1
|
Carrera Silva EA, Puyssegur J, Errasti AE. Coevolutionary interplay: Helminths-trained immunity and its impact on the rise of inflammatory diseases. eLife 2025; 14:e105393. [PMID: 40231720 PMCID: PMC12002795 DOI: 10.7554/elife.105393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
The gut biome, a complex ecosystem of micro- and macro-organisms, plays a crucial role in human health. A disruption in this evolutive balance, particularly during early life, can lead to immune dysregulation and inflammatory disorders. 'Biome repletion' has emerged as a potential therapeutic approach, introducing live microbes or helminth-derived products to restore immune balance. While helminth therapy has shown some promise, significant challenges remain in optimizing clinical trials. Factors such as patient genetics, disease status, helminth species, and the optimal timing and dosage of their products or metabolites must be carefully considered to train the immune system effectively. We aim to discuss how helminths and their products induce trained immunity as prospective to treat inflammatory and autoimmune diseases. The molecular repertoire of helminth excretory/secretory products (ESPs), which includes proteins, peptides, lipids, and RNA-carrying extracellular vesicles (EVs), underscores their potential to modulate innate immune cells and hematopoietic stem cell precursors. Mimicking natural delivery mechanisms like synthetic exosomes could revolutionize EV-based therapies and optimizing production and delivery of ESP will be crucial for their translation into clinical applications. By deciphering and harnessing helminth-derived products' diverse modes of action, we can unleash their full therapeutic potential and pave the way for innovative treatments.
Collapse
Affiliation(s)
- Eugenio Antonio Carrera Silva
- EACS and JP Institute of Experimental Medicine, National Scientific and Technical Research Council, National Academy of Medicine (IMEX-CONICET-ANM)Buenos AiresArgentina
| | - Juliana Puyssegur
- EACS and JP Institute of Experimental Medicine, National Scientific and Technical Research Council, National Academy of Medicine (IMEX-CONICET-ANM)Buenos AiresArgentina
| | - Andrea Emilse Errasti
- AEE Institute of Pharmacology, School of Medicine, University of Buenos AiresBuenos AiresArgentina
- National Scientific and Technical Research Council (CONICET)Buenos AiresArgentina
| |
Collapse
|
2
|
Li H, Shan C, Zhu Y, Yao X, Lin L, Zhang X, Qian Y, Wang Y, Xu J, Zhang Y, Li H, Zhao L, Chen K. Helminth-induced immune modulation in colorectal cancer: exploring therapeutic applications. Front Immunol 2025; 16:1484686. [PMID: 40297577 PMCID: PMC12034720 DOI: 10.3389/fimmu.2025.1484686] [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: 08/22/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Colorectal cancer is one of the most lethal tumors, posing a financial and healthcare burden. This study investigates how helminths and pre-existing diseases such as colitis, obesity, diabetes, and gut microbiota issues influence colon cancer development and prognosis. The immune system's protective immunosuppressive response to helminth invasion minimizes inflammation-induced cell damage and DNA mutations, lowering the risk of colorectal cancer precursor lesions. Helminth infection-mediated immunosuppression can hasten colorectal cancer growth and metastasis, which is detrimental to patient outcomes. Some helminth derivatives can activate immune cells to attack cancer cells, making them potentially useful as colorectal cancer vaccines or therapies. This review also covers gene editing approaches. We discovered that using CRISPR/Cas9 to inhibit live helminths modulates miRNA, which limits tumor growth. We propose more multicenter studies into helminth therapy's long-term effects and immune regulation pathways. We hope to treat colorectal cancer patients with helminth therapy and conventional cancer treatments in an integrative setting.
Collapse
Affiliation(s)
- Hongyu Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- Ocean College, Beibu Gulf University, Qinzhou, China
| | - Chaojun Shan
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yunhuan Zhu
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xiaodong Yao
- School of Marxism, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lijun Lin
- School of Basic Medicine and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Xiaofen Zhang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuncheng Qian
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuqing Wang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jialu Xu
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yijie Zhang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Hairun Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Ling Zhao
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| |
Collapse
|
3
|
Fogang BAN, Meyer J, Debrah LB, Owusu M, Agyei G, Mensah DA, Boateng J, Mensah JO, Klarmann-Schulz U, Horn S, Kroidl I, Ackah EB, Phillips RO, Sylverken A, Debrah AY, Hoerauf A, Adjobimey T. Helminth Seropositivity Inversely Correlated with Th1 and Th17 Cytokines and Severe COVID-19. Vaccines (Basel) 2025; 13:252. [PMID: 40266113 PMCID: PMC11946601 DOI: 10.3390/vaccines13030252] [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: 12/23/2024] [Revised: 02/11/2025] [Accepted: 02/18/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: The COVID-19 pandemic has significantly impacted global health. However, Africa has reported relatively low numbers of cases and fatalities. Although the pandemic has largely receded, the reasons for its milder course on the African continent have not yet been fully clarified. This study explored the hypothesis that helminth co-infections may have contributed to these observations. Methods: A retrospective cohort study was conducted using 104 plasma samples collected during the third wave of the pandemic in the Ashanti Region of Ghana. Luminex assays were used to measure SARS-CoV-2-specific IgA and IgG, neutralizing antibodies, systemic cytokines and helminth-specific IgG. Results: The results indicated that the highest cumulative seroprevalence of helminths (61.5%) was observed in asymptomatic COVID-19 patients. In comparison, mild and moderate patients had helminth seropositivity rates of 43.8% and 34.5%, respectively, which were 1.4 and 1.8 times lower than those of the asymptomatic group, respectively. Notably, the two severe COVID-19 cases investigated were seronegative for all three of the helminths tested. Strikingly, co-exposure resulted in lower SARS-CoV-2-specific IgA/IgG expression and reduced neutralization potential. However, co-seropositive individuals for helminths and SARS-CoV-2 exhibited a higher expression of Th2 cytokines and IL-10 over Th1 cytokines compared to SARS-CoV-2-positive individuals alone. Conclusion: These data suggest that co-exposure to helminths could mitigate the severity of COVID-19 outcomes by reducing the Th1 and Th17 responses; this highlights the potential protective role of helminthiasis against severe COVID-19. These findings provide valuable insights for the development of public health policies in helminth-endemic regions and underscore the importance of considering helminth co-infections in managing viral infections. It also offers a plausible explanation for the milder disease severity observed in helminth-endemic regions while raising critical considerations regarding vaccine efficacy, as helminth-induced immune modulation may influence the magnitude and quality of vaccine-induced immune responses.
Collapse
Affiliation(s)
- Brice Armel Nembot Fogang
- Department of Clinical Microbiology, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- German West African Center for Global Health and Pandemic Prevention (G-WAC), Kumasi 00233, Ghana
| | - Julia Meyer
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
| | - Linda B. Debrah
- Department of Clinical Microbiology, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- German West African Center for Global Health and Pandemic Prevention (G-WAC), Kumasi 00233, Ghana
| | - Michael Owusu
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- German West African Center for Global Health and Pandemic Prevention (G-WAC), Kumasi 00233, Ghana
- Department of Medical Diagnostics, Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
| | - George Agyei
- Department of Clinical Microbiology, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- German West African Center for Global Health and Pandemic Prevention (G-WAC), Kumasi 00233, Ghana
| | - Derrick Adu Mensah
- Department of Clinical Microbiology, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- Department of Medical Laboratory Technology, Royal Ann College of Health, Kumasi P.O. Box KS 6253, Ghana
- Department of Public Health Education, Akenten Appiah-Menka University of Skills Training and Entrepreneurial Development, Kumasi P.O Box 1277, Ghana
| | - John Boateng
- Department of Clinical Microbiology, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- German West African Center for Global Health and Pandemic Prevention (G-WAC), Kumasi 00233, Ghana
| | - Jubin Osei Mensah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- Department of Pathobioogy, School of Veterinary Medicine, Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
| | - Ute Klarmann-Schulz
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
- Bonn-Cologne Site, German Center for Infectious Disease Research (DZIF), Bonn, Germany
| | - Sacha Horn
- Institute of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, 80539 Munich, Germany
| | - Inge Kroidl
- Institute of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, 80539 Munich, Germany
- Munich Site, German Center for Infectious Disease Research (DZIF), Munich, Germany
| | - Ezekiel Bonwin Ackah
- Department of Medical Diagnostics, Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
| | - Richard O. Phillips
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
| | - Augustina Sylverken
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
| | - Alexander Y. Debrah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
- German West African Center for Global Health and Pandemic Prevention (G-WAC), Kumasi 00233, Ghana
- Faculty of Allied Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ghana
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
- German West African Center for Global Health and Pandemic Prevention (G-WAC), Kumasi 00233, Ghana
- Bonn-Cologne Site, German Center for Infectious Disease Research (DZIF), Bonn, Germany
| | - Tomabu Adjobimey
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
- Faculté des Sciences et Techniques (FAST), Université d’Abomey Calavi, Abomey Calavi 05 BP 1604, Benin
| |
Collapse
|
4
|
Wu J, Yang Z, Wang D, Xiao Y, Shao J, Ren K. Human Umbilical Cord Mesenchymal Stem Cell-derived Exosome Regulates Intestinal Type 2 Immunity. Curr Stem Cell Res Ther 2025; 20:302-316. [PMID: 38779734 DOI: 10.2174/011574888x314032240429113240] [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: 03/11/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024]
Abstract
AIMS The aim of this study was to investigate the role of human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-Exo) in regulating the intestinal type 2 immune response for either protection or therapy. BACKGROUND hUCMSC-Exo was considered a novel cell-free therapeutic product that shows promise in the treatment of various diseases. Type 2 immunity is a protective immune response classified as T-helper type 2 (Th2) cells and is associated with helminthic infections and allergic diseases. The effect of hUCMSC-Exo on intestinal type 2 immune response is not clear. METHOD C57BL/6 mice were used to establish intestinal type 2 immune response by administering of H. poly and treated with hUCMSC-Exo before or after H. poly infection. Intestinal organoids were isolated and co-cultured with IL-4 and hUCMSC-Exo. Then, we monitored the influence of hUCMSC-Exo on type 2 immune response by checking adult worms, the hyperplasia of tuft and goblet cells Result: hUCMSC-Exo significantly delays the colonization of H. poly in subserosal layer of duodenum on day 7 post-infection and promotes the hyperplasia of tuft cells and goblet cells on day 14 post-infection. HUCMSC-Exo enhances the expansion of tuft cells in IL-4 treated intestinal organoids, and promotes lytic cell death. CONCLUSION Our study demonstrates hUCMSC-Exo may benefit the host by increasing the tolerance at an early infection stage and then enhancing the intestinal type 2 immune response to impede the helminth during Th2 priming. Our results show hUCMSC-Exo may be a positive regulator of type 2 immune response, suggesting hUCMSC-Exo has a potential therapeutic effect on allergic diseases.
Collapse
Affiliation(s)
- Jiajun Wu
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
- The Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
| | - Zhen Yang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
- The Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
| | - Daoyuan Wang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
- The Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
| | - Yihui Xiao
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
- The Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
| | - Jia Shao
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
- The Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
| | - Kaiqun Ren
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
- The Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
| |
Collapse
|
5
|
Cui X, Cheng Y, Wang H, Li X, Li J, Zhang K, Mou R. Hymenolepis nana antigens alleviate ulcerative colitis by promoting intestinal stem cell proliferation and differentiation via AhR/IL-22 signaling pathway. PLoS Negl Trop Dis 2024; 18:e0012714. [PMID: 39666730 DOI: 10.1371/journal.pntd.0012714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/26/2024] [Accepted: 11/20/2024] [Indexed: 12/14/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease with an unknown etiology and is difficult to treat. Studies have shown that some helminths and their associated products have therapeutic potential in controlling or preventing inflammatory diseases. This study is to investigate the mitigation effects of Hymenolepis nana antigens (HnAg) on the UC model. HnAg significantly improved the disease activity index, colon length, and colonic pathological damage in mice with dextran sulfate sodium (DSS)-induced colitis. HnAg intervention could protect the number of goblet cells and enhance the expression of tight junction proteins and mucins, thereby improving intestinal barrier integrity. HnAg attenuated small intestinal organoid damage and stimulated intestinal stem cells proliferation in a DSS-induced mouse organoid inflammation model. The protective mechanism of HnAg might be related to the activation of the aryl hydrocarbon receptor (AhR)/IL-22 signaling pathway, which regulates intestinal barrier function and promotes the proliferation and differentiation of intestinal stem cells. In conclusion, HnAg has a therapeutic effect on UC mice. Our study provides a new approach for alleviating UC by Hymenolepis nana and its associated products.
Collapse
Affiliation(s)
- Xuanyin Cui
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control / The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Department of Human Parasitology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Yi Cheng
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control / The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Department of Human Parasitology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Hongyan Wang
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control / The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Department of Human Parasitology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Xiaomao Li
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control / The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Department of Human Parasitology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Jinfu Li
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control / The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Department of Human Parasitology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Ke Zhang
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control / The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Department of Human Parasitology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Rong Mou
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control / The Key and Characteristic Laboratory of Modern Pathogenicity Biology, Department of Human Parasitology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| |
Collapse
|
6
|
Wang X, Peng J, Cai P, Xia Y, Yi C, Shang A, Akanyibah FA, Mao F. The emerging role of the gut microbiota and its application in inflammatory bowel disease. Biomed Pharmacother 2024; 179:117302. [PMID: 39163678 DOI: 10.1016/j.biopha.2024.117302] [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: 06/19/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/22/2024] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex disorder with an unknown cause. However, the dysbiosis of the gut microbiome has been found to play a role in IBD etiology, including exacerbated immune responses and defective intestinal barrier integrity. The gut microbiome can also be a potential biomarker for several diseases, including IBD. Currently, conventional treatments targeting pro-inflammatory cytokines and pathways in IBD-associated dysbiosis do not yield effective results. Other therapies that directly target the dysbiotic microbiome for effective outcomes are emerging. We review the role of the gut microbiome in health and IBD and its potential as a diagnostic, prognostic, and therapeutic target for IBD. This review also explores emerging therapeutic advancements that target gut microbiome-associated alterations in IBD, such as nanoparticle or encapsulation delivery, fecal microbiota transplantation, nutritional therapies, microbiome/probiotic engineering, phage therapy, mesenchymal stem cells (MSCs), gut proteins, and herbal formulas.
Collapse
Affiliation(s)
- Xiu Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China
| | - Jianhua Peng
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu 212300, China
| | - Peipei Cai
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yuxuan Xia
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang 212028, China
| | - Anquan Shang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China
| | - Francis Atim Akanyibah
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China.
| |
Collapse
|
7
|
Jamtsho T, Loukas A, Wangchuk P. Pharmaceutical Potential of Remedial Plants and Helminths for Treating Inflammatory Bowel Disease. Pharmaceuticals (Basel) 2024; 17:819. [PMID: 39065669 PMCID: PMC11279646 DOI: 10.3390/ph17070819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/16/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Research is increasingly revealing that inflammation significantly contributes to various diseases, particularly inflammatory bowel disease (IBD). IBD is a major medical challenge due to its chronic nature, affecting at least one in a thousand individuals in many Western countries, with rising incidence in developing nations. Historically, indigenous people have used natural products to treat ailments, including IBD. Ethnobotanically guided studies have shown that plant-derived extracts and compounds effectively modulate immune responses and reduce inflammation. Similarly, helminths and their products offer unique mechanisms to modulate host immunity and alleviate inflammatory responses. This review explored the pharmaceutical potential of Aboriginal remedial plants and helminths for treating IBD, emphasizing recent advances in discovering anti-inflammatory small-molecule drug leads. The literature from Scopus, MEDLINE Ovid, PubMed, Google Scholar, and Web of Science was retrieved using keywords such as natural product, small molecule, cytokines, remedial plants, and helminths. This review identified 55 important Aboriginal medicinal plants and 9 helminth species that have been studied for their anti-inflammatory properties using animal models and in vitro cell assays. For example, curcumin, berberine, and triptolide, which have been isolated from plants; and the excretory-secretory products and their protein, which have been collected from helminths, have demonstrated anti-inflammatory activity with lower toxicity and fewer side effects. High-throughput screening, molecular docking, artificial intelligence, and machine learning have been engaged in compound identification, while clustered regularly interspaced short palindromic repeats (CRISPR) gene editing and RNA sequencing have been employed to understand molecular interactions and regulations. While there is potential for pharmaceutical application of Aboriginal medicinal plants and gastrointestinal parasites in treating IBD, there is an urgent need to qualify these plant and helminth therapies through reproducible clinical and mechanistic studies.
Collapse
Affiliation(s)
- Tenzin Jamtsho
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia
- Australian Institute of Tropical Health and Medicine (AITHM), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia;
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine (AITHM), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia;
| | - Phurpa Wangchuk
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia
- Australian Institute of Tropical Health and Medicine (AITHM), Cairns Campus, James Cook University, Cairns, QLD 4878, Australia;
| |
Collapse
|
8
|
Ishii Y, Matsunaga T, Yasui T, Rini DM, Inoue R, Yamamoto Y, Suzuki T. Supplemental Psyllium Fiber Increases Antimicrobial Proteins via the Tuft Cell-ILC2 Circuit and Type II Immune Response in the Mouse Small Intestine. NUTRACEUTICALS 2024; 4:307-322. [DOI: 10.3390/nutraceuticals4020019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Dietary fibers regulate intestinal barrier function; however, the precise mechanisms remain unclear. This study investigated the effects of psyllium fibers on antimicrobial protein expression, focusing on the type II immunity and tuft cell-group 2 innate lymphoid cell (ILC2) circuit in the small intestine of the mouse. Supplemental psyllium fiber upregulated antimicrobial proteins, such as small proline-rich protein 2A (SPRR2A) and resistin-like beta (RELMβ), in mouse small intestine, evidently affecting cecal microbiota composition. The psyllium fibers also increased the RNA and protein expression of molecules related to ILC2 and tuft cells, such as IL-13, IL-25, DCLK1, Gfi-1b, SH2 domain-containing protein 3C, and Spi-B. In addition, ILC2 inhibitor (disulfiram) and bitter taste receptor blocker administration reduced psyllium-induced SPRR2A and RELMβ expression. Collectively, psyllium supplementation upregulates antimicrobial proteins such as SPRR2A and RELMß via the type II immune response and tuft cell-ILC2 circuit in the mouse small intestine.
Collapse
Affiliation(s)
- Yoshiki Ishii
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
| | - Taiyo Matsunaga
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
| | - Tomoki Yasui
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
| | - Dina Mustika Rini
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
- Department of Food Technology, Faculty of Engineering, Universitas Pembangunan Nasional “Veteran” Jawa Timur, Surabaya 60294, Indonesia
| | - Ryo Inoue
- Faculty of Agriculture, Setsunan University, 45-1 Nagaotoge-cho, Hirakata 573-0101, Japan
| | - Yoshinari Yamamoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
| | - Takuya Suzuki
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima 739-8528, Japan
| |
Collapse
|
9
|
Juarez-Estrada MA, Graham D, Hernandez-Velasco X, Tellez-Isaias G. Editorial: Parasitism: the good, the bad and the ugly. Front Vet Sci 2023; 10:1304206. [PMID: 37915945 PMCID: PMC10616899 DOI: 10.3389/fvets.2023.1304206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 11/03/2023] Open
Affiliation(s)
- Marco A. Juarez-Estrada
- Department of Medicine and Zootechnics of Birds, College of Veterinary Medicine and Zootechnics, National Autonomous University of Mexico, Mexico City, Mexico
| | - Danielle Graham
- Division of Agriculture, Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Xochitl Hernandez-Velasco
- Department of Medicine and Zootechnics of Birds, College of Veterinary Medicine and Zootechnics, National Autonomous University of Mexico, Mexico City, Mexico
| | - Guillermo Tellez-Isaias
- Division of Agriculture, Department of Poultry Science, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
10
|
Pallio G. Editorial: Novel Therapeutic Approaches in Inflammatory Bowel Diseases. Biomedicines 2023; 11:2466. [PMID: 37760907 PMCID: PMC10526183 DOI: 10.3390/biomedicines11092466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) encompass ulcerative colitis (UC) and Crohn's disease (CD), both of which are inflammatory ailments affecting the gastrointestinal tract [...].
Collapse
Affiliation(s)
- Giovanni Pallio
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, Via C. Valeria, 98125 Messina, Italy
| |
Collapse
|
11
|
Thongrin T, Suyapoh W, Wendo W, Tangkawattana P, Sukon P, Salao K, Suttiprapa S, Saichua P, Tangkawatana S. Inflammatory cell responses in biliary mucosa during Opisthorchis viverrini infection: Insights into susceptibility differences among hosts. Open Vet J 2023; 13:1150-1166. [PMID: 37842106 PMCID: PMC10576576 DOI: 10.5455/ovj.2023.v13.i9.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/15/2023] [Indexed: 10/17/2023] Open
Abstract
Background Individual host susceptibility is believed to be a risk factor in the interaction between the host and the parasite. Since studying time series in humans is limited, animal models are replaced. Aim This study aims to explore and compare the pattern of inflammatory cell types along the biliary tract and their association with proliferative lesions in the early development of cholangiocarcinoma from susceptible and nonsusceptible animal models. Methods Thirty male Syrian golden hamsters and 30 BALB/c mice, serving as the susceptible and nonsusceptible animal models, were used in this comparative study. The animals were infected with 50 Opisthorchis viverrini metacercariae via gastric intubation. At days 1, 2, 7, 14, 28, and 56 postinfection (p.i.), five animals were randomly selected from each group and humanely sacrificed. The hepatobiliary tissues were collected and processed for histopathological study. Histochemical and immunohistochemical staining were applied to differentiate the inflammatory cell types. Kruskal-Wallis and Mann-Whitney tests were applied to assess all semi-quantitative and quantitative variables. The correlation between each variable was also analyzed using Spearman rank at a p-value < 0.05. Results The results demonstrated that mice had different patterns of infiltrating cell types when compared to hamsters. This suggested that the cellular response to the infection in mice occurred earlier than that in hamsters. The response in mice reached its peak at D7 to D14 and then rapidly declined at D28. In contrast, although the inflammatory response in hamsters started slowly, the response reached the peak at D28 and maintained a high level until D56. Significant differences in the number of inflammatory cells between mice and hamsters were seen at D1 (p = 0.047), D7 (p = 0.049), D28 (p = 0.040), and D56 (p < 0.040). Conclusion The inflammatory responses to O. viverrini infection in the nonsusceptible animal model occurred and declined earlier while the response in the susceptible animal model occurred later in a gradual manner. Both rodents are suitable animal models for the studies of opisthorchiasis susceptibility.
Collapse
Affiliation(s)
- Theerayut Thongrin
- Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharapol Suyapoh
- Faculty of Veterinary Science, Prince of Songkla University, Songkhla, Thailand
| | - WoroDanur Wendo
- Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | - Peerapol Sukon
- Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kanin Salao
- Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sutas Suttiprapa
- Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| | - Prasert Saichua
- Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| | - Sirikachorn Tangkawatana
- Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|