1
|
Rajković M, Deksne G, Živković L, Leonova E, Spremo-Potparević B, Sjakste N. DNA damage induced by parasitic infections in humans and animals. Comp Immunol Microbiol Infect Dis 2025; 119:102337. [PMID: 40220655 DOI: 10.1016/j.cimid.2025.102337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Pathologies caused by parasitic infections, including protozoans and helminths remain a burden for healthcare in many countries. The DNA damage is produced by numerous parasites, both protozoans and helminths. However, the exact number of cancer-causing parasites and their role in neoplasma formation is still undetermined. The progression and dynamics of parasitic infections are significantly influenced by endogenously induced increase in oxidative stress (OS). Increased ROS production undermines antioxidant defense mechanisms by disrupting the balance between prooxidants and antioxidants, causing structural damage to important biomolecules, including host DNA. The generation of DNA damage possibly leads to the progression of carcinogenesis. However, direct DNA damage by parasites, eggs and factors released by parasites is also possible, and it leads to genomic instability that is a hallmark of most human and animal cancers. Understanding the way parasites induce DNA damage in the hosts may be helpful in the control of parasitic infections and the prevention of parasite-induced malignancies, ultimately benefiting the health of humans and animals. This review article offers an updated overview of parasitic infection-induced DNA damage mechanisms.
Collapse
Affiliation(s)
- Milan Rajković
- Department of Parasitology, Faculty of Veterinary Medicine, University of Belgrade, Bulevar oslobodjenja 18, Belgrade 11000, Serbia.
| | - Gunita Deksne
- Department of Ecology, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas Street 1, LV1004, Latvia; Laboratory of Microbiology and Pathology, Parasitology group, Institute of Food Safety, Animal Health and Environment "BIOR", Lejupes street 3, Riga LV1076, Latvia.
| | - Lada Živković
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade 11000, Serbia
| | - Elina Leonova
- Department of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas Street 3, Riga LV1004, Latvia
| | - Biljana Spremo-Potparević
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade 11000, Serbia
| | - Nikolajs Sjakste
- Department of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas Street 3, Riga LV1004, Latvia.
| |
Collapse
|
2
|
Jain S. Does Schistosoma mansoni trigger colorectal cancer? Mol Biochem Parasitol 2025; 262:111672. [PMID: 39894059 DOI: 10.1016/j.molbiopara.2025.111672] [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: 04/10/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
In this work the relationship between Schistosoma mansoni (Sm) and the induction and progression of colorectal cancer (CRC) is examined. Various clinical studies reviewed here yield inconsistent results, with some reporting no association between Sm infection and CRC and others suggesting a probable to strong association. Here we propose a number of plausible mechanisms whereby Sm infection might contribute to CRC induction and/or progression. These factors are (1) chronic inflammation, (2) exposure to parasite linked antigens and genotoxic products, especially soluble egg antigens (SEAs) and (3) alteration of the intestinal microbiota. These factors probably predispose humans towards CRC and can help in CRC progression however only widespread epidemiological, clinical and pathological studies can firmly establish their role or a complete lack of it.
Collapse
Affiliation(s)
- Sidhant Jain
- Institute for Globally Distributed Open Research and Education (IGDORE), India.
| |
Collapse
|
3
|
Müller H, Straßmann JK, Baier AS, von Bülow V, Stettler F, Hagen MJ, Schmidt FP, Tschuschner A, Schmid AR, Zahner D, Köhler K, Pons-Kühnemann J, Leufkens D, Glebe D, Kaur S, Möscheid MF, Haeberlein S, Grevelding CG, Weiskirchen R, El-Kassas M, Zalata K, Roeb E, Roderfeld M. Liver Fibrosis Is Enhanced by a Higher Egg Burden in Younger Mice Infected with S. mansoni. Cells 2024; 13:1643. [PMID: 39404406 PMCID: PMC11475498 DOI: 10.3390/cells13191643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Schistosomiasis affects over 250 million people worldwide, with the highest prevalence at the age of 10-14 years. The influence of the host's age on the severity of liver damage is unclear. We infected male 8, 14, and 20-week-old mice with S. mansoni. Hepatic damage, inflammation, fibrosis, and metabolism were analyzed by RT-qPCR, Western blotting, ELISA, immunohistochemistry, and mechanistic transwell chamber experiments using S. mansoni eggs and human hepatic stellate cells (HSCs) or primary mouse hepatocytes. Major results were validated in human biopsies. We found that hepatosplenomegaly, granuloma size, egg load, inflammation, fibrosis, and glycogen stores all improved with the increasing age of the host. However, serum alanine transaminase (ALT) levels were lowest in young mice infected with S. mansoni. Hepatic carbohydrate exploitation was characterized by a shift towards Warburg-like glycolysis in S. mansoni-infected animals. Notably, S. mansoni eggs stimulated hepatic stellate cells to an alternatively activated phenotype (GFAP+/desmin+/αSMA-) that secretes IL-6 and MCP-1. The reduction of fibrosis in older age likely depends on the fine-tuning of regulatory and inflammatory cytokines, alternative HSC activation, and the age-dependent preservation of hepatic energy stores. The current results emphasize the significance of investigations on the clinical relevance of host age-dependent liver damage in patients with schistosomiasis.
Collapse
Affiliation(s)
- Heike Müller
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (H.M.); (J.K.S.); (A.S.B.); (V.v.B.); (F.S.); (M.J.H.); (F.P.S.); (A.T.); (E.R.)
| | - Jan K. Straßmann
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (H.M.); (J.K.S.); (A.S.B.); (V.v.B.); (F.S.); (M.J.H.); (F.P.S.); (A.T.); (E.R.)
| | - Anne S. Baier
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (H.M.); (J.K.S.); (A.S.B.); (V.v.B.); (F.S.); (M.J.H.); (F.P.S.); (A.T.); (E.R.)
| | - Verena von Bülow
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (H.M.); (J.K.S.); (A.S.B.); (V.v.B.); (F.S.); (M.J.H.); (F.P.S.); (A.T.); (E.R.)
| | - Frederik Stettler
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (H.M.); (J.K.S.); (A.S.B.); (V.v.B.); (F.S.); (M.J.H.); (F.P.S.); (A.T.); (E.R.)
| | - Maximilian J. Hagen
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (H.M.); (J.K.S.); (A.S.B.); (V.v.B.); (F.S.); (M.J.H.); (F.P.S.); (A.T.); (E.R.)
| | - Fabian P. Schmidt
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (H.M.); (J.K.S.); (A.S.B.); (V.v.B.); (F.S.); (M.J.H.); (F.P.S.); (A.T.); (E.R.)
| | - Annette Tschuschner
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (H.M.); (J.K.S.); (A.S.B.); (V.v.B.); (F.S.); (M.J.H.); (F.P.S.); (A.T.); (E.R.)
| | - Andreas R. Schmid
- Department of Internal Medicine III, Justus Liebig University, 35392 Giessen, Germany;
| | - Daniel Zahner
- Central Laboratory Animal Facility, Justus Liebig University, 35392 Giessen, Germany;
| | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University, 35392, Germany;
| | - Jörn Pons-Kühnemann
- Institute of Medical Informatics, Justus Liebig University, 35392, Germany; (J.P.-K.); (D.L.)
| | - Daniel Leufkens
- Institute of Medical Informatics, Justus Liebig University, 35392, Germany; (J.P.-K.); (D.L.)
| | - Dieter Glebe
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, German Center for Infection Research (DZIF; Partner Site Giessen-Marburg-Langen), Justus Liebig University, 35392 Giessen, Germany; (D.G.); (S.K.)
| | - Surmeet Kaur
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, German Center for Infection Research (DZIF; Partner Site Giessen-Marburg-Langen), Justus Liebig University, 35392 Giessen, Germany; (D.G.); (S.K.)
| | - Max F. Möscheid
- Institute of Parasitology, BFS, Justus Liebig University, 35392 Giessen, Germany; (M.F.M.); (S.H.); (C.G.G.)
| | - Simone Haeberlein
- Institute of Parasitology, BFS, Justus Liebig University, 35392 Giessen, Germany; (M.F.M.); (S.H.); (C.G.G.)
| | - Christoph G. Grevelding
- Institute of Parasitology, BFS, Justus Liebig University, 35392 Giessen, Germany; (M.F.M.); (S.H.); (C.G.G.)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany;
| | - Mohamed El-Kassas
- Endemic Medicine Department, Faculty of Medicine, Helwan University, Cairo 11795, Egypt;
| | - Khaled Zalata
- Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Elke Roeb
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (H.M.); (J.K.S.); (A.S.B.); (V.v.B.); (F.S.); (M.J.H.); (F.P.S.); (A.T.); (E.R.)
| | - Martin Roderfeld
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (H.M.); (J.K.S.); (A.S.B.); (V.v.B.); (F.S.); (M.J.H.); (F.P.S.); (A.T.); (E.R.)
| |
Collapse
|
4
|
Liu M, Wang Y, Deng W, Xie J, He Y, Wang L, Zhang J, Cui M. Combining network pharmacology, machine learning, molecular docking and molecular dynamic to explore the mechanism of Chufeng Qingpi decoction in treating schistosomiasis. Front Cell Infect Microbiol 2024; 14:1453529. [PMID: 39310787 PMCID: PMC11413488 DOI: 10.3389/fcimb.2024.1453529] [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: 06/23/2024] [Accepted: 08/05/2024] [Indexed: 09/25/2024] Open
Abstract
Background Although the Chufeng Qingpi Decoction (CQD) has demonstrated clinical effectiveness in the treatment of schistosomiasis, the precise active components and the underlying mechanisms of its therapeutic action remain elusive. To achieve a profound comprehension, we incorporate network pharmacology, bioinformatics analysis, molecular docking, and molecular dynamics simulations as investigative methodologies within our research framework. Method Utilizing TCMSP and UniProt, we identified formula components and targets. Cytoscape 3.10.0 was used to construct an herb-target interaction network. Genecards, DisGeNET, and OMIM databases were examined for disease-related objectives. A Venn diagram identified the intersection of compound and disease targets. Using Draw Venn, overlapping targets populated STRING for PPI network. CytoNCA identified schistosomiasis treatment targets. GO & KEGG enrichment analysis followed High-scoring genes in PPI were analyzed by LASSO, RF, SVM-RFE. Molecular docking & simulations investigated target-compound interactions. Result The component's target network encompassed 379 nodes, 1629 edges, highlighting compounds such as wogonin, kaempferol, luteolin, and quercetin. Amongst the proteins within the PPI network, PTGS2, TNF, TGFB1, BCL2, TP53, IL10, JUN, MMP2, IL1B, and MYC stood out as the most prevalent entities. GO and KEGG revealed that mainly involved the responses to UV, positive regulation of cell migration and motility. The signal pathways encompassed Pathways in cancer, Lipid and atherosclerosis, Fluid shear stress and atherosclerosis, as well as the AGE-RAGE. Bioinformatics analysis indicated TP53 was the core gene. Ultimately, the molecular docking revealed that wogonin, kaempferol, luteolin, and quercetin each exhibited significant affinity in their respective interactions with TP53. Notably, kaempferol exhibited the lowest binding energy, indicating a highly stable interaction with TP53. Lastly, we validated the stability of the binding interaction between the four small molecules and the TP53 through molecular dynamics simulations. The molecular dynamics simulation further validated the strongest binding between TP53 and kaempferol. In essence, our research groundbreaking in its nature elucidates for the first time the underlying molecular mechanism of CQD in the therapeutic management of schistosomiasis, thereby providing valuable insights and guidance for the treatment of this disease. Conclusion This study uncovered the efficacious components and underlying molecular mechanisms of the Chufeng Qingpi Decoction in the management of schistosomiasis, thereby offering valuable insights for future fundamental research endeavors.
Collapse
Affiliation(s)
- Minglu Liu
- Emergency Department, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yuxin Wang
- Research and Teaching Department of Comparative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Wen Deng
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, China
| | - Jiahao Xie
- Emergency Department, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yanyao He
- Research and Teaching Department of Comparative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Liang Wang
- Research and Teaching Department of Comparative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Jianbin Zhang
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, China
| | - Ming Cui
- Emergency Department, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
5
|
Luh D, Heiles S, Roderfeld M, Grevelding CG, Roeb E, Spengler B. Hepatic Topology of Glycosphingolipids in Schistosoma mansoni-Infected Hamsters. Anal Chem 2024; 96:6311-6320. [PMID: 38594017 PMCID: PMC11044111 DOI: 10.1021/acs.analchem.3c05846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024]
Abstract
Schistosomiasis is a neglected tropical disease caused by worm parasites of the genus Schistosoma. Upon infection, parasite eggs can lodge inside of host organs like the liver. This leads to granuloma formation, which is the main cause of the pathology of schistosomiasis. To better understand the different levels of host-pathogen interaction and pathology, our study focused on the characterization of glycosphingolipids (GSLs). For this purpose, GSLs in livers of infected and noninfected hamsters were studied by combining high-spatial-resolution atmospheric-pressure scanning microprobe matrix-assisted laser desorption/ionization mass spectrometry imaging (AP-SMALDI MSI) with nanoscale hydrophilic interaction liquid chromatography tandem mass spectrometry (nano-HILIC MS/MS). Nano-HILIC MS/MS revealed 60 GSL species with a distinct saccharide and ceramide composition. AP-SMALDI MSI measurements were conducted in positive- and negative-ion mode for the visualization of neutral and acidic GSLs. Based on nano-HILIC MS/MS results, we discovered no downregulated but 50 significantly upregulated GSLs in liver samples of infected hamsters. AP-SMALDI MSI showed that 44 of these GSL species were associated with the granulomas in the liver tissue. Our findings suggest an important role of GSLs during granuloma formation.
Collapse
Affiliation(s)
- David Luh
- Institute
of Inorganic and Analytical Chemistry, Justus
Liebig University Giessen, 35392 Giessen, Germany
| | - Sven Heiles
- Institute
of Inorganic and Analytical Chemistry, Justus
Liebig University Giessen, 35392 Giessen, Germany
- Leibniz-Institut
für Analytische Wissenschaften—ISAS—e.V., 44139 Dortmund, Germany
- Lipidomics,
Faculty of Chemistry, University of Duisburg-Essen, 45141 Essen, Germany
| | - Martin Roderfeld
- Gastroenterology, Justus Liebig University Giessen, 35392Giessen, Germany
| | | | - Elke Roeb
- Gastroenterology, Justus Liebig University Giessen, 35392Giessen, Germany
| | - Bernhard Spengler
- Institute
of Inorganic and Analytical Chemistry, Justus
Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
6
|
Jain S, Rana M. From the discovery of helminths to the discovery of their carcinogenic potential. Parasitol Res 2023; 123:47. [PMID: 38095695 DOI: 10.1007/s00436-023-08022-y] [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: 08/20/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023]
Abstract
Cancer involves a major aberration in the normal behaviour of cells, making them divide continuously, which interferes with the normal physiology of the body. The link between helminths and their cancer-inducing potential has been proposed in the last century. The exact pathway is still not clear but chronic inflammation in response to the deposited eggs, immune response against soluble egg antigens, and co-infection with a third party (a bacteria, a virus, or infection leading to a change in microbiome) seems to be the reasons for cancer induction. This review looks into the historical outlook on helminths along with their epidemiology, morphology, and life cycle. It then focuses on providing correlations between helminth infection and molecular mechanism of carcinogenesis by elaborating upon epidemiological, clinical, and surgical studies. While the cancer-inducing potential has been convincingly established only for a few helminths and studies point out towards possible cancer-inducing ability of the rest of the helminths elucidated in this work, however, more insights into the immunobiology of helminths as well as infected patients are required to conclusively comment upon this ability of the latter.
Collapse
Affiliation(s)
- Sidhant Jain
- Institute for Globally Distributed Open Research and Education (IGDORE), Rewari, Haryana, India.
| | - Meenakshi Rana
- Dyal Singh College, University of Delhi, Lodhi Road, Pragati Vihaar, New Delhi, India
| |
Collapse
|
7
|
Jain S, Rana M, Choubey P, Kumar S. Schistosoma japonicum Associated Colorectal Cancer and Its Management. Acta Parasitol 2023; 68:723-734. [PMID: 37594685 DOI: 10.1007/s11686-023-00707-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND An association between Schistosoma japonicum and colorectal cancer in humans has been known since a long time; however, this association remains understudied and lacks comprehensive experimentation support. OBJECTIVE Various epidemiological and pathological studies have established the role of chronic inflammation as a major factor behind the induction of colorectal cancer. The aim of this review is to present the current knowledge on the association of Schistosoma japonicum with colorectal cancer. RESULT Mechanisms which lead to induction and progression of colorectal cancer are highlighted along with diagnosis and treatment for the same. Further, various methodologies, including mass drug administration, use of new drugs and vaccines, role of apoptosis, and histone-modifying enzymes, have been described which can either prevent the schistosomal infection itself or can check it from reaching an advanced stage. CONCLUSIONS Epidemiological, clinical, pathological and surgical studies suggest that Schistosoma japonicum is responsible for induction of colorectal cancer. However, thorough clinical studies are required to support and globally accept this notion. Further, methodologies highlighted in this work can be employed in order to take care of schistosomal infection or address the cancer induction and progression.
Collapse
Affiliation(s)
- Sidhant Jain
- Institute for Globally Distributed Open Research and Education (IGDORE), Rewari, Haryana, India.
| | - Meenakshi Rana
- Dyal Singh College, University of Delhi, Lodhi Road, Pragati Vihaar, New Delhi, 110003, India
| | - Pooja Choubey
- Department of Zoology, Gate No. 3, Chaatra Marg, University of Delhi, North Campus, Delhi, 110007, India
| | - Sahil Kumar
- Department of Pharmacology, ESIC Dental College and Hospital, Rohini, Sector-15, Delhi, 110089, India
| |
Collapse
|
8
|
von Bülow V, Schneider M, Dreizler D, Russ L, Baier A, Buss N, Lichtenberger J, Härle L, Müller H, Tschuschner A, Schramm G, Pons-Kühnemann J, Grevelding CG, Roeb E, Roderfeld M. Schistosoma mansoni-Induced Oxidative Stress Triggers Hepatocellular Proliferation. Cell Mol Gastroenterol Hepatol 2023; 17:107-117. [PMID: 37696392 PMCID: PMC10665951 DOI: 10.1016/j.jcmgh.2023.08.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND & AIMS Schistosomiasis is one of the most prominent parasite-induced infectious diseases, affecting more than 250 million people. Schistosoma mansoni causes metabolic exhaustion and a strong redox imbalance in the liver, causing parenchymal damage, and may predispose for cancer. We investigated whether oxidative stress provokes hepatocellular proliferation upon S. mansoni infection. METHODS The cell cycle, replication stress response, and proliferation were analyzed on transcriptional and protein levels in the livers of S. mansoni-infected hamsters and by mechanistic gain- and loss-of-function experiments in human hepatoma cells. Major results were validated in human biopsy specimens of S. mansoni-infected patients. RESULTS S. mansoni infection induced licensing factors of DNA replication and cell-cycle checkpoint cyclins in parallel with a DNA damage response in hamster hepatocytes. Moreover, even unisexual infection without egg effects, as a reflection of a chronic inflammatory process, resulted in a moderate activation of several cell-cycle markers. S. mansoni soluble egg antigens induced proliferation of human hepatoma cells that could be abolished by reduced glutathione. CONCLUSIONS Our data suggest that hepatocellular proliferation is triggered by S. mansoni egg-induced oxidative stress.
Collapse
Affiliation(s)
- Verena von Bülow
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Maryam Schneider
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Dorothee Dreizler
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Lena Russ
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Anne Baier
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Nicola Buss
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Jakob Lichtenberger
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Lukas Härle
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Heike Müller
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Annette Tschuschner
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Gabriele Schramm
- Early Life Origin of Chronic Lung Diseases, Priority Research Area Chronic Lung Diseases, Research Center Borstel, Borstel, Germany
| | - Jörn Pons-Kühnemann
- Institute of Medical Informatics, Justus Liebig University Giessen, Giessen, Germany
| | - Christoph G Grevelding
- Institute of Parasitology, Biomedizinisches Forschungszentrum Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Elke Roeb
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Martin Roderfeld
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
9
|
von Bülow V, Gindner S, Baier A, Hehr L, Buss N, Russ L, Wrobel S, Wirth V, Tabatabai K, Quack T, Haeberlein S, Kadesch P, Gerbig S, Wiedemann KR, Spengler B, Mehl A, Morlock G, Schramm G, Pons-Kühnemann J, Falcone FH, Wilson RA, Bankov K, Wild P, Grevelding CG, Roeb E, Roderfeld M. Metabolic reprogramming of hepatocytes by Schistosoma mansoni eggs. JHEP Rep 2022; 5:100625. [PMID: 36590323 PMCID: PMC9800334 DOI: 10.1016/j.jhepr.2022.100625] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
Abstract
Background & Aims Schistosomiasis is a parasitic infection which affects more than 200 million people globally. Schistosome eggs, but not the adult worms, are mainly responsible for schistosomiasis-specific morbidity in the liver. It is unclear if S. mansoni eggs consume host metabolites, and how this compromises the host parenchyma. Methods Metabolic reprogramming was analyzed by matrix-assisted laser desorption/ionization mass spectrometry imaging, liquid chromatography with high-resolution mass spectrometry, metabolite quantification, confocal laser scanning microscopy, live cell imaging, quantitative real-time PCR, western blotting, assessment of DNA damage, and immunohistology in hamster models and functional experiments in human cell lines. Major results were validated in human biopsies. Results The infection with S. mansoni provokes hepatic exhaustion of neutral lipids and glycogen. Furthermore, the distribution of distinct lipid species and the regulation of rate-limiting metabolic enzymes is disrupted in the liver of S. mansoni infected animals. Notably, eggs mobilize, incorporate, and store host lipids, while the associated metabolic reprogramming causes oxidative stress-induced DNA damage in hepatocytes. Administration of reactive oxygen species scavengers ameliorates these deleterious effects. Conclusions Our findings indicate that S. mansoni eggs completely reprogram lipid and carbohydrate metabolism via soluble factors, which results in oxidative stress-induced cell damage in the host parenchyma. Impact and implications The authors demonstrate that soluble egg products of the parasite S. mansoni induce hepatocellular reprogramming, causing metabolic exhaustion and a strong redox imbalance. Notably, eggs mobilize, incorporate, and store host lipids, while the metabolic reprogramming causes oxidative stress-induced DNA damage in hepatocytes, independent of the host's immune response. S. mansoni eggs take advantage of the host environment through metabolic reprogramming of hepatocytes and enterocytes. By inducing DNA damage, this neglected tropical disease might promote hepatocellular damage and thus influence international health efforts.
Collapse
Key Words
- DMPE, dimethyl-phosphatidylethanolamine
- DNA damage
- GS, glycogen synthase
- GSH, reduced L-glutathione
- HCC, hepatocellular carcinoma
- Lipid
- MALDI-MSI, matrix assisted laser desorption/ionization mass spectrometry imaging
- MDA, malondialdehyde
- OA, oleic acid
- Oxidative stress
- PAS, periodic acid-Schiff
- PC, phosphatidylcholine
- PDH, pyruvate dehydrogenase
- PE, phosphatidylethanolamine
- PLIN2, perilipin 2
- Parasite
- ROS, reactive oxygen species
- S. japonicum, Schistosoma japonicum
- S. mansoni, Schistosoma mansoni
- SEA, soluble egg antigens
- Schistosomiasis
- TG, triglyceride
- bs, bisex
- flOA, fluorescently labelled OA
- hRF, retention factor ∗ 100
- ms, monosex
- ni, non-infected
Collapse
Affiliation(s)
- Verena von Bülow
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany
| | - Sarah Gindner
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany
| | - Anne Baier
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany
| | - Laura Hehr
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany
| | - Nicola Buss
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany
| | - Lena Russ
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany
| | - Sarah Wrobel
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany
| | - Victoria Wirth
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany
| | - Kuscha Tabatabai
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany
| | - Thomas Quack
- Institute of Parasitology, BFS, Justus Liebig University, Schubertstr. 81, 35392 Giessen, Germany
| | - Simone Haeberlein
- Institute of Parasitology, BFS, Justus Liebig University, Schubertstr. 81, 35392 Giessen, Germany
| | - Patrik Kadesch
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| | - Stefanie Gerbig
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| | - Katja R. Wiedemann
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| | - Annabel Mehl
- Institute of Nutritional Science, Food Science Department, and Interdisciplinary Research Center (iFZ), Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany
| | - Gertrud Morlock
- Institute of Nutritional Science, Food Science Department, and Interdisciplinary Research Center (iFZ), Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany
| | - Gabriele Schramm
- Experimental Pneumology, Priority Research Area Asthma & Allergy, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany
| | - Jörn Pons-Kühnemann
- Institute of Medical Informatics, Justus Liebig University, Rudolf-Buchheim-Str. 6. 35392 Giessen, Germany
| | - Franco H. Falcone
- Institute of Parasitology, BFS, Justus Liebig University, Schubertstr. 81, 35392 Giessen, Germany
| | - R. Alan Wilson
- York Biomedical Research Institute, Department of Biology, University of York, York YO10 5DD, UK
| | - Katrin Bankov
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Peter Wild
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Christoph G. Grevelding
- Institute of Parasitology, BFS, Justus Liebig University, Schubertstr. 81, 35392 Giessen, Germany
| | - Elke Roeb
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany
| | - Martin Roderfeld
- Department of Gastroenterology, Justus Liebig University, Klinikstr. 33, 35392 Giessen, Germany,Corresponding author. Address: Department of Gastroenterology, Justus Liebig University Giessen, Gaffkystr. 11, 35392 Giessen, Germany. Tel.: +49 641 99 42527, fax: +49 641 99 42333.
| |
Collapse
|
10
|
Abstract
INTRODUCTION Schistosomes are long-lived blood dwelling helminth parasites using intricate mechanisms to invade, mature, and reproduce inside their vertebrate hosts, whilst simultaneously deploying immune evasion strategies. Their multi-tissue organization and solid body plan presents particular problems for the definition of sub-proteomes. AREAS COVERED Here, we focus on the two host-parasite interfaces of the adult worm accessible to the immune system, namely the tegument and the alimentary tract, but also on the secretions of the infective cercaria, the migrating schistosomulum and the mature egg. In parallel, we introduce the concepts of "leakyome' and 'disintegrome' to emphasize the importance of interpreting data in the context of schistosome biology so that misleading conclusions about the distinct proteome compositions are avoided. Lastly, we highlight the possible clinical implications of the reviewed proteomic findings for pathogenesis, vaccine design and diagnostics. EXPERT OPINION Proteomics has provided considerable insights into the biology of schistosomes, most importantly for rational selection of novel vaccine candidates that might confer protective immunity, but also into the pathogenesis of schistosomiasis. However, given the increasing sensitivity of mass spectrometric instrumentation, we stress the need for care in data interpretation since schistosomes do not deviate from the fundamental rules of eukaryotic cell biology.
Collapse
Affiliation(s)
- William Castro-Borges
- Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brasil
| | - R Alan Wilson
- Department of Biology and York Biomedical Research Institute, University of York, Heslington, York, UK
| |
Collapse
|
11
|
Changes in the lipid profile of hamster liver after Schistosoma mansoni infection, characterized by mass spectrometry imaging and LC-MS/MS analysis. Anal Bioanal Chem 2022; 414:3653-3665. [PMID: 35320368 PMCID: PMC9035427 DOI: 10.1007/s00216-022-04006-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/18/2022] [Accepted: 03/03/2022] [Indexed: 11/01/2022]
Abstract
Schistosomiasis, caused by the human parasite Schistosoma mansoni, is one of the WHO-listed neglected tropical diseases (NTDs), and it has severe impact on morbidity and mortality, especially in Africa. Not only the adult worms but also their eggs are responsible for health problems. Up to 50% of the eggs produced by the female worms are not excreted with the feces but are trapped in the host tissue, such as the liver, where they provoke immune responses and a change in the lipid profile. We built up a database with 372 infection markers found in livers of S. mansoni-infected hamsters, using LC-MS/MS for identification, followed by statistical analysis. Most of them belong to the lipid classes of phosphatidylcholines (PCs), phosphatidylethanolamines (PEs), and triglycerides (TGs). We assigned some of these markers to specific anatomical structures by applying high-resolution MALDI MSI to cryosections of hamster liver and generating ion images based on the marker list from the LC-MS/MS experiments. Furthermore, enrichment and depletion of several markers were visualized.
Collapse
|
12
|
Liu Y, Cheng W, Zhao Y, Gao L, Chang Y, Tong Z, Li H, Jing J. Cyclic Mechanical Strain Regulates Osteoblastic Differentiation of Mesenchymal Stem Cells on TiO 2 Nanotubes Through GCN5 and Wnt/β-Catenin. Front Bioeng Biotechnol 2021; 9:735949. [PMID: 34869255 PMCID: PMC8634263 DOI: 10.3389/fbioe.2021.735949] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 09/30/2021] [Indexed: 02/03/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) play a critical role in bone formation and are extremely sensitive to external mechanical stimuli. Mechanical signals can regulate the biological behavior of cells on the surface of titanium-related prostheses and inducing osteogenic differentiation of BMSCs, which provides the integration of host bone and prosthesis benefits. But the mechanism is still unclear. In this study, BMSCs planted on the surface of TiO2 nanotubes were subjected to cyclic mechanical stress, and the related mechanisms were explored. The results of alkaline phosphatase staining, real-time PCR, and Western blot showed that cyclic mechanical stress can regulate the expression level of osteogenic differentiation markers in BMSCs on the surface of TiO2 nanotubes through Wnt/β-catenin. As an important member of the histone acetyltransferase family, GCN5 exerted regulatory effects on receiving mechanical signals. The results of the ChIP assay indicated that GCN5 could activate the Wnt promoter region. Hence, we concluded that the osteogenic differentiation ability of BMSCs on the surface of TiO2 nanotubes was enhanced under the stimulation of cyclic mechanical stress, and GCN5 mediated this process through Wnt/β-catenin.
Collapse
Affiliation(s)
- Yanchang Liu
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Wendan Cheng
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yao Zhao
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Liang Gao
- Sino Euro Orthopaedics Network, Berlin, Germany
| | - Yongyun Chang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhicheng Tong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huiwu Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Juehua Jing
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
von Bülow V, Lichtenberger J, Grevelding CG, Falcone FH, Roeb E, Roderfeld M. Does Schistosoma Mansoni Facilitate Carcinogenesis? Cells 2021; 10:1982. [PMID: 34440754 PMCID: PMC8393187 DOI: 10.3390/cells10081982] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/21/2021] [Accepted: 08/01/2021] [Indexed: 12/24/2022] Open
Abstract
Schistosomiasis is one of the most prominent parasite-induced infectious diseases, causing tremendous medical and socioeconomic problems. Current studies have reported on the spread of endemic regions and the fear of development of resistance against praziquantel, the only effective drug available. Among the Schistosoma species, only S. haematobium is classified as a Group 1 carcinogen (definitely cancerogenic to humans), causing squamous cell carcinoma of the bladder, whereas infection with S. mansoni is included in Group 3 of carcinogenic hazards to humans by the International Agency for Research on Cancer (IARC), indicating insufficient evidence to determine its carcinogenicity. Nevertheless, although S. mansoni has not been discussed as an organic carcinogen, the multiplicity of case reports, together with recent data from animal models and cell culture experiments, suggests that this parasite can predispose patients to or promote hepatic and colorectal cancer. In this review, we discuss the current data, with a focus on new developments regarding the association of S. mansoni infection with human cancer and the recently discovered biomolecular mechanisms by which S. mansoni may predispose patients to cancer development and carcinogenesis.
Collapse
Affiliation(s)
- Verena von Bülow
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (V.v.B.); (J.L.); (E.R.)
| | - Jakob Lichtenberger
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (V.v.B.); (J.L.); (E.R.)
| | - Christoph G. Grevelding
- Institute of Parasitology, BFS, Justus Liebig University, 35392 Giessen, Germany; (C.G.G.); (F.H.F.)
| | - Franco H. Falcone
- Institute of Parasitology, BFS, Justus Liebig University, 35392 Giessen, Germany; (C.G.G.); (F.H.F.)
| | - Elke Roeb
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (V.v.B.); (J.L.); (E.R.)
| | - Martin Roderfeld
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (V.v.B.); (J.L.); (E.R.)
| |
Collapse
|