer effects because of a negative correlation between parasitic infections and tumour growth despite some parasites that are known to exhibit pro-carcinogenic effects. It has been observed that parasites exert an anticancer effect either by activating the host's immune response or by secreting certain molecules that exhibit anticancer potential. The activation of the immune response by these parasitic organisms results in the inhibition of some of the hallmarks of cancer such as tumour proliferation, angiogenesis, and metastasis. This review summarizes the current advances as well as the mechanisms underlying the possible implications of this diverse group of organisms as anticancer agents.
Collapse
Affiliation(s)
- Neha Sylvia Walter
- Department of Biophysics, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Shalmoli Bhattacharyya
- Department of Biophysics, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India.
| |
Collapse
|
7
|
Barati N, Shojaeian A, Ramezani M, Kalhori F, Yavari Bazl MS, Zafari S, Asl SS, Motavallihaghi S. Investigating the effect of parasites (toxoplasma gondii RH strain, Leishmania major (MRHO/IR/75/ER), and hydatid cyst) antigens on Alzheimer's disease: An in vivo evaluation. Exp Neurol 2024; 377:114813. [PMID: 38735456 DOI: 10.1016/j.expneurol.2024.114813] [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: 03/07/2024] [Revised: 04/22/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024]
Abstract
This study aimed to investigate the effects of parasite antigens on Alzheimer's symptoms in animal model. Alzheimer's model was induced in Wistar rats using Amyloid-beta peptide, and treated with parasite crude antigens from T. gondii RH strain, L. major (MRHO/IR/75/ER), and HC. Spectrophotometry and real-time PCR were used to evaluate the oxidative stress levels, antioxidant enzyme activity, and gene expression of NLRP3, IL-8, IL-1β, and Caspase-1. Histological assays were performed to investigate structural changes in the hippocampus. Apoptosis was analyzed using an Annexin V Apoptosis by Flow cytometer. The levels of total oxidant, antioxidant, and SOD increased in the Alzheimer's group compared with the control group, but these factors were lower in the L. major group. The apoptosis in the treated groups was lower compared to the Alzheimer's group. IL-8 expression was significantly higher in all Alzheimer's groups, but decreased in the HC and L. major treated group compared to Alzheimer's. IL-1β and Caspase-1 expression were similarly increased in all groups compared with the control group, but decreased in the antigen-treated groups compared with Alzheimer's. NLRP3 expression was increased in all groups compared with the control group, with lower expression in HC group, but significantly decreased in L. major group compared with Alzheimer's. In histological results, only L. major group could play a therapeutic role in pathological damage of the hippocampus. The results showed that parasite antigens, specifically L. major antigens, may have neuroprotective effects that reduce oxidative stress, apoptosis, and histopathological changes in response to AD in animal model.
Collapse
Affiliation(s)
- Nastaran Barati
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Shojaeian
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahdi Ramezani
- Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fereshte Kalhori
- Department of Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Salman Zafari
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Soleimani Asl
- Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyedmousa Motavallihaghi
- Department of Medical Parasitology and Mycology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
8
|
Younis SS, Salama AM, Elmehy DA, Heabah NA, Rabah HM, Elakshar SH, Awad RA, Gamea GA. Trichinella spiralis Larval Extract as a Biological Anti-Tumor Therapy in a Murine Model of Ehrlich Solid Carcinoma. Parasite Immunol 2024; 46:e13035. [PMID: 38712475 DOI: 10.1111/pim.13035] [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: 09/07/2023] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 05/08/2024]
Abstract
Trichinella spiralis (T. spiralis) is an immunomodulating parasite that can adversely affect tumor growth and extend host lifespan. The aim of this study was to elucidate the mechanisms by which T. spiralis larval antigens achieve this effect using Ehrlich solid carcinoma (ESC) murine model. Assessment was done by histopathological and immunohistochemical analysis of caspase-3, TNF-α, Ki-67 and CD31. Additionally, Bcl2 and Bcl2-associated protein X (Bax) relative gene expression was assessed by molecular analysis for studying the effect of T. spiralis crude larval extract (CLE) antigen on tumor necrosis, apoptosis, cell proliferation and angiogenesis. We found that both T. spiralis infection and CLE caused a decrease in the areas of necrosis in ESC. Moreover, they led to increased apoptosis through activation of caspase-3, up-regulation of pro-apoptotic gene, Bax and down-regulation of anti-apoptotic gene, Bcl2. Also, T. spiralis infection and CLE diminished ESC proliferation, as evidenced by decreasing Ki-67. T. spiralis infection and CLE were able to suppress the development of ESC by inhibiting tumor proliferation, inducing apoptosis and decreasing tumor necrosis, with subsequent decrease in tumor metastasis. T. spiralis CLE antigen may be considered as a promising complementary immunotherapeutic agent in the treatment of cancer.
Collapse
Affiliation(s)
- Salwa S Younis
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Amina M Salama
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Dalia A Elmehy
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Nehal A Heabah
- Department of Pathology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hanem M Rabah
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Sara H Elakshar
- Department of Clinical Oncology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Radwa A Awad
- Department of Clinical Oncology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ghada A Gamea
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
9
|
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
|
10
|
Kandalai S, Li H, Zhang N, Peng H, Zheng Q. The human microbiome and cancer: a diagnostic and therapeutic perspective. Cancer Biol Ther 2023; 24:2240084. [PMID: 37498047 PMCID: PMC10376920 DOI: 10.1080/15384047.2023.2240084] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/09/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023] Open
Abstract
Recent evidence has shown that the human microbiome is associated with various diseases, including cancer. The salivary microbiome, fecal microbiome, and circulating microbial DNA in blood plasma have all been used experimentally as diagnostic biomarkers for many types of cancer. The microbiomes present within local tissue, other regions, and tumors themselves have been shown to promote and restrict the development and progression of cancer, most often by affecting cancer cells or the host immune system. These microbes have also been shown to impact the efficacy of various cancer therapies, including radiation, chemotherapy, and immunotherapy. Here, we review the research advances focused on how microbes impact these different facets and why they are important to the clinical care of cancer. It is only by better understanding the roles these microbes play in the diagnosis, development, progression, and treatment of cancer, that we will be able to catch and treat cancer early.
Collapse
Affiliation(s)
- Shruthi Kandalai
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Huapeng Li
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Nan Zhang
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Haidong Peng
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Qingfei Zheng
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Biological Chemistry and Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
11
|
Kaufman CD, Farré C, Biscari L, Pérez AR, Alloatti A. Trypanosoma cruzi, Chagas disease and cancer: putting together the pieces of a complex puzzle. Front Cell Dev Biol 2023; 11:1260423. [PMID: 38188016 PMCID: PMC10768204 DOI: 10.3389/fcell.2023.1260423] [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: 07/17/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024] Open
Abstract
Considering the extensive and widespread impact on individuals, cancer can presently be categorized as a pandemic. In many instances, the development of tumors has been linked to endemic microbe infections. Among parasitic infections, Trypanosoma cruzi stands out as one of the most extensively discussed protozoans in the literature that explores the association between diseases of parasite origin and cancer. However, the effective association remains an unsolved paradox. Both the parasite, along with protozoan-derived molecules, and the associated antiparasitic immune response can induce alterations in various host cell pathways, leading to modifications in cell cycle, metabolism, glycosylation, DNA mutations, or changes in neuronal signaling. Furthermore, the presence of the parasite can trigger cell death or a senescent phenotype and modulate the immune system, the metastatic cascade, and the formation of new blood vessels. The interaction among the parasite (and its molecules), the host, and cancer undoubtedly encompasses various mechanisms that operate differentially depending on the context. Remarkably, contrary to expectations, the evidence tilts the balance toward inhibiting tumor growth or resisting tumor development. This effect is primarily observed in malignant cells, rather than normal cells, indicating a selective or specific component. Nevertheless, nonspecific bystander mechanisms, such as T. cruzi's adjuvancy or the presence of proinflammatory cytokines, may also play a significant role in this phenomenon. This work aims to elucidate this complex scenario by synthesizing the main findings presented in the literature and by proposing new questions and answers, thereby adding pieces to this challenging puzzle.
Collapse
Affiliation(s)
- Cintia Daniela Kaufman
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Cecilia Farré
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Lucía Biscari
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Ana Rosa Pérez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Andrés Alloatti
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
12
|
Yousefi M, Akbari M, Hadipour M, Dehkordi AB, Farahbakhsh Z, Darani HY. Parasites as potential targets for cancer immunotherapy. J Cancer Res Clin Oncol 2023; 149:8027-8038. [PMID: 36949175 DOI: 10.1007/s00432-023-04694-2] [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: 11/30/2022] [Accepted: 03/13/2023] [Indexed: 03/24/2023]
Abstract
Parasites and cancers have some common antigens. Much scientific evidence in the human population, animal models, and in vitro experiments exhibit that parasites have significant anti-cancer effects. The larval stage of the tapeworm Echinococcus granulosus, Toxoplasma gondii, Trypanosoma cruzy, Plasmodium's, and Trichinella spiralis are among the parasites that have been subjects of anti-cancer research in the last decades. Anti-tumor effects of parasites may be due to the direct impact of the parasites per se or indirectly due to the immune response raised against common antigens between malignant cells and parasites. This manuscript reviews the anti-cancer effects of parasites and possible mechanisms of these effects. Options for using parasites or their antigens for cancer treatment in the future have been discussed.
Collapse
Affiliation(s)
- Morteza Yousefi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadesmail Akbari
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mahboubeh Hadipour
- Department of Parasitology and Mycology, Faculty of Medicine, Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azar Balouti Dehkordi
- Department of Parasitology and Mycology, Faculty of Medicine, Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zohreh Farahbakhsh
- Department of Parasitology and Mycology, Faculty of Medicine, Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Yousofi Darani
- Department of Parasitology and Mycology, Faculty of Medicine, Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
13
|
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
|
14
|
Eissa MM, Gaafar MR, Younis LK, Ismail CA, El Skhawy N. Prophylactic antineoplastic activity of Toxoplasma gondii RH derived antigen against ehrlich solid carcinoma with evidence of shared antigens by comparative immunoblotting. Infect Agent Cancer 2023; 18:21. [PMID: 37029378 PMCID: PMC10082516 DOI: 10.1186/s13027-023-00500-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/22/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND With cancer cases escalation, an urgent request to develop novel combating strategies arise. Pathogen-based cancer-immunotherapy is getting more consideration. Autoclaved parasitic antigens seem promising candidates, taking steadily their first steps. Our aim was to examine the prophylactic antineoplastic activity of autoclaved Toxoplasma vaccine (ATV) and to test for the shared antigen theory between Toxoplasma gondii and cancer cells. METHODS Mice were immunized with ATV followed by Ehrlich solid carcinoma (ESC) inoculation. Tumor weight, volume, histopathology, and immunohistochemistry for CD8+ T cells, Treg cells and VEGF were assessed. In addition, the proposed shared antigen theory between parasites and cancer was also verified using SDS-PAGE and immunoblotting. RESULTS Results revealed powerful prophylactic activity of ATV with 13.3% inhibition of ESC incidence, significant reduction in tumor weight and volume in ATV vaccinated mice. Immunologically, significantly higher CD8+T cells and lower FOXP3+ Treg cells surrounded and infiltrated ESC in ATV immunized mice with higher CD8+T/Treg cells ratio and significant antiangiogenic effect. Moreover, SDS-PAGE and immunoblotting showed four shared bands between Ehrlich carcinoma and ATV of approximate molecular weights 60, 26, 22 and 12.5 KDa. CONCLUSION Exclusively, we demonstrated a prophylactic antineoplastic activity of autoclaved Toxoplasma vaccine against ESC. Moreover, to the best of our knowledge this is the first report highlighting the existence of cross-reactive antigens between Toxoplasma gondi parasite and cancer cells of Ehrlich carcinoma.
Collapse
Affiliation(s)
- Maha M Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Maha R Gaafar
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Layla K Younis
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Cherine A Ismail
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nahla El Skhawy
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
15
|
Ribeiro Franco PI, do Carmo Neto JR, Miguel MP, Machado JR, Nunes Celes MR. Cancer and Trypanosoma cruzi: Tumor induction or protection? Biochimie 2023; 207:113-121. [PMID: 36368477 DOI: 10.1016/j.biochi.2022.10.019] [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: 09/28/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022]
Abstract
Trypanosoma cruzi causes Chagas disease, a neglected disease that can be divided, overall, into acute and chronic phases. Understanding the mechanisms underlying its progression is based on the parasite-host interactions occurring during the infection. Although the pathophysiology of the main symptomatic forms of Chagas disease has been the subject of several studies, little is known about their relationship with the development of different types of cancer. Therefore, knowledge regarding the molecular aspects of infection in the host, as well as the influence of the immune response in the parasite and the host, can help to understand the association between Chagas disease and tumor development. This review aims to summarize the main molecular mechanisms related to T. cruzi-dependent carcinogenic development and the mechanisms associated with tumor protection mediated by different parasite components.
Collapse
Affiliation(s)
- Pablo Igor Ribeiro Franco
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, 74605-050, Goiania, Goiás, Brazil.
| | - José Rodrigues do Carmo Neto
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, 74605-050, Goiania, Goiás, Brazil
| | - Marina Pacheco Miguel
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, 74605-050, Goiania, Goiás, Brazil; Veterinary and Animal Science School, Federal University of Goiás, 74605-050, Goiania, Goiás, Brazil
| | - Juliana Reis Machado
- Department of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, 38025-180, Uberaba, Minas Gerais, Brazil
| | - Mara Rúbia Nunes Celes
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, 74605-050, Goiania, Goiás, Brazil
| |
Collapse
|
16
|
Freire T, Landeira M, Giacomini C, Festari MF, Pittini Á, Cardozo V, Brosque A, Monin L, da Costa V, Faral-Tello P, Robello C, Osinaga E. Trypanosoma cruzi-Derived Molecules Induce Anti-Tumour Protection by Favouring Both Innate and Adaptive Immune Responses. Int J Mol Sci 2022; 23:ijms232315032. [PMID: 36499361 PMCID: PMC9739173 DOI: 10.3390/ijms232315032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022] Open
Abstract
Lung cancer remains the leading cause of cancer mortality worldwide. Thus, the development of strategies against this type of cancer is of high value. Parasite infections can correlate with lower cancer incidence in humans and their use as vaccines has been recently explored in preclinical models. In this study, we investigated whether immunisations with a Trypanosoma cruzi lysate from epimastigotes protect from lung tumour growth in mice. We also explore the role of parasite glycans in the induction of the protective immune response. A pre-clinical murine cancer model using the lung tumour cell line LL/2 was used to evaluate the anti-tumour potential, both in preventive and therapeutic settings, of a T. cruzi epimastigote-derived protein lysate. Immunisation with the parasite lysate prevents tumour growth and induces both humoral and cellular anti-tumour immune responses to LL-2 cancer cells. The induced immunity and tumour protection were associated with the activation of natural killer (NK) cells, the production of interferon-γ (IFN-γ) and tumour cell cytotoxicity. We also show that mannose residues in the T. cruzi lysate induce Toll-like receptor (TLR) signalling. The evaluated T. cruzi lysate possesses anti-tumour properties likely by activating innate and adaptive immunity in a process where carbohydrates seem to be essential.
Collapse
Affiliation(s)
- Teresa Freire
- Laboratorio de Inmunomodulación y Vacunas, Departamento Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
- Correspondence: (T.F.); (E.O.)
| | - Mercedes Landeira
- Laboratorio de Inmunomodulación y Vacunas, Departamento Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
| | - Cecilia Giacomini
- Laboratorio de Bioquímica, Departamento de Biociencias, Facultad de Química, UdelaR, Gral Flores 2124, Montevideo 11800, Uruguay
| | - María Florencia Festari
- Laboratorio de Inmunomodulación y Vacunas, Departamento Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
| | - Álvaro Pittini
- Departamento de Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Viviana Cardozo
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Alina Brosque
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Leticia Monin
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Valeria da Costa
- Laboratorio de Inmunomodulación y Vacunas, Departamento Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
| | - Paula Faral-Tello
- Laboratorio de Interacciones Hospedero-Patógeno, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Carlos Robello
- Laboratorio de Interacciones Hospedero-Patógeno, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
| | - Eduardo Osinaga
- Departamento de Inmunobiología, Facultad de Medicina, UdelaR, Gral Flores 2125, Montevideo 11800, Uruguay
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- Correspondence: (T.F.); (E.O.)
| |
Collapse
|
17
|
Eligio García L, Crisóstomo Vázquez MDP, Maravelez Acosta VA, Soria Guerrero M, Cortés Campos A, Jiménez Cardoso E. Trypanosoma cruzi Antigenic Proteins Shared with Acute Lymphoblastic Leukemia and Neuroblastoma. Pharmaceuticals (Basel) 2022; 15:1421. [PMID: 36422551 PMCID: PMC9693088 DOI: 10.3390/ph15111421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 02/19/2024] Open
Abstract
Background. Research studies indicate that immunization with protein extracts of Trypanosoma cruzi, the protozoan parasite that causes Chagas disease, prevents the appearance of tumors in 60% of mice injected with the murine lung carcinoma tumor line. The molecular basis of this process is unknown, although the presence of specific antigens in tumor cells and on the surface of T. cruzi suggests an antiparasitic immune response, with an effective cross-reaction against cancer cells, hence the importance to identify the antigens involved and determine their potential as target cells in anticancer therapy. Aim. This study aimed to determine the presence of antigenic proteins of T. cruzi shared with acute lymphoblastic leukemia and neuroblastoma cells. Material and methods. To achieve this, polyclonal antibodies against T. cruzi were developed in rabbits, and reactivity was determined with protein extracts of acute lymphoblastic leukemia cells and neuroblastoma. The immunodetection of five different strains of T. cruzi against anti-T. cruzi polyclonal antibodies was also performed. Conclusion. The study allows the knowledge of the immunological interactions between cancer and parasites to be expanded and, therefore, contributes to the design of more and better projects that improve the therapeutic strategies applied in oncology.
Collapse
Affiliation(s)
| | | | | | | | | | - Enedina Jiménez Cardoso
- Laboratorio de Investigación en Parasitología, Hospital Infantil de México Federico Gómez (HIMFG), Dr. Márquez 162. Col Doctores, Cuauhtémoc, México City 06720, Mexico
| |
Collapse
|
18
|
Oliveira FMS, Cruz RE, Pinheiro GRG, Caliari MV. Comorbidities involving parasitic diseases: A look at the benefits and complications. Exp Biol Med (Maywood) 2022; 247:1819-1826. [PMID: 35876147 PMCID: PMC9679356 DOI: 10.1177/15353702221108387] [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: 12/29/2022] Open
Abstract
Parasitic infections acquired by the population cause substantial morbidity worldwide, with individuals from developing countries being most affected. Some parasites remain in the host for long periods, settling in different organs, manipulating the flow of nutrients and metabolites, and influencing the immune response, favoring their adaptation. The host attempts to counteract the metabolic and immunological alterations and the possible damage caused by infection. These metabolic and immunological changes experienced by the host can influence the progression of other existing morbidities or those that will be acquired in the future. Cancer and metabolic diseases are also frequent causes of morbidity in the world population. The large numbers of individuals affected by cancer and metabolic diseases and the high prevalence of morbidity caused by parasitic diseases favor the development of comorbidity involving these pathologies. This review provides an overview of major advances in research on cancer and metabolic diseases associated with parasitic infections. Information about hosts and parasites such as alterations of the immune response, metabolism and adaptation mechanisms of the parasites, and parasitic molecules with therapeutic potential is provided, as well as the beneficial results or complications related to the comorbidities discussed herein. We emphasize the need to conduct additional studies addressing comorbidities associated with parasitic infections to improve the understanding of the impact of this association on the progression of morbidities, as well as the possibility of the therapeutic use of and therapeutic approaches involving parasites.
Collapse
Affiliation(s)
- Fabrício Marcus Silva Oliveira
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Ruth Elizabeth Cruz
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Guilherme Rafael Gomide Pinheiro
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Marcelo Vidigal Caliari
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil,Marcelo Vidigal Caliari.
| |
Collapse
|
19
|
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
|
20
|
Berois N, Pittini A, Osinaga E. Targeting Tumor Glycans for Cancer Therapy: Successes, Limitations, and Perspectives. Cancers (Basel) 2022; 14:cancers14030645. [PMID: 35158915 PMCID: PMC8833780 DOI: 10.3390/cancers14030645] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Aberrant glycosylation is a common feature of many cancers, and it plays crucial roles in tumor development and biology. Cancer progression can be regulated by several physiopathological processes controlled by glycosylation, such as cell–cell adhesion, cell–matrix interaction, epithelial-to-mesenchymal transition, tumor proliferation, invasion, and metastasis. Different mechanisms of aberrant glycosylation lead to the formation of tumor-associated carbohydrate antigens (TACAs), which are suitable for selective cancer targeting, as well as novel antitumor immunotherapy approaches. This review summarizes the strategies developed in cancer immunotherapy targeting TACAs, analyzing molecular and cellular mechanisms and state-of-the-art methods in clinical oncology. Abstract Aberrant glycosylation is a hallmark of cancer and can lead to changes that influence tumor behavior. Glycans can serve as a source of novel clinical biomarker developments, providing a set of specific targets for therapeutic intervention. Different mechanisms of aberrant glycosylation lead to the formation of tumor-associated carbohydrate antigens (TACAs) suitable for selective cancer-targeting therapy. The best characterized TACAs are truncated O-glycans (Tn, TF, and sialyl-Tn antigens), gangliosides (GD2, GD3, GM2, GM3, fucosyl-GM1), globo-serie glycans (Globo-H, SSEA-3, SSEA-4), Lewis antigens, and polysialic acid. In this review, we analyze strategies for cancer immunotherapy targeting TACAs, including different antibody developments, the production of vaccines, and the generation of CAR-T cells. Some approaches have been approved for clinical use, such as anti-GD2 antibodies. Moreover, in terms of the antitumor mechanisms against different TACAs, we show results of selected clinical trials, considering the horizons that have opened up as a result of recent developments in technologies used for cancer control.
Collapse
Affiliation(s)
- Nora Berois
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay;
- Correspondence: (N.B.); (E.O.)
| | - Alvaro Pittini
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay;
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Eduardo Osinaga
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay;
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
- Correspondence: (N.B.); (E.O.)
| |
Collapse
|
21
|
Excretory-secretory product of Trichinella spiralis inhibits tumor cell growth by regulating the immune response and inducing apoptosis. Acta Trop 2022; 225:106172. [PMID: 34627760 DOI: 10.1016/j.actatropica.2021.106172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/24/2021] [Accepted: 09/27/2021] [Indexed: 12/30/2022]
Abstract
The excretory-secretory product (ESP) of Trichinella spiralis (T. spiralis) has been reported to inhibit the growth of various tumor cells, but the mechanism is not yet clear. To explore the effect and mechanism of ESP on liver cancer cells, tumor models were established with H22 cells and then infected with T. spiralis. The results showed that the growth of tumors in mice infected with T. spiralis was significantly inhibited. ESP from adult worms or muscle larvae were then incubated with H22 cells in vitro, and it was found that the ESP could inhibit cell proliferation and promote apoptosis. Subsequently, apoptosis-related proteins in stimulated H22 cells were evaluated, and ESP was found to induce cell apoptosis through the mitochondrial pathway. Additionally, Th-related cytokines were investigated in vivo, and the results showed that the levels of Th1 cytokines were significantly increased in the early stage of T. spiralis infection, while Th2 cytokines increased later than Th1 cytokines, implying that Th1 cytokines with antitumor effects may play a role in inhibiting tumor growth at early stage. In short, ESP can directly induce tumor cell apoptosis and indirectly inhibit tumor cell growth through the host immune system, which may be the antitumor mechanism of T. spiralis infection.
Collapse
|
22
|
Yue TT, Zhang N, Li JH, Lu XY, Wang XC, Li X, Zhang HB, Cheng SQ, Wang BB, Gong PT, Zhang XC. Anti-osteosarcoma effect of antiserum against cross antigen TPD52 between osteosarcoma and Trichinella spiralis. Parasit Vectors 2021; 14:498. [PMID: 34565443 PMCID: PMC8474799 DOI: 10.1186/s13071-021-05008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Trichinella spiralis (T. spiralis) is a parasite occurring worldwide that has been proven to have antitumour ability. However, studies on the antitumour effects of cross antigens between the tumour and T. spiralis or antibodies against cross antigens between tumours and T. spiralis are rare. METHODS To study the role of cross antigens between osteosarcoma and T. spiralis, we first screened the cDNA expression library of T. spiralis muscle larvae to obtain the cross antigen gene tumour protein D52 (TPD52), and prepared fusion protein TPD52 and its antiserum. The anti-osteosarcoma effect of the anti-TPD52 antiserum was studied using cell proliferation and cytotoxicity assays as well as in vivo animal models; preliminary data on the mechanism were obtained using western blot and immunohistochemistry analyses. RESULTS Our results indicated that TPD52 was mainly localized in the cytoplasm of MG-63 cells. Anti-TPD52 antiserum inhibited the proliferation of MG-63 cells and the growth of osteosarcoma in a dose-dependent manner. The tumour inhibition rate in the 100 μg treatment group was 61.95%. Enzyme-linked immunosorbent assay showed that injection of anti-TPD52 antiserum increased the serum levels of IFN-γ, TNF-α, and IL-12 in nude mice. Haematoxylin and eosin staining showed that anti-TPD52 antiserum did not cause significant pathological damage. Apoptosis of osteosarcoma cells was induced by anti-TPD52 antiserum in vivo and in vitro. CONCLUSIONS Anti-TPD52 antiserum exerts an anti-osteosarcoma effect by inducing apoptosis without causing histopathological damage.
Collapse
Affiliation(s)
- Tao-Tao Yue
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Nan Zhang
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Jian-Hua Li
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xiang-Yun Lu
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xiao-Cen Wang
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xin Li
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Hong-Bo Zhang
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Shu-Qin Cheng
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Bo-Bo Wang
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Peng-Tao Gong
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China.
| | - Xi-Chen Zhang
- Key Laboratory of Zoonosis Research By Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China.
| |
Collapse
|
23
|
Human hydatid cyst fluid-induced therapeutic anti-cancer immune responses via NK1.1 + cell activation in mice. Cancer Immunol Immunother 2021; 70:3617-3627. [PMID: 33944981 DOI: 10.1007/s00262-021-02948-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/19/2021] [Indexed: 01/21/2023]
Abstract
Echinococcus granulosus is a cestode parasite which causes cystic echinococcosis disease. Previously we observed that vaccination with E. granulosus antigens from human hydatid cyst fluid (HCF) significantly inhibits colon cancer growth. In the present work, we evaluate the anti-tumor immune response induced by human HCF against LL/2 lung cancer in mice. HCF vaccination protected from tumor growth, both in prophylactic and therapeutic settings, and significantly increased mouse survival compared to control mice. Considering that tumor-associated carbohydrate antigens are expressed in E. granulosus, we oxidized terminal carbohydrates in HCF with sodium periodate. This treatment abrogates the anti-tumor activity induced by HCF vaccination. We found that HCF vaccination-induced IgG antibodies that recognize LL/2 tumor cells by flow cytometry. An antigen-specific immune response is induced with HCF vaccination in the tumor-draining lymph nodes and spleen characterized by the production of IL-5 and, in less extent, IFNɣ. In the tumor microenvironment, we found that NK1.1 positive cells from HCF-treated mice showed higher expression of CD69 than control mice ones, indicating a higher level of activation. When we depleted these cells by administrating the NK-specific antibody NK1.1, a significantly decreased survival was observed in HCF-induced mice, suggesting that NK1.1+ cells mediate the anti-tumor protection induced by HCF. These results suggest that HCF can evoke an integrated anti-tumor immune response involving both, the innate and adaptive components, and provide novel insights into the understanding of the intricate relationship between HCF vaccination and tumor growth.
Collapse
|
24
|
Herrero de la Parte B, González-Arribas M, Diaz-Sanz I, Palomares T, García-Alonso I. Partial hepatectomy enhances the growth of CC531 rat colorectal cancer cells both in vitro and in vivo. Sci Rep 2021; 11:5356. [PMID: 33686132 PMCID: PMC7970880 DOI: 10.1038/s41598-021-85082-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Partial hepatectomy (PHx) is the gold standard for the treatment of colorectal cancer liver metastases. However, after removing a substantial amount of hepatic tissue, growth factors are released to induce liver regeneration, which may promote the proliferation of liver micrometastases or circulating tumour cells still present in the patient. The aim of this study is to assess the effect of PHx on the growth of liver metastases induced by intrasplenic cell inoculation as well as on in vitro proliferation of the same cancer cell line. Liver tumours were induced in 18 WAG/RijHsd male rats, by seeding 250,000 syngeneic colorectal cancer cells (CC531) into the spleen. The left lateral lobe of the liver was mobilized and in half of the animals it was removed to achieve a 40% hepatectomy. Twenty-eight days after tumour induction, the animals were sacrificed and the liver was removed and sliced to assess the relative tumour surface area (RTSA%). CC531 cells were cultured in presence of foetal calf serum, non-hepatectomised (NRS) or hepatectomized rat serum (HRS), and their proliferation rate at 24, 48, and 72 h was measured. RTSA% was significantly higher in animals which had undergone PHx than in the controls (non-hepatectomised) (46.98 ± 8.76% vs. 18.73 ± 5.65%; p < 0.05). Analysing each lobe separately, this difference in favour of hepatectomized animals was relevant and statistically significant in the paramedian and caudate lobes. But in the right lobe the difference was scarce and not significant. In vitro, 2.5% HRS achieved stronger proliferative rates than the control cultures (10% FCS) or their equivalent of NRS. In this experimental model, a parallelism has been shown between the effect of PHx on the growth of colorectal cancer cells in the liver and the effect of the serum on those cells in vitro.
Collapse
Affiliation(s)
- Borja Herrero de la Parte
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Vizcaya, Spain. .,Biocruces Bizkaia Health Research Institute, Plaza de Cruces s/n, 48903, Barakaldo, Spain.
| | - Mikel González-Arribas
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Vizcaya, Spain
| | - Iñaki Diaz-Sanz
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Vizcaya, Spain
| | - Teodoro Palomares
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Vizcaya, Spain
| | - Ignacio García-Alonso
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Vizcaya, Spain.,Biocruces Bizkaia Health Research Institute, Plaza de Cruces s/n, 48903, Barakaldo, Spain
| |
Collapse
|
25
|
Borges BC, Uehara IA, Dos Santos MA, Martins FA, de Souza FC, Junior ÁF, da Luz FAC, da Costa MS, Notário AFO, Lopes DS, Teixeira SC, Teixeira TL, de Castilhos P, da Silva CV, Silva MJB. The Recombinant Protein Based on Trypanosoma cruzi P21 Interacts With CXCR4 Receptor and Abrogates the Invasive Phenotype of Human Breast Cancer Cells. Front Cell Dev Biol 2020; 8:569729. [PMID: 33195200 PMCID: PMC7604327 DOI: 10.3389/fcell.2020.569729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/22/2020] [Indexed: 01/07/2023] Open
Abstract
Trypanosoma cruzi P21 is a protein secreted by the parasite that plays biological roles directly involved in the progression of Chagas disease. The recombinant protein (rP21) demonstrates biological properties, such as binding to CXCR4 receptors in macrophages, chemotactic activity of immune cells, and inhibiting angiogenesis. This study aimed to verify the effects of rP21 interaction with CXCR4 from non-tumoral cells (MCF-10A) and triple-negative breast cancer cells (MDA-MB-231). Our data showed that the MDA-MB-231 cells expressed higher levels of CXCR4 than did the non-tumor cell lines. Besides, cytotoxicity assays using different concentrations of rP21 showed that the recombinant protein was non-toxic and was able to bind to the cell membranes of both cell lineages. In addition, rP21 reduced the migration and invasion of MDA-MB-231 cells by the downregulation of MMP-9 gene expression. In addition, treatment with rP21 blocked the cell cycle, arresting it in the G1 phase, mainly in MDA-MB-231 cells. Finally, rP21 prevents the chemotaxis and proliferation induced by CXCL12. Our data showed that rP21 binds to the CXCR4 receptors in both cells, downregulates CXCR4 gene expression, and decreases the receptors in the cytoplasm of MDA-MB-231 cells, suggesting CXCR4 internalization. This internalization may explain the desensitization of the receptors in these cells. Thus, rP21 prevents migration, invasion, and progression in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Bruna Cristina Borges
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil.,Laboratório de Biomarcadores Tumorais e Osteoimunologia, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Isadora Akemi Uehara
- Laboratório de Biomarcadores Tumorais e Osteoimunologia, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Marlus Alves Dos Santos
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Flávia Alves Martins
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Fernanda Carvalho de Souza
- Laboratório de Biomarcadores Tumorais e Osteoimunologia, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Álvaro Ferreira Junior
- Departamento de MedicinaVeterinária, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, Brazil
| | - Felipe Andrés Cordero da Luz
- Laboratório de Biomarcadores Tumorais e Osteoimunologia, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Mylla Spirandelli da Costa
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Ana Flávia Oliveira Notário
- Laboratório de Nanobiotecnologia, Instituto de Genético e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Daiana Silva Lopes
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| | - Samuel Cota Teixeira
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Thaise Lara Teixeira
- Laboratório de Biologia Molecular de Trypanosoma Cruzi, Departamento de Parasitologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Patrícia de Castilhos
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Claudio Vieira da Silva
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Marcelo José Barbosa Silva
- Laboratório de Biomarcadores Tumorais e Osteoimunologia, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| |
Collapse
|
26
|
Sosoniuk-Roche E, Cruz P, Maldonado I, Duaso L, Pesce B, Michalak M, Valck C, Ferreira A. In vitro Treatment of a Murine Mammary Adenocarcinoma Cell Line with Recombinant Trypanosoma cruzi Calreticulin Promotes Immunogenicity and Phagocytosis. Mol Immunol 2020; 124:51-60. [PMID: 32526557 DOI: 10.1016/j.molimm.2020.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/16/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022]
Abstract
American Trypanosomiasis, a parasitic disease produced by Trypanosoma cruzi (T. cruzi), endemic in Latin America, infects about 6 million people. During the chronic stage of the infection, approximately 30% of infected people will develop Chagas Disease, the clinical manifestation. Few decades ago it was reported that, during the chronic stage, the parasite interferes with the development of solid tumors. However, the identification of parasite molecules responsible for such effects remained elusive. Years later, we described T.cruzi Calreticulin (TcCalr), an endoplasmic reticulum resident chaperone that infective trypomastigotes translocate to the parasite exterior, where it displays anticomplement activities. Most likely, at least some of these activities are related with the antitumor properties of TcCalr, as shown in in vitro, ex vivo, in ovum, and in vivo models. In this context we, we have seen that in vivo subcutaneous peritumoral inoculation of rTcCalr enhances local infiltration of T cells and slows tumor development. Based on these precedents, we propose that in vitro treatment of a mammary adenocarcinoma (TA3 cell line) with rTcCalr, will enhance tumor immunogenicity. In agreement with this proposal, we have shown that: i). rTcCalr binds to TA3 cells in a concentration-dependent fashion, ii). C1q binds to TA3 cells in an rTcCalr-dependent fashion, confirmed by the reversion attained using anti-TcS (a central TcCalr domain that binds C1) F(ab')2 antibody fragments, iii). incubation of TA3 cells with rTcCalr, promotes cell phagocytosis by murine macrophages and, iv). rTcCalr decreases the membrane expression of MHC class II, m-Dectin-1, Galectin-9 and PD-L1, while increasing the expression of Rae-1γ. In synthesis, herein we show that in vitro treatment of a murine mammary adenocarcinoma with rTcCalr enhances phagocytosis and modulates the expression of a variety of membrane molecules that correlates with increased tumor immunogenicity.
Collapse
Affiliation(s)
- Eduardo Sosoniuk-Roche
- Immunology of Microbial Aggressions, Immunology Disciplinary Program, Biomedical Science Institute, Faculty of Medicine, Universidad de Chile, Chile
| | - Pamela Cruz
- Immunology of Microbial Aggressions, Immunology Disciplinary Program, Biomedical Science Institute, Faculty of Medicine, Universidad de Chile, Chile
| | - Ismael Maldonado
- Immunology of Microbial Aggressions, Immunology Disciplinary Program, Biomedical Science Institute, Faculty of Medicine, Universidad de Chile, Chile
| | - Leonora Duaso
- Immunology of Microbial Aggressions, Immunology Disciplinary Program, Biomedical Science Institute, Faculty of Medicine, Universidad de Chile, Chile
| | - Bárbara Pesce
- MED.UCHILE-FACS Laboratory, Biomedical Science Institute, Faculty of Medicine, Universidad de Chile, Chile
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Alberta, Canada
| | - Carolina Valck
- Immune Response Modulation by the Complement System, Immunology Disciplinary Program, Biomedical Science Institute, Faculty of Medicine, Universidad de Chile, Chile.
| | - Arturo Ferreira
- Immunology of Microbial Aggressions, Immunology Disciplinary Program, Biomedical Science Institute, Faculty of Medicine, Universidad de Chile, Chile.
| |
Collapse
|
27
|
Ramírez-Toloza G, Sosoniuk-Roche E, Valck C, Aguilar-Guzmán L, Ferreira VP, Ferreira A. Trypanosoma cruzi Calreticulin: Immune Evasion, Infectivity, and Tumorigenesis. Trends Parasitol 2020; 36:368-381. [PMID: 32191851 DOI: 10.1016/j.pt.2020.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/25/2020] [Accepted: 01/26/2020] [Indexed: 02/06/2023]
Abstract
To successfully infect, Trypanosoma cruzi evades and modulates the host immune response. T. cruzi calreticulin (TcCalr) is a multifunctional, endoplasmic reticulum (ER)-resident chaperone that, translocated to the external microenvironment, mediates crucial host-parasite interactions. TcCalr binds and inactivates C1 and mannose-binding lectin (MBL)/ficolins, important pattern- recognition receptors (PRRs) of the complement system. Using an apoptotic mimicry strategy, the C1-TcCalr association facilitates the infection of target cells. T. cruzi infection also seems to confer protection against tumorigenesis. Thus, recombinant TcCalr has important antiangiogenic properties, detected in vitro, ex vivo, and in ovum, most likely contributing at least in part, to its antitumor properties. Consequently, TcCalr is useful for investigating key issues of host-parasite interactions and possible new immunological/pharmacological interventions in the areas of Chagas' disease and experimental cancer.
Collapse
Affiliation(s)
- Galia Ramírez-Toloza
- Faculty of Veterinary Medicine and Livestock Sciences, University of Chile, Santiago, Chile.
| | | | - Carolina Valck
- Department of Immunology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Lorena Aguilar-Guzmán
- Faculty of Veterinary Medicine and Livestock Sciences, University of Chile, Santiago, Chile
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, OH, USA
| | - Arturo Ferreira
- Department of Immunology, Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
28
|
Cruz P, Sosoniuk-Roche E, Maldonado I, Torres CG, Ferreira A. Trypanosoma cruzi calreticulin: In vitro modulation of key immunogenic markers of both canine tumors and relevant immune competent cells. Immunobiology 2019; 225:151892. [PMID: 31837774 DOI: 10.1016/j.imbio.2019.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/01/2019] [Accepted: 12/01/2019] [Indexed: 10/25/2022]
Abstract
Recombinant calreticulin from Trypanosoma cruzi (rTcCalr), the parasite responsible for Chagas' disease, binds to Canine Transmissible Venereal Tumor (CTVT) cells from primary cultures and to a canine mammary carcinoma cell line. A Complement-binding assay indicated that interaction of the first component C1q with these tumor cells operated independently of the rTcCalr-presence. This apparent independence could be explained by the important structural similarities that exist among rTcCarl, endogenous normal canine and/or mutated calreticulins present in several types of cancer. In phagocytosis assays, tumor cells treated with rTcCalr were readily engulfed by macrophages and, co-cultured with DCs, accelerated their maturation. In addition, DCs maturation, induced by tumor cells co-cultured with rTcCalr, activated T cells more efficiently than DCs, treated or not with LPS. In an apparent paradox, a decrease in MHC Class I expression was observed when these tumor cells were co-cultivated with rTcCalr. This decrease may be related to a down regulation signaling promoting the rescue of MHC I. Possibly, these in vitro assays may be valid correlates of in vivo sceneries. Based on these results, we propose that rTcCalr improves in vitro the immunogenicity of two widely different tumor cell lines, thus suggesting that the interesting properties of rTcCalr to boost immune responses warrant future studies.
Collapse
Affiliation(s)
- P Cruz
- Laboratory of Immunology of Microbial Aggression, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, 8380453, Chile; Laboratory of Biomedicine and Regenerative Medicine, Department of Clinical Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santiago, 8820808, Chile
| | - E Sosoniuk-Roche
- Laboratory of Immunology of Microbial Aggression, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, 8380453, Chile
| | - I Maldonado
- Laboratory of Immunology of Microbial Aggression, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, 8380453, Chile
| | - C G Torres
- Laboratory of Biomedicine and Regenerative Medicine, Department of Clinical Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santiago, 8820808, Chile.
| | - A Ferreira
- Laboratory of Immunology of Microbial Aggression, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, 8380453, Chile.
| |
Collapse
|
29
|
Structural bases that underline Trypanosoma cruzi calreticulin proinfective, antiangiogenic and antitumor properties. Immunobiology 2019; 225:151863. [PMID: 31732192 DOI: 10.1016/j.imbio.2019.10.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 10/29/2019] [Indexed: 12/24/2022]
Abstract
Microbes have developed mechanisms to resist the host immune defenses and some elicit antitumor immune responses. About 6 million people are infected with Trypanosoma cruzi, the protozoan agent of Chagas' disease, the sixth neglected tropical disease worldwide. Eighty years ago, G. Roskin and N. Klyuyeva proposed that T. cruzi infection mediates an anti-cancer activity. This observation has been reproduced by several other laboratories, but no molecular basis has been proposed. We have shown that the highly pleiotropic chaperone calreticulin (TcCalr, formerly known as TcCRT), translocates from the parasite ER to the exterior, where it mediates infection. Similar to its human counterpart HuCALR (formerly known as HuCRT), TcCalr inhibits C1 in its capacity to initiate the classical pathway of complement activation. We have also proposed that TcCalr inhibits angiogenesis and it is a likely mediator of antitumor effects. We have generated several in silico structural TcCalr models to delimit a peptide (VC-TcCalr) at the TcCalr N-domain. Chemically synthesized VC-TcCalr did bind to C1q and was anti-angiogenic in Gallus gallus chorioallantoic membrane assays. These properties were associated with structural features, as determined in silico. VC-TcCalr, a strong dipole, interacts with charged proteins such as collagen-like tails and scavenger receptors. Comparatively, HuCALR has less polarity and spatial stability, probably due to at least substitutions of Gln for Gly, Arg for Lys, Arg for Asp and Ser for Arg that hinder protein-protein interactions. These differences can explain, at least in part, how TcCalr inhibits the complement activation pathway and has higher efficiency as an antiangiogenic and antitumor agent than HuCALR.
Collapse
|
30
|
Daneshpour S, Rostamirad S, Kefayat A, Mofid M, Safavi A, Darani HY. Identifying the Most Effective Hydatid Cyst Fluid Fraction for Anticancer Vaccination of 4T1 Breast Tumor-Bearing Mice. Int J Prev Med 2019; 10:143. [PMID: 31516684 PMCID: PMC6716223 DOI: 10.4103/ijpvm.ijpvm_508_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/21/2019] [Indexed: 11/22/2022] Open
Abstract
Background: The hydatid cyst fluid antigens have high homology with cancer cell antigens and also exhibit considerable immunogenicity. Therefore, their utilization for cancer immunization can cause an effective antitumor immune response. However, the main challenge is identifying the most effective antigens for this purpose. Methods: Hydatid cyst fluid fractions including the glycolipid fraction, glycoprotein fraction, 78 kDa fraction, and antigen B fraction were prepared. Then, the BALB/c mice were immunized against different antigens and, subsequently, 4T1 cells were subcutaneously implanted. The tumors' growth, metastasis, and tumor-bearing mice survival were assessed in different immunized groups. In addition, IL-2, IL-4, IFN-γ, and TNF-α serum levels were estimated to evaluate the immune system response. Results: BALB/c mice immunization against the complete hydatid cyst fluid antigens exhibited more significant inhibition of the tumors' growth and metastasis and increase of tumor-bearing mice survival in comparison with its derived fractions. However, the 78 kDa fraction exhibited the best results according to the same factors in comparison with all the prepared fractions. Conclusions: The 78 kDa fraction of the hydatid cyst fluid was the most effective fraction of hydatid cyst fluid for immunization against 4T1 breast tumors.
Collapse
Affiliation(s)
- Shima Daneshpour
- Department of Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahla Rostamirad
- Clinical Research Development Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Amirhosein Kefayat
- Cancer Prevention Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - MohammadReza Mofid
- Department of Biochemistry, Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ashkan Safavi
- Cancer Prevention Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Yousofi Darani
- Department of Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
31
|
Mohamadi F, Shakibapour M, Sharafi SM, Reza AA, Tolouei S, Darani HY. Anti- Toxoplasma gondii antibodies attach to mouse cancer cell lines but not normal mouse lymphocytes. Biomed Rep 2019; 10:183-188. [PMID: 30906547 DOI: 10.3892/br.2019.1186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 01/04/2019] [Indexed: 01/03/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is prevalent intracellular parasite and a cause of worldwide infection in the human population. An inhibitory effect of this parasite on cancer growth has been demonstrated in cell culture and animal models. To determine whether the anticancer activities of T. gondii are associated with host immune response, in the current study the reactivity of anti-T. gondii antiserum with the surface of cancer cell lines was investigated. Anti-T. gondii antibodies were raised in rabbit and the reaction of this antiserum in comparison with other anti-parasite antisera (anti-T. vaginalis, anti-hydatid cyst fluid, anti-protoscolices antigens) with mouse melanoma or breast cancer cells lines was investigated using flow cytometry. Anti-T. gondii antiserum reacted markedly with the surface of mouse melanoma and breast cancer cells, and less so with the normal mouse spleen lymphocytes. Meanwhile, the other anti-parasite antisera did not react strongly with the surface of cancer cells compared with normal mouse spleen lymphocytes. In summary, it has been demonstrated herein that anti-T. gondii antiserum may selectively react with the surface of mouse cancer cells but not with normal mouse spleen lymphocytes. Therefore, further study on anti-Toxoplasma antibodies may be useful for directing the application of selective drug delivery in cancer treatment.
Collapse
Affiliation(s)
- Fereshteh Mohamadi
- Department of Medical Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| | - Mahshid Shakibapour
- Department of Medical Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| | - Seyedeh Maryam Sharafi
- Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| | - Ali Andalib Reza
- Department of Medical Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| | - Sepideh Tolouei
- Department of Medical Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| | - Hossein Yousofi Darani
- Department of Medical Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran.,Cancer Prevention Research Centre, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| |
Collapse
|
32
|
Callejas BE, Martínez-Saucedo D, Terrazas LI. Parasites as negative regulators of cancer. Biosci Rep 2018; 38:BSR20180935. [PMID: 30266743 PMCID: PMC6200699 DOI: 10.1042/bsr20180935] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 12/16/2022] Open
Abstract
Several environmental factors (chemical, physical, and biological) can cause the initiation, promotion, and progression of cancer. Regarding the biological factors, several studies have found that infections caused by some bacteria, viruses and protozoan, and helminth parasites are related to carcinogenesis. However, in recent years a different approach has been implemented on the antitumor impact of parasitic diseases caused by some protozoan and helminths, mainly because such infections may affect several hallmarks of cancer, but the involved mechanisms still remain unknown. The beneficial effects reported for some parasitic diseases on tumorigenesis range from the induction of apoptosis, activation of the immune response, avoiding metastasis and angiogenesis, inhibition of proliferative signals, to the regulation of inflammatory responses that promote cancer. In this work, we reviewed the available information regarding how parasitic infections may modulate cancer progression. Despite the fact that specific mechanisms of action on tumors are not yet totally clear, we consider that detailed studies of the antitumor action of these organisms and their products could lead to the discovery and use of new molecules from these biological agents that may work as adjuvant therapy in the treatment of various types of cancer.
Collapse
Affiliation(s)
- Blanca E Callejas
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, 54090, México
| | - Diana Martínez-Saucedo
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, 54090, México
| | - Luis I Terrazas
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, 54090, México
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, 54090, México
| |
Collapse
|
33
|
Immuno-therapeutic potential of Schistosoma mansoni and Trichinella spiralis antigens in a murine model of colon cancer. Invest New Drugs 2018; 37:47-56. [PMID: 29808307 DOI: 10.1007/s10637-018-0609-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023]
Abstract
Considerable evidence indicates a negative correlation between the prevalence of some parasitic infections and cancer and their interference with tumor growth. Therefore, parasitic antigens seem to be promising candidates for cancer immunotherapy. In this study, the therapeutic efficacy of autoclaved Schistosoma mansoni and Trichinella spiralis antigens against a colon cancer murine model was investigated. Both antigens showed immunomodulatory potential, as evidenced by a significant decrease in serum IL-17, a significant increase in serum IL-10, and the percentage of splenic CD4+T-cells and intestinal FoxP3+ Treg cells. However, treatment with S. mansoni antigen yielded protection against the deleterious effect of DMH-induced colon carcinogenesis only, with a significant decrease in the average lesion size and number of neoplasias per mouse. For the first time, we report an inhibitory effect of S. mansoni antigen on the progression of chemically induced colon carcinogenesis, but the exact mechanism has yet to be clarified. This anti-tumor strategy could introduce a new era of medicine in which a generation of anticancer vaccines of parasitic origin would boost the therapy for incurable cancers.
Collapse
|
34
|
Study on the mitochondrial apoptosis pathways of small cell lung cancer H446 cells induced by Trichinella spiralis muscle larvae ESPs. Parasitology 2017; 144:793-800. [PMID: 28073393 DOI: 10.1017/s0031182016002535] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Trichinella spiralis (T.spiralis) muscle-larva (ML) excretory-secretory proteins (ESPs) contain antitumour-active substances. ESPs have been shown to inhibit tumour growth. To explore the effects of these proteins on small cell lung cancer cells and the possible mechanisms of their antineoplastic action, H446 SCLC cells were co-cultured with different concentrations of T. spiralis ML ESPs for 12, 24 and 48 h. Our results showed that T. spiralis ML ESPs significantly inhibited H446 cell proliferation, which was dose-and time-dependent. The results of flow cytometry testing indicate a clear apoptosis trend in H446 cells co-cultured with ESPs for 24 h. Reverse transcription polymerase chain reaction and Western blotting results showed increased expression of pro-apoptosis genes Bax, Cyt-C, Apaf-1, caspase-9 and caspase-3, compared with the negative control group, and decreased the expression of anti-apoptosis genes Bcl-2 and Livin. Our results suggest that T. spiralis ML ESPs can induce apoptosis in H446 cells through a mitochondrial pathway, which may be a mechanism of antineoplastic action in T. spiralis ML ESPs.
Collapse
|
35
|
van Tong H, Brindley PJ, Meyer CG, Velavan TP. Parasite Infection, Carcinogenesis and Human Malignancy. EBioMedicine 2016; 15:12-23. [PMID: 27956028 PMCID: PMC5233816 DOI: 10.1016/j.ebiom.2016.11.034] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 11/24/2016] [Accepted: 11/29/2016] [Indexed: 12/14/2022] Open
Abstract
Cancer may be induced by many environmental and physiological conditions. Infections with viruses, bacteria and parasites have been recognized for years to be associated with human carcinogenicity. Here we review current concepts of carcinogenicity and its associations with parasitic infections. The helminth diseases schistosomiasis, opisthorchiasis, and clonorchiasis are highly carcinogenic while the protozoan Trypanosoma cruzi, the causing agent of Chagas disease, has a dual role in the development of cancer, including both carcinogenic and anticancer properties. Although malaria per se does not appear to be causative in carcinogenesis, it is strongly associated with the occurrence of endemic Burkitt lymphoma in areas holoendemic for malaria. The initiation of Plasmodium falciparum related endemic Burkitt lymphoma requires additional transforming events induced by the Epstein-Barr virus. Observations suggest that Strongyloides stercoralis may be a relevant co-factor in HTLV-1-related T cell lymphomas. This review provides an overview of the mechanisms of parasitic infection-induced carcinogenicity. The helminth diseases schistosomiasis, opisthorchiasis, and clonorchiasis are highly carcinogenic. Trypanosoma cruzi has a dual role in cancer development including both carcinogenic and anticancer properties. Initiation of Plasmodium falciparum related endemic Burkitt lymphoma requires additional transforming events induced by EBV. Strongyloides stercoralis may be a relevant co-factor in HTLV-1-related T cell lymphomas.
We searched MEDLINE database and PubMed for articles from 1970 through June 30, 2016. Search terms used in various combinations were “parasite infection”, “carcinogenesis”, “cancer”, “human malignancy”, “parasite and cancer”, “infection-associated cancer”, “parasite-associated cancer” “schistosomiasis”, “opisthorchiasis”, “malaria”, “Chagas disease”, and “strongyloidiasis”. Articles resulting from these searches and relevant references cited in those articles were selected based on their related topics and were reviewed. Abstracts and reports from meetings were also included. Articles published in English were included.
Collapse
Affiliation(s)
- Hoang van Tong
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; Biomedical and Pharmaceutical Applied Research Center, Vietnam Military Medical University, Hanoi, Vietnam.
| | - Paul J Brindley
- Research Center for Neglected Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, D.C., USA
| | - Christian G Meyer
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; Health Focus GmbH, Potsdam, Germany; Duy Tan University, Da Nang, Viet Nam; Vietnamese - German Centre for Medical Research (VG-CARE), Hanoi, Viet Nam
| | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; Duy Tan University, Da Nang, Viet Nam; Vietnamese - German Centre for Medical Research (VG-CARE), Hanoi, Viet Nam.
| |
Collapse
|
36
|
Osinaga E, Freire T, Ubillos L. Author's reply to: Could cross-immunological reactivity to Trypanosoma cruziantigens be considered a rational strategy for designing vaccines against cancer? Int J Cancer 2016; 139:2144. [DOI: 10.1002/ijc.30246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 06/07/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Eduardo Osinaga
- Departamento de Inmunobiología; Facultad De Medicina, Universidad de la, República; Montevideo Uruguay
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de; Montevideo Uruguay
| | - Teresa Freire
- Departamento de Inmunobiología; Facultad De Medicina, Universidad de la, República; Montevideo Uruguay
| | - Luis Ubillos
- Departamento de Inmunobiología; Facultad De Medicina, Universidad de la, República; Montevideo Uruguay
- Servicio de Oncología Clínica, Hospital de Clínicas, Facultad De Medicina, Universidad de la República; Montevideo Uruguay
| |
Collapse
|
37
|
Novaes RD, Gonçalves RV, Caldas IS. Could cross-immunological reactivity to Trypanosoma cruzi antigens be considered a rational strategy for designing vaccines against cancer? Int J Cancer 2016; 139:2142-3. [PMID: 27352084 DOI: 10.1002/ijc.30247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/07/2016] [Indexed: 11/05/2022]
Affiliation(s)
- Rômulo Dias Novaes
- Department of Structural Biology, Federal University of Alfenas, Alfenas, MG, 37130-000, Brazil
| | | | - Ivo Santana Caldas
- Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, MG, 37130-000, Brazil
| |
Collapse
|
38
|
Puzzling and ambivalent roles of malarial infections in cancer development and progression. Parasitology 2016; 143:1811-1823. [DOI: 10.1017/s0031182016001591] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SUMMARYScientific evidence strongly suggests that parasites are directly or indirectly associated with carcinogenesis in humans. However, studies have also indicated that parasites or their products might confer resistance to tumour growth.Plasmodiumprotozoa, the causative agents of malaria, exemplify the ambivalent link between parasites and cancer. Positive relationships between malaria and virus-associated cancers are relatively well-documented; for example, malaria can reactivate the Epstein-Barr Virus, which is the known cause of endemic Burkitt lymphoma. Nevertheless, possible anti-tumour properties of malaria have also been reported and, interestingly, this disease has long been thought to be beneficial to patients suffering from cancers. Current knowledge of the potential pro- and anti-cancer roles of malaria suggests that, contrary to other eukaryotic parasites affecting humans,Plasmodium-related cancers are principally lymphoproliferative disorders and attributable to virus reactivation, whereas, similar to other eukaryotic parasites, the anti-tumour effects of malaria are primarily associated with carcinomas and certain sarcomas. Moreover, malarial infection significantly suppresses murine cancer growth by inducing both innate and specific adaptive anti-tumour responses. This review aims to present an update regarding the ambivalent association between malaria and cancer, and further studies may open future pathways to develop novel strategies for anti-cancer therapies.
Collapse
|
39
|
Abello-Cáceres P, Pizarro-Bauerle J, Rosas C, Maldonado I, Aguilar-Guzmán L, González C, Ramírez G, Ferreira J, Ferreira A. Does native Trypanosoma cruzi calreticulin mediate growth inhibition of a mammary tumor during infection? BMC Cancer 2016; 16:731. [PMID: 27619675 PMCID: PMC5020520 DOI: 10.1186/s12885-016-2764-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/27/2016] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND For several decades now an antagonism between Trypanosoma cruzi infection and tumor development has been detected. The molecular basis of this phenomenon remained basically unknown until our proposal that T. cruzi Calreticulin (TcCRT), an endoplasmic reticulum-resident chaperone, translocated-externalized by the parasite, may mediate at least an important part of this effect. Thus, recombinant TcCRT (rTcCRT) has important in vivo antiangiogenic and antitumor activities. However, the relevant question whether the in vivo antitumor effect of T. cruzi infection is indeed mediated by the native chaperone (nTcCRT), remains open. Herein, by using specific modified anti-rTcCRT antibodies (Abs), we have neutralized the antitumor activity of T. cruzi infection and extracts thereof, thus identifying nTcCRT as a valid mediator of this effect. METHODS Polyclonal anti-rTcCRT F(ab')2 Ab fragments were used to reverse the capacity of rTcCRT to inhibit EAhy926 endothelial cell (EC) proliferation, as detected by BrdU uptake. Using these F(ab')2 fragments, we also challenged the capacity of nTcCRT, during T. cruzi infection, to inhibit the growth of an aggressive mammary adenocarcinoma cell line (TA3-MTXR) in mice. Moreover, we determined the capacity of anti-rTcCRT Abs to reverse the antitumor effect of an epimastigote extract (EE). Finally, the effects of these treatments on tumor histology were evaluated. RESULTS The rTcCRT capacity to inhibit ECs proliferation was reversed by anti-rTcCRT F(ab')2 Ab fragments, thus defining them as valid probes to interfere in vivo with this important TcCRT function. Consequently, during infection, these Ab fragments also reversed the in vivo experimental mammary tumor growth. Moreover, anti-rTcCRT Abs also neutralized the antitumor effect of an EE, again identifying the chaperone protein as an important mediator of this anti mammary tumor effect. Finally, as determined by conventional histological parameters, in infected animals and in those treated with EE, less invasive tumors were observed while, as expected, treatment with F(ab')2 Ab fragments increased malignancy. CONCLUSION We have identified translocated/externalized nTcCRT as responsible for at least an important part of the anti mammary tumor effect of the chaperone observed during experimental infections with T. cruzi.
Collapse
Affiliation(s)
- Paula Abello-Cáceres
- Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Avenida Independencia 1027, Independencia, Santiago, Chile
| | - Javier Pizarro-Bauerle
- Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Avenida Independencia 1027, Independencia, Santiago, Chile
| | - Carlos Rosas
- Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Avenida Independencia 1027, Independencia, Santiago, Chile
| | - Ismael Maldonado
- Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Avenida Independencia 1027, Independencia, Santiago, Chile
| | - Lorena Aguilar-Guzmán
- Faculty of Veterinary Medicine and Livestock Sciences, University of Chile, Avenida Santa Rosa 11735, La Pintana, Santiago, Chile
| | - Carlos González
- Faculty of Veterinary Medicine, Andrés Bello University, Avenida República 440, Santiago Centro, Santiago, Chile
| | - Galia Ramírez
- Faculty of Veterinary Medicine and Livestock Sciences, University of Chile, Avenida Santa Rosa 11735, La Pintana, Santiago, Chile
| | - Jorge Ferreira
- Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Avenida Independencia 1027, Independencia, Santiago, Chile
| | - Arturo Ferreira
- Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Avenida Independencia 1027, Independencia, Santiago, Chile
- University of Chile, Avenida Independencia 1027, Santiago, Chile
| |
Collapse
|
40
|
Zheng J, Zhou Y, Li Y, Xu DP, Li S, Li HB. Spices for Prevention and Treatment of Cancers. Nutrients 2016; 8:E495. [PMID: 27529277 PMCID: PMC4997408 DOI: 10.3390/nu8080495] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/22/2016] [Accepted: 08/05/2016] [Indexed: 12/14/2022] Open
Abstract
Spices have been widely used as food flavorings and folk medicines for thousands of years. Numerous studies have documented the antioxidant, anti-inflammatory and immunomodulatory effects of spices, which might be related to prevention and treatment of several cancers, including lung, liver, breast, stomach, colorectum, cervix, and prostate cancers. Several spices are potential sources for prevention and treatment of cancers, such as Curcuma longa (tumeric), Nigella sativa (black cumin), Zingiber officinale (ginger), Allium sativum (garlic), Crocus sativus (saffron), Piper nigrum (black pepper) and Capsicum annum (chili pepper), which contained several important bioactive compounds, such as curcumin, thymoquinone, piperine and capsaicin. The main mechanisms of action include inducing apoptosis, inhibiting proliferation, migration and invasion of tumors, and sensitizing tumors to radiotherapy and chemotherapy. This review summarized recent studies on some spices for prevention and treatment of cancers, and special attention was paid to bioactive components and mechanisms of action.
Collapse
Affiliation(s)
- Jie Zheng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Yue Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Ya Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Dong-Ping Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Sha Li
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
- South China Sea Bioresource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-Sen University, Guangzhou 510006, China.
| |
Collapse
|
41
|
Ramírez-Toloza G, Abello P, Ferreira A. Is the Antitumor Property of Trypanosoma cruzi Infection Mediated by Its Calreticulin? Front Immunol 2016; 7:268. [PMID: 27462315 PMCID: PMC4939398 DOI: 10.3389/fimmu.2016.00268] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 06/27/2016] [Indexed: 12/31/2022] Open
Abstract
Eight to 10 million people in 21 endemic countries are infected with Trypanosoma cruzi. However, only 30% of those infected develop symptoms of Chagas' disease, a chronic, neglected tropical disease worldwide. Similar to other pathogens, T. cruzi has evolved to resist the host immune response. Studies, performed 80 years ago in the Soviet Union, proposed that T. cruzi infects tumor cells with similar capacity to that displayed for target tissues such as cardiac, aortic, or digestive. An antagonistic relationship between T. cruzi infection and cancer development was also proposed, but the molecular mechanisms involved have remained largely unknown. Probably, a variety of T. cruzi molecules is involved. This review focuses on how T. cruzi calreticulin (TcCRT), exteriorized from the endoplasmic reticulum, targets the first classical complement component C1 and negatively regulates the classical complement activation cascade, promoting parasite infectivity. We propose that this C1-dependent TcCRT-mediated virulence is critical to explain, at least an important part, of the parasite capacity to inhibit tumor development. We will discuss how TcCRT, by directly interacting with venous and arterial endothelial cells, inhibits angiogenesis and tumor growth. Thus, these TcCRT functions not only illustrate T. cruzi interactions with the host immune defensive strategies, but also illustrate a possible co-evolutionary adaptation to privilege a prolonged interaction with its host.
Collapse
Affiliation(s)
- Galia Ramírez-Toloza
- Faculty of Veterinary Medicine and Livestock Sciences, University of Chile , Santiago , Chile
| | - Paula Abello
- Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences (ICBM), University of Chile , Santiago , Chile
| | - Arturo Ferreira
- Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences (ICBM), University of Chile , Santiago , Chile
| |
Collapse
|