1
|
Shakra MY, Geneedy MRM, Ahmad HK, Mazen MAI, Mostafa ME. Impact of Soluble Schistosomal Egg Antigens on Type 1 Diabetes Mellitus in an Induced Diabetic Mouse Model. Acta Parasitol 2025; 70:98. [PMID: 40299229 PMCID: PMC12041111 DOI: 10.1007/s11686-025-01035-w] [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: 01/03/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE This study aimed to investigate the effects of Schistosoma mansoni soluble egg antigen (SEA) on type 1 diabetes (T1D) in a streptozotocin (STZ)-induced diabetic mouse model. METHODS The study examined the effects of Schistosoma mansoni soluble egg antigen (SEA) on type 1 diabetes (T1D) using a mouse model, involving 50 mice divided into three groups: a healthy control group receiving phosphate-buffered saline (PBS), a diabetic control group with STZ-induced T1D also receiving PBS, and a diabetic treated group receiving SEA. Biochemical and immunological analyses were conducted on blood samples collected at four and eight weeks post-treatment to assess metabolic markers like blood glucose and insulin levels, as well as immune markers including TNF-α, TGF-β, FOXp3, IL-4, and IL-10. RESULTS SEA treatment induced early immune modulation at four weeks and sustained metabolic and immunological improvements at eight weeks, marked by increased regulatory T cells (elevated FOXp3), activation of immunosuppressive pathways (increased TGF-β), reduced inflammation (decreased TNF-α), a shift to an anti-inflammatory Th2 response (elevated IL-4 and IL-10), improved glycemic control, lower blood glucose levels, and higher insulin levels. CONCLUSION SEA exhibits potential therapeutic effects against T1D by modulating immune responses, promoting Th2 polarization, and increasing regulatory T cell activity. This immunological shift reduces systemic inflammation and enhances glycemic control.
Collapse
Affiliation(s)
- Mohammed Y Shakra
- Department of Parasitology, Faculty of Medicine, Al-Azhar University, Damietta, Egypt.
| | - Morsy R M Geneedy
- Department of Parasitology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Haitham Kh Ahmad
- Department of Parasitology, Faculty of Medicine, Al-Azhar University, Assuit, Egypt
| | - Moamen A I Mazen
- Department of Parasitology, Faculty of Medicine, Al-Azhar University, Assuit, Egypt
| | - Mostafa E Mostafa
- Department of Parasitology, Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| |
Collapse
|
2
|
El-Kady AM, Altwaim SA, Albohiri HH, Wakid MH, Mohamed K, Alshehri EA, Elshazly H, Elshabrawy HA, Hamed D. Administration of Trichinella spiralis Antigens Alleviated Diabetic Nephropathy in Diabetic Mice. Acta Parasitol 2025; 70:83. [PMID: 40178750 PMCID: PMC11968495 DOI: 10.1007/s11686-025-01016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/16/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a leading cause of end-stage renal disease (ESRD) among individuals with diabetes, highlighting the urgent need for effective therapeutic strategies to combat this condition. Prior research has indicated that T. spiralis possesses hypoglycemic properties. In this investigation, we aimed to evaluate the efficacy of T. spiralis antigens, derived from both adult and larval forms, in treating diabetic nephropathy in alloxan-induced diabetic mice (AIDM). METHODS A total of forty Swiss albino mice were allocated into four groups, each consisting of ten mice. Diabetes was induced in three of the groups using alloxan, while one group served as a control without diabetes. Two diabetic groups received treatment with either crude larva (CLA) antigen or adult worm antigen (AWA), while one group remained untreated. The study assessed various parameters, including fasting blood glucose levels, blood urea, serum creatinine, and serum albumin across all groups. Additionally, histopathological examinations of the kidneys were conducted. RESULTS The results indicated that treatment with CLA or AWA antigens led to a significant reduction in blood glucose, serum creatinine, and blood urea levels, alongside an increase in serum albumin. Notably, the administration of AWA antigens resulted in substantial improvements in renal pathological changes induced by diabetes, as evidenced by hematoxylin and eosin staining and Masson trichrome staining, which also demonstrated a reduction in fibrosis. CONCLUSIONS The findings suggest that T. spiralis antigens may mitigate renal damage in diabetic mice by alleviating hyperglycemia-induced inflammation and oxidative stress, warranting further investigation into their potential role in preventing DN in diabetic patients.
Collapse
Affiliation(s)
- Asmaa M El-Kady
- Department of Medical Parasitology, Faculty of Medicine, South Valley University, Qena, 83523, Egypt.
| | - Sarah A Altwaim
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Haleema H Albohiri
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, 21589, Saudi Arabia
| | - Majed H Wakid
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Khalil Mohamed
- Department of Epidemiology and Medical Statistics, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Eman Abdullah Alshehri
- Department of Zoology, College of Science, King Saud University, Riyadh, 11362, Saudi Arabia
| | - Hayam Elshazly
- Department of Biology, Faculty of Sciences-Scientific Departments, Qassim University, Buraidah, Qassim, 52571, Saudi Arabia
| | - Hatem A Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX, 77304, USA.
| | - Dina Hamed
- Department of Medical Parasitology, Faculty of Medicine, South Valley University, Qena, 83523, Egypt
| |
Collapse
|
3
|
Chaimon S, Phuphisut O, Reamtong O, Ampawong S, Fongsodsri K, Chantree P, Thanongsaksrikul J, Malaithong P, Sreesai S, Maleewong W, Sadaow L, Martviset P, Adisakwattana P. Molecular and biological characterization of transforming growth factor-β homolog derived from Trichinella spiralis. Sci Rep 2024; 14:31229. [PMID: 39732815 DOI: 10.1038/s41598-024-82599-x] [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: 07/30/2024] [Accepted: 12/06/2024] [Indexed: 12/30/2024] Open
Abstract
The cytokine homologs, particularly transforming growth factor (TGF)-β, is a crucial immunomodulatory molecule and involved in growth and developmental processes in several helminths. In this study, the basic properties and functions of T. spiralis TGF-β homolog 2 (TsTGH2) were characterized using bioinformatics and molecular biology approaches. Bioinformatics analyses indicated that TsTGH2 belongs to the TGF-β subfamily. Recombinant TsTGH2 (rTsTGH2) expressed in Escherichia coli was used to produce a polyclonal antibody (pAb) in mice. Western blot and immunolocalization using pAb detected native TsTGH2 in crude worm antigens from muscle larvae and adults, showing it was mainly localized in the body wall muscles and the epithelia of the ovary and uterus. To assess the interplay between TsTGH2 and the human TGF-β signaling pathway, rTsTGH2 produced in a HEK293T cell was incubated with the SBE luciferase-HEK293 cell. The result indicated a significant increase in luciferase activity after treatment with rTsTGH2 compared to untreated control (p < 0.05). In conclusion, these findings are the first to characterize the basic properties and functions of TGF-β homologs in T. spiralis, demonstrating their interaction with the human TGF-β receptor. Further investigation is required to identify and optimize an appropriate expression system or conditions for TsTGH2. Additionally, studies are needed to clarify the specific role of native TsTGH2 in parasite development and host immunomodulation.
Collapse
Affiliation(s)
- Salisa Chaimon
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
- Graduate Program in Applied Biosciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Orawan Phuphisut
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Kamonpan Fongsodsri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Pathanin Chantree
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
- Graduate Program in Applied Biosciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani, 12120, Thailand
| | - Jeeraphong Thanongsaksrikul
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12120, Thailand
- Thammasat University Research Unit in Molecular Pathogenesis and Immunology of Infectious Diseases, Thammasat University, Pathum Thani, 12120, Thailand
| | - Preeyarat Malaithong
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Suthasinee Sreesai
- Central Equipment Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Wanchai Maleewong
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Lakkhana Sadaow
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Pongsakorn Martviset
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
- Graduate Program in Applied Biosciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani, 12120, Thailand.
| | - Poom Adisakwattana
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
4
|
Heidari M, Maleki Vareki S, Yaghobi R, Karimi MH. Microbiota activation and regulation of adaptive immunity. Front Immunol 2024; 15:1429436. [PMID: 39445008 PMCID: PMC11496076 DOI: 10.3389/fimmu.2024.1429436] [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: 05/08/2024] [Accepted: 08/30/2024] [Indexed: 10/25/2024] Open
Abstract
In the mucosa, T cells and B cells of the immune system are essential for maintaining immune homeostasis by suppressing reactions to harmless antigens and upholding the integrity of intestinal mucosal barrier functions. Host immunity and homeostasis are regulated by metabolites produced by the gut microbiota, which has developed through the long-term coevolution of the host and the gut biome. This is achieved by the immunological system's tolerance for symbiote microbiota, and its ability to generate a proinflammatory response against invasive organisms. The imbalance of the intestinal immune system with commensal organisms is causing a disturbance in the homeostasis of the gut microbiome. The lack of balance results in microbiota dysbiosis, the weakened integrity of the gut barrier, and the development of inflammatory immune reactions toward symbiotic organisms. Researchers may uncover potential therapeutic targets for preventing or regulating inflammatory diseases by understanding the interactions between adaptive immunity and the microbiota. This discussion will explore the connection between adaptive immunity and microbiota.
Collapse
Affiliation(s)
- Mozhdeh Heidari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saman Maleki Vareki
- Department of Oncology, Western University, London, ON, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada
| | - Ramin Yaghobi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
5
|
El-kady AM, Altwaim SA, Wakid MH, Banjar AS, Mohammed K, Alfaifi MS, Elshazly H, Al-Megrin WAI, Alshehri EA, Sayed E, Elshabrawy HA. Prior Trichinella spiralis infection protects against Schistosoma mansoni induced hepatic fibrosis. Front Vet Sci 2024; 11:1443267. [PMID: 39439825 PMCID: PMC11494294 DOI: 10.3389/fvets.2024.1443267] [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: 06/03/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Background Schistosomiasis affects approximately 250 million people worldwide, with 200,000 deaths annually. It has been documented that the granulomatous response to Schistosoma mansoni (S. mansoni) oviposition is the root cause of progressive liver fibrosis in chronic infection, in 20% of the patients, and can lead to liver cirrhosis and/or liver cancer. The influence of helminths coinfection on schistosomiasis-induced liver pathological alterations remains poorly understood. Therefore, in this study, we investigated the effect of Trichinella spiralis (T. spiralis) infection on S. mansoni-induced hepatic fibrosis. Materials and methods Thirty adult male Balb-c mice were divided into three groups. Group 1 was left uninfected; group 2 was infected with S. mansoni cercariae and group 3 was orally infected with T. spiralis larvae, then 28 days later, this group was infected with S. mansoni cercariae. All groups were sacrificed at the end of the 8th week post infection with S. mansoni to evaluate the effect of pre-infection with T. spiralis on S. mansoni induced liver fibrosis was evaluated parasitologically (worm burden and egg count in tissues), biochemically (levels of alanine aminotransferase and aspartate aminotransferase), histopathologically (H&E and MT staining, and immunohistochemical staining for the expression of α-SMA, IL-6, IL-1β, IL-17, IL-23, TNF-α, and TGF-β). Results The results in the present study demonstrated marked protective effect of T. spiralis against S. mansoni induced liver pathology. We demonstrated that pre-infection with T. spirais caused marked reduction in the number of S. mansoni adult worms (3.17 ± 0.98 vs. 18 ± 2.16, P = 0.114) and egg count in both the intestine (207.2 ± 64.3 vs. 8,619.43 ± 727.52, P = 0.009) and liver tissues (279 ± 87.2 vs. 7,916.86 ± 771.34; P = 0.014). Consistently, we found significant reductions in both number (3.4 ± 1.1 vs. 11.8.3 ± 1.22; P = 0.007) and size (84 ± 11 vs. 294.3 ± 16.22; P = 0.001) of the hepatic granulomas in mice pre-infected with T. spiralis larvae compared to those infected with only S. mansoni. Furthermore, pre- infection with T. spiralis markedly reduced S. mansoni- induced hepatic fibrosis, as evidenced by decreased collagen deposition, low expression of α-SMA, and significantly reduced levels of IL-17, IL-1B, IL-6, TGF-B, IL-23, and TNF-α compared to mice infected with S. mansoni only. Conclusions Our data show that pre-infection with T. spiralis effectively protected mice from severe schistosomiasis and liver fibrosis. We believe that our findings support the potential utility of helminths for the preventing and ameliorating severe pathological alterations induced by schistosomiasis.
Collapse
Affiliation(s)
- Asmaa M. El-kady
- Department of Medical Parasitology, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Sarah A. Altwaim
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, Jeddah, Saudi Arabia
| | - Majed H. Wakid
- Special Infectious Agents Unit, King Fahd Medical Research Center, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alaa S. Banjar
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khalil Mohammed
- Department of Epidemiology and Medical Statistics, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Mashael S. Alfaifi
- Department of Epidemiology and Medical Statistics, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Hayam Elshazly
- Department of Biology, Faculty of Sciences-Scientific Departments, Qassim University, Buraidah, Qassim, Saudi Arabia
- Department of Zoology, Faculty of Science, Beni-Suef University, Beni Suef, Egypt
| | - Wafa Abdullah I. Al-Megrin
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | - Eman Sayed
- Department of Parasitology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Hatem A. Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX, United States
| |
Collapse
|
6
|
Grondin JA, Jamal A, Mowna S, Seto T, Khan WI. Interaction between Intestinal Parasites and the Gut Microbiota: Implications for the Intestinal Immune Response and Host Defence. Pathogens 2024; 13:608. [PMID: 39204209 PMCID: PMC11356857 DOI: 10.3390/pathogens13080608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024] Open
Abstract
Intestinal parasites, including helminths and protozoa, account for a significant portion of the global health burden. The gastrointestinal (GI) tract not only serves as the stage for these parasitic infections but also as the residence for millions of microbes. As the intricacies of the GI microbial milieu continue to unfold, it is becoming increasingly apparent that the interactions between host, parasite, and resident microbes help dictate parasite survival and, ultimately, disease outcomes. Across both clinical and experimental models, intestinal parasites have been shown to impact microbial composition and diversity. Reciprocally, microbes can directly influence parasitic survival, colonization and expulsion. The gut microbiota can also indirectly impact parasites through the influence and manipulation of the host. Studying this host-parasite-microbiota axis may help bring about novel therapeutic strategies for intestinal parasitic infection as well as conditions such as inflammatory bowel disease (IBD). In this review, we explore the relationship between intestinal parasites, with a particular focus on common protozoa and helminths, and the gut microbiota, and how these interactions can influence the host defence and intestinal immune response. We will also explore the impact of this tripartite relationship in a clinical setting and its broader implications for human health.
Collapse
Affiliation(s)
- Jensine A. Grondin
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; (J.A.G.); (A.J.); (S.M.); (T.S.)
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Asif Jamal
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; (J.A.G.); (A.J.); (S.M.); (T.S.)
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sadrina Mowna
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; (J.A.G.); (A.J.); (S.M.); (T.S.)
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Tyler Seto
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; (J.A.G.); (A.J.); (S.M.); (T.S.)
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Waliul I. Khan
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; (J.A.G.); (A.J.); (S.M.); (T.S.)
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
7
|
Chakraborty A, Bayry J, Mukherjee S. Helminth-derived biomolecules as potential therapeutics against ulcerative colitis. Immunotherapy 2024; 16:635-640. [PMID: 38888436 PMCID: PMC11404699 DOI: 10.1080/1750743x.2024.2360382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Affiliation(s)
- Ankita Chakraborty
- Integrative Biochemistry & Immunology Laboratory (IBIL), Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, 713340, India
| | - Jagadeesh Bayry
- Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad, Kerala, 678623, India
| | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory (IBIL), Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, 713340, India
| |
Collapse
|
8
|
Noel S, LaFrancois R, Scott ME. Gastrointestinal nematode infection during pregnancy and lactation enhances spatial reference memory and reduces indicators of anxiety-like behaviour in uninfected adult female mouse offspring. Parasitology 2024; 151:722-731. [PMID: 38808523 PMCID: PMC11474017 DOI: 10.1017/s0031182024000696] [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: 03/06/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
Maternal bacterial and viral infections that induce neuroinflammation in the developing brain are associated with impaired cognitive function and increased anxiety in the offspring. In contrast, maternal infection with the immunoregulatory murine gastrointestinal (GI) nematode, Heligmosomoides bakeri, appears to benefit neurodevelopment as juvenile 2- and 3-week-old male and female offspring had enhanced spatial memory, which may be due to a Th2/Treg biased neuroimmune environment. Here, the impact of maternal H. bakeri infection during pregnancy and lactation on the spatial and anxiety-like behaviours of adult, 3-month-old uninfected male and female offspring was explored for the first time. It was observed that adult female offspring of H. bakeri-infected dams had enhanced spatial reference memory and reduced anxiety-like behaviour compared to females of uninfected dams. These effects were not observed in adult male offspring. Thus, the positive influence of a maternal GI nematode infection on spatial memory of juvenile offspring persists in adult female offspring.
Collapse
Affiliation(s)
- Sophia Noel
- Institute of Parasitology, McGill University (Macdonald Campus), Quebec H9X 3V9, Canada
| | - Ryan LaFrancois
- Institute of Parasitology, McGill University (Macdonald Campus), Quebec H9X 3V9, Canada
| | - Marilyn E. Scott
- Institute of Parasitology, McGill University (Macdonald Campus), Quebec H9X 3V9, Canada
| |
Collapse
|
9
|
Singh I, Kanichery A, Kotimoole CN, Modi PK, Prasad TSK, Hoti SL. Unpacking Immune Modulation as a Site of Therapeutics Innovation for Nematode Parasite Wuchereria bancrofti: A Temporal Quantitative Phosphoproteomics Profiling of Macrophage Migration Inhibitory Factor 2. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:125-137. [PMID: 38527276 DOI: 10.1089/omi.2024.0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Nematode infections are common in both humans and livestock, with major adverse planetary health and economic impacts. Wuchereria bancrofti is a parasitic nematode that causes lymphatic filariasis, a neglected tropical disease that can lead to severe disability and deformity worldwide. For the long-term survival of the bancroftian parasites in the host, a complex immune invasion strategy is involved through immunomodulation. Therefore, immunomodulation can serve as a site of research and innovation for molecular targets. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine crucial to the host antimicrobial alarm system and stress response. Interestingly, the nematode parasite W. bancrofti also produces two homologs of MIF (Wba-MIF1 and 2). Using a mass spectrometry-based phosphoproteomics approach, we report new findings on the immunomodulatory effect and signaling mechanism of Wba-MIF2 in macrophage cells. Accordingly, we observed 1201 phosphorylated sites on 467 proteins. Out of the 1201 phosphorylated sites, 1075, 117, and 9 were found on serine (S), threonine (T), and tyrosine (Y) residues, respectively. Our bioinformatics analysis led to identification of major pathways, including spliceosomes, T cell receptor signaling pathway, Th17 differentiation pathway, interleukin-17 signaling pathway, and insulin signaling pathway upon Wba-MIF2 treatment. Wba-MIF2 treatment also enriched CDK4, CDK1, and DNAPK kinases. The comparison of the signaling pathway of Wba-MIF2 with that of human-MIF suggests both share similar signaling pathways. These findings collectively offer new insights into the role and mechanism of Wba-MIF2 as an immunomodulator and inform future diagnostics and drug discovery research for W. bancrofti.
Collapse
Affiliation(s)
- Ishwar Singh
- Department of Neglected Tropical Diseases and Translational Research, ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, India
- Department of the Interdisciplinary Science, KLE Academy of Higher Education and Research, (Deemed to be University), Belagavi, Karnataka, India
| | - Anagha Kanichery
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Chinmaya Narayana Kotimoole
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | | | - Sugeerappa Laxamannappa Hoti
- Department of Neglected Tropical Diseases and Translational Research, ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, India
| |
Collapse
|
10
|
Long SR, Shang WX, Zhang HR, Jiang M, Wang JJ, Liu RD, Wang ZQ, Cui J, Sun H. Trichinella-derived protein ameliorates colitis by altering the gut microbiome and improving intestinal barrier function. Int Immunopharmacol 2024; 127:111320. [PMID: 38064817 DOI: 10.1016/j.intimp.2023.111320] [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: 08/24/2023] [Revised: 10/29/2023] [Accepted: 11/27/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) encompasses Crohn's Disease and Ulcerative Colitis. Reports have highlighted the potential use of helminths or their byproducts as a possible treatment for IBD; however, the mechanisms underlying their ability to modulate inflammation remain incompletely understood. In the present study, we analyze the possible mechanism of a serine protease inhibitor from adult T. spiralis excretion-secretion products (rTsSPI) on the improvement of colitis. METHODS The immune protective effect of rTsSPI was studied by using DSS or Salmonella-induced colitis in female C56BL/6 mice. The effect of rTsSPI on the immune and inflammatory responses, gut microbiota, permeability of colon epithelium and junction proteins was analyzed. RESULTS Treating mice with rTsSPI induced type 2 immunity and significantly attenuated clinical symptoms, macroscopical and histological features of DSS or bacteria-induced colonic inflammation. This was accompanied by decreasing neutrophil recruitment in the colonic lamina propria, and reducing TNF-α mRNA levels in the colon; in contrast, the recruitment of M2 macrophages, the expression level of IL-10 and adhesion molecules increased in the colon tissue. Moreover, treatment with rTsSPI led to an improvement in gut microbiota diversity, as well as an increase in the abundance of the bacterial genera Bifidobacterium and Ruminclostridium 5. CONCLUSIONS Collective findings suggest that pretreatment with rTsSPI can ameliorate colitis in mice by inducing a Th2-type response with M2 macrophages. Data also indicate that immunotherapy with rTsSPI represents an additional strategy to ameliorate inflammatory processes in IBD by enhancing probiotic colonization and maintaining intestinal epithelial barrier function.
Collapse
Affiliation(s)
- Shao Rong Long
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Wen Xuan Shang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Hui Ran Zhang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Miao Jiang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jing Jing Wang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jing Cui
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Hualei Sun
- Department of Nutrition, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
11
|
Zhang QZ, Liu JH, Gao YR, Liang J, Tang CL. Effect of macrophage polarization on parasitic protection against type 1 diabetes mellitus. Exp Parasitol 2024; 256:108649. [PMID: 37914152 DOI: 10.1016/j.exppara.2023.108649] [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: 06/21/2023] [Revised: 10/06/2023] [Accepted: 10/29/2023] [Indexed: 11/03/2023]
Abstract
Type 1 diabetes mellitus is a chronic disease caused by the destruction of pancreatic beta cells. Based on the hygiene hypothesis, a growing body of evidence suggests a negative association between parasitic infections and diabetes in humans and animal models. The mechanism of parasite-mediated prevention of type 1 diabetes mellitus may be related to the adaptive and innate immune systems. Macrophage polarization is a new paradigm for the treatment of type 1 diabetes mellitus, and different host macrophage subsets play various roles during parasite infection. Proinflammatory cytokines are released by M1 macrophages, which are important in the development of type 1 diabetes mellitus. Parasite-activated M2 macrophages prevent the development of type 1 diabetes mellitus and can influence the development of adaptive immune responses through several mechanisms, including Th2 cells and regulatory T cells. Here, we review the role and mechanism of macrophage polarization in parasitic protection against type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Qi-Zhi Zhang
- Wuchang Hospital Affiliated with Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Jun-Hui Liu
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430030, China
| | - Yan-Ru Gao
- Basic Medical Science Teaching Center, Medical Department, Wuhan City College, Wuhan, 430083, China
| | - Jun Liang
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430030, China.
| | - Chun-Lian Tang
- Wuchang Hospital Affiliated with Wuhan University of Science and Technology, Wuhan, 430063, China.
| |
Collapse
|
12
|
Bertrand L, Lehuen A. Lymph node sharing is caring for pancreatic immunity. Immunity 2023; 56:1980-1982. [PMID: 37703826 DOI: 10.1016/j.immuni.2023.08.016] [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: 08/07/2023] [Revised: 08/17/2023] [Accepted: 08/17/2023] [Indexed: 09/15/2023]
Abstract
Lymph nodes can be shared among several organs, notably in the gastrointestinal system. In this issue of Immunity, Brown et al. describe how pancreatic immunity is shaped by the mixing of different migratory dendritic cells issued from co-drainage from liver, pancreas, and duodenum.
Collapse
Affiliation(s)
- Léo Bertrand
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratoire d'Excellence Inflamex, F-75014, Paris, France
| | - Agnès Lehuen
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratoire d'Excellence Inflamex, F-75014, Paris, France.
| |
Collapse
|
13
|
Wang X, Shen J, Xu C, Wan C, Yang H, Qiu Y, Xu M, Duo W, Sun T, Cui J, Chu L, Yang X. Proteomic profile of Trichinella spiralis infected mice with acute spinal cord injury: A 4D label-free quantitative analysis. Comp Immunol Microbiol Infect Dis 2023; 97:101994. [PMID: 37207504 DOI: 10.1016/j.cimid.2023.101994] [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: 03/04/2023] [Revised: 04/25/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023]
Abstract
Spinal cord injury (SCI) can cause severe loss of locomotor and sensory activities, with no ideal treatment. Emerging reports suggest that the helminth therapy is highly effective in relieving numerous inflammatory diseases. Proteomic profiling is often used to elucidate the underlying mechanism behind SCI. Herein, we systematically compared the protein expression profiles of murine SCI spinal cord and Trichinella spiralis treated murine SCI spinal cord, using a 4D label-free technique known for its elevated sensitivity. Relative to the SCI mice, the T. spiralis-treated mice exhibited marked alterations in 91 proteins (31 up- and 60 down-regulated). Based on our Gene Ontology (GO) functional analysis, the differentially expressed proteins (DEPs) were primarily enriched in the processes of metabolism, biological regulation, cellular process, antioxidant activity, and other cell functions. In addition, according to the Clusters of Orthologous Groups of protein/EuKaryotic Orthologous Groups (COG/KOG) functional stratification, proteins involved in signaling transduction mechanisms belonged to the largest category. Over-expressed DEPs were also enriched in the "NADPH oxidase complex", "superoxide anion generation", "other types of O-glycan biosynthesis", and "HIF-1 signaling pathway". Furthermore, the protein-protein interaction (PPI) network identified the leading 10 hub proteins. In conclusion, we highlighted the dynamic proteomic profiling of T. spiralis-treated SCI mice. Our findings provide significant insight into the molecular mechanism behind T. spiralis regulation of SCI.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| | - Junhong Shen
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| | - Changyan Xu
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| | - Chen Wan
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| | - Haoyu Yang
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| | - Yu Qiu
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| | - Mengmeng Xu
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| | - Wenjuan Duo
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| | - Tongjun Sun
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| | - Jie Cui
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| | - Liang Chu
- Second Affiliated Hospital of Bengbu Medical College, Bengbu, China.
| | - Xiaodi Yang
- Department of Microbiology and Parasitology, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunology, Bengbu Medical College, Bengbu, China.
| |
Collapse
|
14
|
Chakraborty P, Aravindhan V, Mukherjee S. Helminth-derived biomacromolecules as therapeutic agents for treating inflammatory and infectious diseases: What lessons do we get from recent findings? Int J Biol Macromol 2023; 241:124649. [PMID: 37119907 DOI: 10.1016/j.ijbiomac.2023.124649] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Despite the tremendous progress in healthcare sectors, a number of life-threatening infectious, inflammatory, and autoimmune diseases are continuously challenging mankind throughout the globe. In this context, recent successes in utilizing helminth parasite-derived bioactive macromolecules viz. glycoproteins, enzymes, polysaccharides, lipids/lipoproteins, nucleic acids/nucleotides, and small organic molecules for treating various disorders primarily resulted from inflammation. Among the several parasites that infect humans, helminths (cestodes, nematodes, and trematodes) are known as efficient immune manipulators owing to their explicit ability to modulate and modify the innate and adaptive immune responses of humans. These molecules selectively bind to immune receptors on innate and adaptive immune cells and trigger multiple signaling pathways to elicit anti-inflammatory cytokines, expansion of alternatively activated macrophages, T-helper 2, and immunoregulatory T regulatory cell types to induce an anti-inflammatory milieu. Reduction of pro-inflammatory responses and repair of tissue damage by these anti-inflammatory mediators have been exploited for treating a number of autoimmune, allergic, and metabolic diseases. Herein, the potential and promises of different helminths/helminth-derived products as therapeutic agents in ameliorating immunopathology of different human diseases and their mechanistic insights of function at cell and molecular level alongside the molecular signaling cross-talks have been reviewed by incorporating up-to-date findings achieved in the field.
Collapse
Affiliation(s)
- Pritha Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India
| | | | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India.
| |
Collapse
|
15
|
Khueangchiangkhwang S, Wu Z, Nagano I, Maekawa Y. Trichinella pseudospiralis-secreted 53 kDa protein ameliorates imiquimod-induced psoriasis by inhibiting the IL-23/IL-17 axis in mice. Biochem Biophys Rep 2022; 33:101415. [PMID: 36620087 PMCID: PMC9813687 DOI: 10.1016/j.bbrep.2022.101415] [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: 09/14/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Trichinella infection can experimentally ameliorate many autoimmune diseases. However, the immune mechanism of the amelioration and the identification of corresponding Trichinella-derived molecule(s) are still not fully elucidated. Fifty-three kDa excretory-secretory (ES) protein from Trichinella pseudospiralis (Tpp53) is a molecule like TsP53 reported as a protein exerting immune-inhibitory effect in T. spiralis. In this study, we investigated the immunomodulatory effect of Tpp53 using imiquimod (IMQ)-induced psoriasis-like dermatitis model, which is a mouse model of autoimmune disease with the pathogenic interleukin 17 (IL-17) producing CD4+ T cells (Th17) via IL-23/IL17 axis. Administrating the recombinant Tpp53 (rTpp53) mixed with IMQ cream on the skin of mice ameliorated psoriatic lesions, as revealed by the improvement of erythema, scaling, skin thickening, epidermis hyperplasia and parakeratosis, thickening of acanthosis cell layer, epidermal extension of dermis, less infiltration of inflammatory cells, and decreased expression of inflammatory marker. The increased expression of the factors related to the IL-23/IL-17 axis, including IL-17A, IL-6, Il17F and Il23a, in the skins of IMQ-treated mice was inhibited by rTpp53 treatment. Moreover, the expression of activated keratinocyte-produced cytokines, chemokines, and antimicrobial peptides in the skin was also down-regulated in rTpp53-treated IMQ-treated mice. Co-culture of splenocytes with rTpp53 inhibited IL-17A and treatment of macrophages with rTpp53 reduced IL-6 production. Overall, our study revealed that the Trichinella-secreted 53 kDa ES protein could ameliorate IMQ-induced psoriasis by inhibiting the IL-23/IL-17 axis, suggesting that Tpp53 might involve in regulating host Th17 for immune evasion and have an alternative potential for psoriasis therapy.
Collapse
Affiliation(s)
| | - Zhiliang Wu
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan,Cocorresponding author. 1-1 Yanagido, Gifu, 501-1194, Japan.
| | - Isao Nagano
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yoichi Maekawa
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan,Preemptive Food Research Center, Gifu University, Gifu, Japan,Corresponding author. 1-1 Yanagido, Gifu, 501-1194, Japan.
| |
Collapse
|
16
|
Dalmolin SP, Pedó RT, da Rosa TH, de Souza Silva JM, Farinon M, Gasparini ML, Chiela ECF, Paz AH, Sehabiague MPC, Ferreira HB, do Espírito Santo RC, da Costa Gonçalves F, Xavier RM. Fasciola hepatica extract suppresses fibroblast-like synoviocytes in vitro and alleviates experimental arthritis. Adv Rheumatol 2022; 62:43. [PMID: 36371346 DOI: 10.1186/s42358-022-00275-y] [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: 05/16/2022] [Accepted: 10/29/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by synovial inflammation, fibroblast-like synoviocytes (FLS) activation and joint destruction. Fasciola hepatica is a platyhelminth that releases excretory-secretory immunomodulatory products capable of suppressing the Th1 immune response. Despite the effectiveness of available treatments for inducing disease remission, current options are not successful in all patients and may cause side effects. Thus, we evaluated the therapeutic potential of F. hepatica extract on FLS from RA patients and arthritis models. METHODS FLS were isolated from synovial fluid of RA patients, cultured, and exposed to F. hepatica extract (60, 80, and 100 µg/ml) for different time points to assess cell viability, adherence, migration and invasion. For in vivo experiments, mice with antigen (AIA) and collagen (CIA) induced arthritis received a 200 µg/dose of F. hepatica extract daily. Statistical analysis was performed by ANOVA and Student's t-test using GraphPad Prism 6.0. RESULTS In vitro assays showed that extract decreased FLS cell viability at concentration of 100 µg/ml (83.8% ± 5.0 extract vs. 100.0% ± 0.0 control; p < 0.05), adherence in 20% (92.0 cells ± 5.8 extract vs. 116.3 cells ± 7.9 control; p < 0.05), migratory potential (69.5% ± 17.6 extract vs. 100.0% control; p < 0.05), and cell invasiveness potential through the matrigel (76.0% ± 8.4 extract vs. 100.0% control; p < 0.01). The extract reduced leukocyte migration by 56% (40 × 104 leukocytes/knee ± 19.00) compared to control (90.90 × 104 leukocytes/knee ± 12.90) (p < 0.01) and nociception (6.37 g ± 0.99 extract vs. 3.81 g ± 1.44 control; p < 0.001) in AIA and delayed clinical onset of CIA (11.75 ± 2.96 extract vs. 14.00 ± 2.56 control; p = 0.126). CONCLUSION Our results point out a potential immunomodulatory effect of F. hepatica extract in RA models. Therefore, the characterization of promising new immunomodulatory molecules should be pursued, as they can promote the development of new therapies. Trial registration Collection of synovial liquid and in vitro procedures were approved by the Ethics Committee with Certificate of Presentation of Ethical Appreciation in Plataforma Brasil (CAAE: 89044918.8.0000.5327; date of registration: 26/07/2018).
Collapse
Affiliation(s)
- Suelen Pizzolatto Dalmolin
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Renata Ternus Pedó
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Thales Hein da Rosa
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jordana Miranda de Souza Silva
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mirian Farinon
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Maria Luísa Gasparini
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduardo Cremonese Filippi Chiela
- Programa de Pós-Graduação Ciências em Gastroenterologia e Hepatologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Ciências Morfológicas, ICBS, Serviço de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Centro de Pesquisas Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Ana Helena Paz
- Programa de Pós-Graduação Ciências em Gastroenterologia e Hepatologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Ciências Morfológicas, ICBS, Serviço de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Centro de Pesquisas Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Martín Pablo Cancela Sehabiague
- Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Henrique Bunselmeyer Ferreira
- Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafaela Cavalheiro do Espírito Santo
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fabiany da Costa Gonçalves
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, GD 3015, Rotterdam, The Netherlands.
| | - Ricardo Machado Xavier
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil. .,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil. .,Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350; 6º Andar, Porto Alegre, RS, 90035-903, Brazil.
| |
Collapse
|
17
|
Majumdar S, Lin Y, Bettini ML. Host-microbiota interactions shaping T-cell response and tolerance in type 1 diabetes. Front Immunol 2022; 13:974178. [PMID: 36059452 PMCID: PMC9434376 DOI: 10.3389/fimmu.2022.974178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 11/29/2022] Open
Abstract
Type-1 Diabetes (T1D) is a complex polygenic autoimmune disorder involving T-cell driven beta-cell destruction leading to hyperglycemia. There is no cure for T1D and patients rely on exogenous insulin administration for disease management. T1D is associated with specific disease susceptible alleles. However, the predisposition to disease development is not solely predicted by them. This is best exemplified by the observation that a monozygotic twin has just a 35% chance of developing T1D after their twin's diagnosis. This makes a strong case for environmental triggers playing an important role in T1D incidence. Multiple studies indicate that commensal gut microbiota and environmental factors that alter their composition might exacerbate or protect against T1D onset. In this review, we discuss recent literature highlighting microbial species associated with T1D. We explore mechanistic studies which propose how some of these microbial species can modulate adaptive immune responses in T1D, with an emphasis on T-cell responses. We cover topics ranging from gut-thymus and gut-pancreas communication, microbial regulation of peripheral tolerance, to molecular mimicry of islet antigens by microbial peptides. In light of the accumulating evidence on commensal influences in neonatal thymocyte development, we also speculate on the link between molecular mimicry and thymic selection in the context of T1D pathogenesis. Finally, we explore how these observations could inform future therapeutic approaches in this disease.
Collapse
Affiliation(s)
- Shubhabrata Majumdar
- Immunology Graduate Program, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology, University of Utah, Salt Lake City, UT, United States
| | - Yong Lin
- Immunology Graduate Program, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology, University of Utah, Salt Lake City, UT, United States
| | - Matthew L. Bettini
- Department of Pathology, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
18
|
Huang H, Hu D, Chen Z, Xu J, Xu R, Gong Y, Fang Z, Wang T, Chen W. Immunotherapy for type 1 diabetes mellitus by adjuvant-free Schistosoma japonicum-egg tip-loaded asymmetric microneedle patch (STAMP). J Nanobiotechnology 2022; 20:377. [PMID: 35964125 PMCID: PMC9375265 DOI: 10.1186/s12951-022-01581-9] [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: 03/03/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
Background Type 1 diabetes mellitus (T1DM) is an autoimmune disease mediated by autoreactive T cells and dominated by Th1 response polarization. Insulin replacement therapy faces great challenges to this autoimmune disease, requiring highly frequent daily administration. Intriguingly, the progression of T1DM has proven to be prevented or attenuated by helminth infection or worm antigens for a relatively long term. However, the inevitable problems of low safety and poor compliance arise from infection with live worms or direct injection of antigens. Microneedles would be a promising candidate for local delivery of intact antigens, thus providing an opportunity for the clinical immunotherapy of parasitic products. Methods We developed a Schistosoma japonicum-egg tip-loaded asymmetric microneedle patch (STAMP) system, which serves as a new strategy to combat TIDM. In order to improve retention time and reduce contamination risk, a specific imperfection was introduced on the STAMP (asymmetric structure), which allows the tip to quickly separate from the base layer, improving reaction time and patient’s comfort. After loading Schistosoma japonicum-egg as the immune regulator, the effects of STAMP on blood glucose control and pancreatic pathological progression improvement were evaluated in vivo. Meanwhile, the immunoregulatory mechanism and biosafety of STAMP were confirmed by histopathology, qRT-PCR, ELISA and Flow cytometric analysis. Results Here, the newly developed STAMP was able to significantly reduce blood glucose and attenuate the pancreatic injury in T1DM mice independent of the adjuvants. The isolated Schistosoma japonicum-eggs micron slowly degraded in the skin and continuously released egg antigen for at least 2 weeks, ensuring localization and safety of antigen stimulation. This phenomenon should be attributed to the shift of Th2 immune response to reduce Th1 polarization. Conclusion Our results exhibited that STAMP could significantly regulate the blood glucose level and attenuate pancreatic pathological injury in T1DM mice by balancing the Th1/Th2 immune responses, which is independent of adjuvants. This technology opens a new window for the application of parasite products in clinical immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01581-9.
Collapse
Affiliation(s)
- Haoming Huang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Dian Hu
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhuo Chen
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jiarong Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Rengui Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yusheng Gong
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhengming Fang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ting Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Wei Chen
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Hubei Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
19
|
Bruschi F, Ashour D, Othman A. Trichinella-induced immunomodulation: Another tale of helminth success. Food Waterborne Parasitol 2022; 27:e00164. [PMID: 35615625 PMCID: PMC9125654 DOI: 10.1016/j.fawpar.2022.e00164] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 01/01/2023] Open
Abstract
Trichinella spiralis is a unique parasite in that both the adults and larvae survive in two different intracellular niches in the same host. The immune response, albeit intense, is highly modulated to ensure the survival of both the host and the parasite. It is skewed to T helper 2 and regulatory arms. Diverse cells from both the innate and adaptive compartments of immunity, including dendritic cells, T regulatory cells, and alternatively activated macrophages are thought to mediate such immunomodulation. The parasite has also an outstanding ability to evade the immune system by several elaborate processes. The molecules derived from the parasites including Trichinella, particularly the components of the excretory-secretory products, are being continually identified and explored for the potential of ameliorating the immunopathology in animal models of diverse inflammatory and autoimmune human diseases. Herein we discuss the various aspects of Trichinella-induced immunomodulation with a special reference to the practical implications of the immune system manipulation in alleviating or possibly curing human diseases.
Collapse
Key Words
- AAM, alternatively activated macrophage
- AW, adult worm
- Allergy
- Autoimmune diseases
- Breg, regulatory B cell
- CAM, classically activated macrophage
- Cancer
- ES L1, ES product of T. spiralis muscle larva
- ES, excretory–secretory
- IFN- γ, interferon-γ
- IIL, intestinal infective larva
- IL, interleukin
- Immune evasion
- Immunomodulation
- ML, muscle larva
- NBL, newborn larva
- NOS, nitric oxide synthase
- TGF-β, transforming growth factor-β
- TLR, toll-like receptor
- TNF- α, tumor necrosis factor-α
- Th, T helper
- Tol-DC, tolerogenic dendritic cell
- Treg, regulatory T cell
- Trichinella
- Trichinella-derived molecules
- Ts-AES, ES from adult T. spiralis
Collapse
Affiliation(s)
- F. Bruschi
- School of Medicine, Department of Translational Research, N.T.M.S., Università di Pisa, Pisa, Italy
| | - D.S. Ashour
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - A.A. Othman
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
20
|
Ruenchit P, Reamtong O, Khowawisetsut L, Adisakwattana P, Chulanetra M, Kulkeaw K, Chaicumpa W. Peptide of Trichinella spiralis Infective Larval Extract That Harnesses Growth of Human Hepatoma Cells. Front Cell Infect Microbiol 2022; 12:882608. [PMID: 35558100 PMCID: PMC9086976 DOI: 10.3389/fcimb.2022.882608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/28/2022] [Indexed: 11/15/2022] Open
Abstract
Trichinella spiralis, a tissue-dwelling helminth, causes human trichinellosis through ingestion of undercooked meat containing the parasite’s infective larvae. However, benefits from T. spiralis infection have been documented: reduction of allergic diseases, inhibition of collagen-induced arthritis, delay of type 1 diabetes progression, and suppression of cancer cell proliferation. Since conventional cancer treatments have limited and unreliable efficacies with adverse side effects, novel adjunctive therapeutic agents and strategies are needed to enhance the overall treatment outcomes. This study aimed to validate the antitumor activity of T. spiralis infective larval extract (LE) and extricate the parasite-derived antitumor peptide. Extracts of T. spiralis infective larvae harvested from striated muscles of infected mice were prepared and tested for antitumor activity against three types of carcinoma cells: hepatocellular carcinoma HepG2, ovarian cancer SK-OV-3, and lung adenocarcinoma A549. The results showed that LE exerted the greatest antitumor effect on HepG2 cells. Proteomic analysis of the LE revealed 270 proteins. They were classified as cellular components, proteins involved in metabolic processes, and proteins with diverse biological functions. STRING analysis showed that most LE proteins were interconnected and played pivotal roles in various metabolic processes. In silico analysis of anticancer peptides identified three candidates. Antitumor peptide 2 matched the hypothetical protein T01_4238 of T. spiralis and showed a dose-dependent anti-HepG2 effect, not by causing apoptosis or necrosis but by inducing ROS accumulation, leading to inhibition of cell proliferation. The data indicate the potential application of LE-derived antitumor peptide as a complementary agent for human hepatoma treatment.
Collapse
Affiliation(s)
- Pichet Ruenchit
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Ladawan Khowawisetsut
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Poom Adisakwattana
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Monrat Chulanetra
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kasem Kulkeaw
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanpen Chaicumpa
- Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- *Correspondence: Wanpen Chaicumpa,
| |
Collapse
|
21
|
Helminth protection against type-1 diabetes: an insight into immunomodulatory effect of helminth-induced infection. Mol Biol Rep 2021; 48:6581-6588. [PMID: 34432219 DOI: 10.1007/s11033-021-06663-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
Helminths are the old dirty friends of humans from decades and may live undetected by the immune system for years in the tissues. They have evolved as good experts at subverting the immune system. Despite of their pathogenicity, they provide protection to their host against certain inflammatory diseases such as diabetes by modulating the immune mechanisms. These parasites are extra-cellular and induce Th2 response which triggers the adaptive immune cells as well as innate immune cells to work synergistically allowing Tregs to work in a toll-like receptor-dependent manure. T-helper cells type-2 also secrete certain anti-inflammatory cytokines including IL-4, IL-10, IL-13 and TGF-β which also provide protection against type-1 diabetes. Several helminths such as T. crassiceps, S. venezuelensis, filarial worms, Schistosoma spp. and T. spiralis have been reported to prevent diabetes in mouse models as well as in some clinical trials. Immunomodulatory talent of helminths is receiving greater attention to prevent diabetes. Herein, an attempt has been made to review and highlight the possible immuno-modulatory mechanisms by which helminths provide protection against diabetes. Moreover, this review also emphasizes on the use of helminth-derived molecules or synthetic derivatives of helminth-antigens in clinical trials to overcome rapidly growing autoimmune disorders including diabetes.
Collapse
|
22
|
Ilić N, Kosanović M, Gruden-Movsesijan A, Glamočlija S, Sofronić-Milosavljević L, Čolić M, Tomić S. Harnessing immunomodulatory mechanisms of Trichinella spiralis to design novel nanomedical approaches for restoring self-tolerance in autoimmunity. Immunol Lett 2021; 238:57-67. [PMID: 34363897 DOI: 10.1016/j.imlet.2021.04.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/28/2021] [Accepted: 04/28/2021] [Indexed: 01/13/2023]
Abstract
The rapid increase in the prevalence of autoimmune diseases in recent decades, especially in developed countries, coincided with improved living conditions and healthcare. Part of this increase could be ascribed to the lack of exposure to infectious agents like helminths that co-evolved with us and display potent immune regulatory actions. In this review we discussed many investigations, including our own, showing that Trichinella spiralis via its excretory-secretory products attenuate Th1/Th17 immunopathological response in autoimmunity and potentiate the protective Th2 and or regulatory T cell response, acting as an effective induction of tolerogenic dendritic cells (DCs), and probably mimicking the autoantigen in some diseases. A recent discovery of T. spiralis extracellular vesicles (TsEVs) suggested that inducing a complex regulation of the immune response requires simultaneous delivery of different signals in nano-sized packages. Indeed, different artificial nanomedical approaches discussed here suggested that co-delivery of multiple signals via nanoparticles is the most promising strategy for the treatment of autoimmune diseases. Although a long way is ahead of us before we could completely replicate natural nano-delivery systems which are both safe and potent in restoring self-tolerance, a clear path is being opened from a careful examination of parasite-host interactions.
Collapse
Affiliation(s)
- Nataša Ilić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia
| | - Maja Kosanović
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia
| | - Alisa Gruden-Movsesijan
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia
| | - Sofija Glamočlija
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia
| | - Ljiljana Sofronić-Milosavljević
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia
| | - Miodrag Čolić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia; Medical Faculty Foča, University of East Sarajevo, Bosnia and Hercegovina; Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Sergej Tomić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia.
| |
Collapse
|
23
|
Wu Z, Nagano I, Khueangchiangkhwang S, Maekawa Y. Proteomics of Trichinella. TRICHINELLA AND TRICHINELLOSIS 2021:103-183. [DOI: 10.1016/b978-0-12-821209-7.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
24
|
DC-SIGN signalling induced by Trichinella spiralis products contributes to the tolerogenic signatures of human dendritic cells. Sci Rep 2020; 10:20283. [PMID: 33219293 PMCID: PMC7679451 DOI: 10.1038/s41598-020-77497-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/11/2020] [Indexed: 12/28/2022] Open
Abstract
Tolerogenic dendritic cells (tolDCs) are central players in the maintenance of immune tolerance and thereby have been identified as the most favourable candidates for cell therapy of autoimmune diseases. We have recently shown that excretory-secretory products (ES L1) released by Trichinella spiralis larvae induce stable human tolDCs in vitro via Toll-like receptor 2 (TLR2) and TLR4. However, engagement of these receptors did not fully explain the tolerogenic profile of DCs. Here, we observed for the first time that dendritic cell-specific ICAM-3 grabbing non-integrin (DC-SIGN) interacts with highly glycosylated ES L1 and contributes to the generation of ES L1-induced tolDCs. Blocking DC-SIGN interfered with the ES L1-induced higher expression of CD40 and CCR7 and the production of IL-10 and TGF-β by DCs. The cooperation of TLR2, TLR4 and DC-SIGN receptors is of importance for the capacity of DCs to prime T cell response toward Th2 and to induce expansion of CD4+CD25+Foxp3+ T cells, as well as for the production of IL-10 and TGF-β by these cells. Overall, these results indicate that induction of tolDCs by ES L1 involves engagement of multiple pattern recognition receptors namely, TLR2, TLR4 and DC-SIGN.
Collapse
|
25
|
Abstract
The hygiene hypothesis posits that the decreased incidence of parasitic infection in developed countries may underlie an increased prevalence of allergic and autoimmune diseases in these countries. As unique inflammation modulator of intracellular parasitism, Trichinella spiralis, or its excretory-secretory (ES) product, shows improved responses to allergies, autoimmune diseases, inflammatory bowel disease, type 1 diabetes, rheumatic arthritis and autoimmune encephalomyelitis by exerting immunomodulatory effects on both innate and adaptive immune cells in animal models. Research has shown that T. spiralis differs from other helminths in manipulation of the host immune response not only by well-known characteristics of its life cycle, but also by its inflammation modulation pathway. How the parasite achieves inflammation modulation has not been fully elucidated yet. This review will generalize the mechanism and focuses on ES immunomodulatory molecules of T. spiralis that may be important for developing new therapeutics for inflammatory disorders.
Collapse
|
26
|
Jackson-Thompson BM, Torrero M, Mitre BK, Long J, Packiam M, Mitre E. Axenic Caenorhabditis elegans antigen protects against development of type-1 diabetes in NOD mice. J Transl Autoimmun 2020; 3:100065. [PMID: 32939449 PMCID: PMC7476867 DOI: 10.1016/j.jtauto.2020.100065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/23/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Studies in humans and animals have demonstrated that infection with helminths (parasitic worms) is protective against a range of hyperinflammatory diseases. A number of factors limit translation into clinical use, including: potential contamination of helminths obtained from infected humans or animals, lack of batch to batch stability, and potential pathological risks derived from live worm infections. To overcome these limitations we tested whether an antigen homogenate of the non-pathogenic nematode Caenorhabditis elegans confers protection against type 1 diabetes mellitus (T1D) using the Non Obese Diabetic (NOD) mouse model. Our study demonstrates that twice weekly intraperitoneal injections of axenically cultured C. elegans antigen (aCeAg) confers substantial protection against type 1 diabetes in NOD mice. Whereas 80% of control mice (PBS-injected) developed clinical disease, only 10% of aCeAg-treated mice became diabetic. Additionally, aCeAg treated mice had significantly greater numbers of insulin-producing pancreatic islets and greater numbers of islets negative for lymphocyte infiltration. Immunological changes observed in aCeAg treated mice included increases in total IgE and total IgG1, consistent with induction of a type 2 immune response similar to that typically seen in parasitic worm infection. Although evidence suggests that helminth infections induce strong immunoregulatory signals, we did not observe significant changes in regulatory T cell numbers or in production of the regulatory cytokines TGFβ and IL-10. The lack of a regulatory response may be due to our time point of observation, or perhaps the mechanism of aCeAg efficacy may differ from that of helminth infection. Discovery that antigens obtained from a non-parasitic environmental nematode replicate the protective phenotype induced by parasitic worm infections may accelerate our ability to develop nematode-derived therapies for allergy and autoimmune diseases. Axenically cultured C. elegans is protective against T1D in NOD mice. aCeAg protection is comparable to helminth immune therapy in NOD mice. aCeAg administration induces total IgE and total IgG similar to helminth infection. aCeAg does not induce TLR4 nor antigen-specific IgE activity. aCeAg efficacy finding may aid in nematode therapy for hyperinflammatory diseases.
Collapse
Affiliation(s)
| | - Marina Torrero
- Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | | | - James Long
- Walter Reed National Military Medical Center, 8901 Wisconsin Ave, Bethesda, MD, 20889, USA
| | - Mathanraj Packiam
- Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Edward Mitre
- Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| |
Collapse
|
27
|
Cleenewerk L, Garssen J, Hogenkamp A. Clinical Use of Schistosoma mansoni Antigens as Novel Immunotherapies for Autoimmune Disorders. Front Immunol 2020; 11:1821. [PMID: 32903582 PMCID: PMC7438586 DOI: 10.3389/fimmu.2020.01821] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
The hygiene hypothesis states that improved hygiene and the resulting disappearance of once endemic diseases is at the origin of the enormous increase in immune related disorders such as autoimmune diseases seen in the industrialized world. Helminths, such as Schistosoma mansoni, are thought to provide protection against the development of autoimmune diseases by regulating the host's immune response. This modulation primarily involves induction of regulatory immune responses, such as generation of tolerogenic dendritic cells and alternatively activated macrophages. This points toward the potential of employing helminths or their products/metabolites as therapeutics for autoimmune diseases that are characterized by an excessive inflammatory state, such as multiple sclerosis (MS), type I diabetes (T1D) and inflammatory bowel disease (IBD). In this review, we examine the known mechanisms of immune modulation by S. mansoni, explore preclinical and clinical studies that investigated the use of an array helminthic products in these diseases, and propose that helminthic therapy opens opportunities in the treatment of chronic inflammatory disorders.
Collapse
Affiliation(s)
- L Cleenewerk
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Beta Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Beta Sciences, Utrecht University, Utrecht, Netherlands.,Division of Immunology, Danone Nutricia Research B.V., Utrecht, Netherlands
| | - Astrid Hogenkamp
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Beta Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
28
|
Shayesteh Z, Hosseini H, Nasiri V, Haddadi Z, Moradi N, Beikzadeh L, Sezavar M, Heidari A, Zibaei M. Evaluating the preventive and curative effects of Toxocara canis larva in Freund's complete adjuvant-induced arthritis. Parasite Immunol 2020; 42:e12760. [PMID: 32472559 DOI: 10.1111/pim.12760] [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: 07/17/2019] [Revised: 05/16/2020] [Accepted: 05/21/2020] [Indexed: 11/29/2022]
Abstract
Helminthic infection and the parallel host immune reactions are the results of a protracted dynamic co-interaction between the host and worms. An assessment of the effect of Toxocara canis infection on arthritis in rats stimulated by Freund's complete adjuvant (FCA) was the main purpose of the investigation. An arthritis model was established by the administration of 0.1 mL FCA in the palmar surface. Cytokine assessment, evaluating oedema and the use of a rheumatoid arthritis (RA) score provided evidence of the protective effects of T canis against adjuvant-induced arthritis (AIA). The cytokines TGF-β, IFN-ɣ, IL-10 and IL-17 were measured to assess the anti-inflammatory effect of T canis infection. Besides, arthritis swelling findings were evaluated in rat paws. The data showed that T canis infection significantly modulated the immune response by alleviating inflammatory cytokines and increasing TGF-β as an anti-inflammatory cytokine. Evaluations of arthritis swelling showed low severity and faster recuperation. These findings suggest that the products derived from T canis eggs might be a potential therapeutic candidate to treat autoimmune diseases like the arthritis.
Collapse
Affiliation(s)
- Zahra Shayesteh
- Department of Immunology, School of Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Hamid Hosseini
- Department of Parasitology and Mycology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Vahid Nasiri
- Protozoology Laboratory, Parasitology Department, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Zeinab Haddadi
- Department of Medical Laboratory Sciences, School of Allied Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Najmeh Moradi
- Department of Immunology, School of Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Leila Beikzadeh
- Department of Medical Laboratory Sciences, School of Allied Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Monireh Sezavar
- Department of Medical Laboratory Sciences, School of Allied Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Aliehsan Heidari
- Department of Parasitology and Mycology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Zibaei
- Department of Parasitology and Mycology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
29
|
Shimokawa C, Kato T, Takeuchi T, Ohshima N, Furuki T, Ohtsu Y, Suzue K, Imai T, Obi S, Olia A, Izumi T, Sakurai M, Arakawa H, Ohno H, Hisaeda H. CD8 + regulatory T cells are critical in prevention of autoimmune-mediated diabetes. Nat Commun 2020; 11:1922. [PMID: 32321922 PMCID: PMC7176710 DOI: 10.1038/s41467-020-15857-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 03/31/2020] [Indexed: 02/08/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which insulin-producing pancreatic β-cells are destroyed. Intestinal helminths can cause asymptomatic chronic and immunosuppressive infections and suppress disease in rodent models of T1D. However, the underlying regulatory mechanisms for this protection are unclear. Here, we report that CD8+ regulatory T (Treg) cells prevent the onset of streptozotocin -induced diabetes by a rodent intestinal nematode. Trehalose derived from nematodes affects the intestinal microbiota and increases the abundance of Ruminococcus spp., resulting in the induction of CD8+ Treg cells. Furthermore, trehalose has therapeutic effects on both streptozotocin-induced diabetes and in the NOD mouse model of T1D. In addition, compared with healthy volunteers, patients with T1D have fewer CD8+ Treg cells, and the abundance of intestinal Ruminococcus positively correlates with the number of CD8+ Treg cells in humans. Helminth infections are associated with a reduction in inflammatory pathology in rodent models of type 1 diabetes. Here, the authors show patient data and that trehalose (produced by H. polygyrus) can alter the microbiome of mice, inducing regulatory CD8+ T cells and reducing susceptibility to autoimmune diabetes.
Collapse
Affiliation(s)
- Chikako Shimokawa
- Department of Parasitology, National Institute of Infectious Disease, Tokyo, 162-8640, Japan. .,Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan. .,Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.
| | - Tamotsu Kato
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Immunobiolgy Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Tadashi Takeuchi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Grauduate School of Medicine, Keio University, Tokyo, 160-8582, Japan
| | - Noriyasu Ohshima
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Takao Furuki
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8502, Japan
| | - Yoshiaki Ohtsu
- Department of Pediatrics, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Kazutomo Suzue
- Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Takashi Imai
- Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Seiji Obi
- Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Alex Olia
- Department of Parasitology, National Institute of Infectious Disease, Tokyo, 162-8640, Japan.,Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Takashi Izumi
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Minoru Sakurai
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8502, Japan
| | - Hirokazu Arakawa
- Department of Pediatrics, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan. .,Immunobiolgy Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan. .,Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Ebina, 243-0435, Japan.
| | - Hajime Hisaeda
- Department of Parasitology, National Institute of Infectious Disease, Tokyo, 162-8640, Japan. .,Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan.
| |
Collapse
|
30
|
Maizels RM. Regulation of immunity and allergy by helminth parasites. Allergy 2020; 75:524-534. [PMID: 31187881 DOI: 10.1111/all.13944] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/26/2022]
Abstract
There is increasing interest in helminth parasite modulation of the immune system, both from the fundamental perspective of the "arms race" between host and parasite, and equally importantly, to understand if parasites offer new pathways to abate and control untoward immune responses in humans. This article reviews the epidemiological and experimental evidence for parasite down-regulation of host immunity and immunopathology, in allergy and other immune disorders, and recent progress towards defining the mechanisms and molecular mediators which parasites exploit in order to modulate their host. Among these are novel products that interfere with epithelial cell alarmins, dendritic cell activation, macrophage function and T-cell responsiveness through the promotion of an immunoregulatory environment. These modulatory effects assist parasites to establish and survive, while dampening immune reactivity to allergens, autoantigens and microbiome determinants.
Collapse
Affiliation(s)
- Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunology and Inflammation University of Glasgow Glasgow UK
| |
Collapse
|
31
|
Itami N, Kondo Y, Tademoto S, Ito D, Fukumoto S, Otsuki H. Alternative Activation of Macrophages in Mice Peritoneal Cavities and Diaphragms by Newborn Larvae of Trichinella spiralis. Yonago Acta Med 2020; 63:34-41. [PMID: 32158331 DOI: 10.33160/yam.2020.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/26/2019] [Indexed: 12/11/2022]
Abstract
Background Trichinellosis is a serious zoonosis with a worldwide distribution. Fecund adult worms in the intestine release newborn larvae (NBL) that enter the general circulation from 4 days post infection (dpi). Alternatively activated macrophages in the peritoneal cavities and the diaphragms in Trichinella spiralis infected mice have been reported. However, a role of newborn larvae is poorly understood. Methods The total numbers of peritoneal macrophages in mice infected with 500 muscle-stage larvae were counted during early infection and then total RNA was extracted. Peritoneal macrophages from uninfected C57BL/6 mice were incubated with IL-4 or LPS as a control, or co-cultured with live NBL, and peritoneal macrophages were obtained from mice injected with live or frozen dead NBL into peritoneal cavity. Total RNA was extracted from these macrophages. Two types of gene expression, classical and alternative activation, were examined in the macrophages and diaphragms of the infected mice using semi-quantitative reverse transcription-PCR. Results The number of peritoneal macrophages in T. spiralis infected mice increased significantly. mRNA peak expression of alternative activation markers, Ym1 and arginase-1 (Arg1), was confirmed in the peritoneal macrophages and in diaphragm of mice around 15 dpi, while mRNA expression of classical activation markers, TNFα, IP-10, and iNOS was not detected. Injection of live NBL into the peritoneal cavities induced mRNA expression of Ym1 and Arg1 in the peritoneal macrophages of mice 9 dpi. However, dead NBL did not induce such gene expression. Alternative activation was not detected in the peritoneal macrophages co-cultured with NBL in vitro. Conclusion Gene expression of alternative activation makers, Ym1 and Arg1, was confirmed in the peritoneal macrophages and diaphragms of mice infected with T. spiralis. However, gene expression of classical activation markers was not detected. Live NBL induced an alternative activation of peritoneal macrophages in vivo, but not in vitro.
Collapse
Affiliation(s)
- Nanase Itami
- Division of Medical Zoology, Department of Microbiology and Immunology, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Yoko Kondo
- Division of Medical Zoology, Department of Microbiology and Immunology, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Sayuri Tademoto
- Technical Department, Tottori University, Yonago 683-8503, Japan
| | - Daisuke Ito
- Division of Medical Zoology, Department of Microbiology and Immunology, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Soji Fukumoto
- Division of Medical Zoology, Department of Microbiology and Immunology, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan.,Tottori Medical Career Support Center, Tottori University Hospital, Yonago 683-8504, Japan
| | - Hitoshi Otsuki
- Division of Medical Zoology, Department of Microbiology and Immunology, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| |
Collapse
|
32
|
Abdoli A, Mirzaian Ardakani H. Potential application of helminth therapy for resolution of neuroinflammation in neuropsychiatric disorders. Metab Brain Dis 2020; 35:95-110. [PMID: 31352539 DOI: 10.1007/s11011-019-00466-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/14/2019] [Indexed: 12/19/2022]
Abstract
Neuropsychiatric disorders (NPDs) are among the major debilitating disorders worldwide with multiple etiological factors. However, in recent years, psychoneuroimmunology uncovered the role of inflammatory condition and autoimmune disorders in the etiopathogenesis of different NPDs. Hence, resolution of inflammation is a new therapeutic target of NPDs. On the other hand, Helminth infections are among the most prevalent infectious diseases in underdeveloped countries, which usually caused chronic infections with minor clinical symptoms. Remarkably, helminths are among the master regulator of inflammatory reactions and epidemiological studies have shown an inverse association between prevalence of autoimmune disorders with these infections. As such, changes of intestinal microbiota are known to be associated with inflammatory conditions in various NPDs. Conversely, helminth colonization alters the intestinal microbiota composition that leads to suppression of intestinal inflammation. In animal models and human studies, helminths or their antigens have shown to be protected against severe autoimmune and allergic disorders, decline the intensity of inflammatory reactions and improved clinical symptoms of the patients. Therefore, "helminthic therapy" have been used for modulation of immune disturbances in different autoimmunity illnesses, such as Multiple Sclerosis (MS) and Inflammatory Bowel Disease (IBD). Here, it is proposed that "helminthic therapy" is able to ameliorate neuroinflammation of NPDs through immunomodulation of inflammatory reactions and alteration of microbiota composition. This review discusses the potential application of "helminthic therapy" for resolution of neuroinflammation in NPDs.
Collapse
Affiliation(s)
- Amir Abdoli
- Department of Parasitology and Mycology, School of Medicine, Jahrom University of Medical Sciences, POBox 74148-46199, Ostad Motahari Ave, Jahrom, Iran.
- Zoonoses Research Center, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran.
- Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran.
| | - Hoda Mirzaian Ardakani
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
33
|
Amelioration of type 1 diabetes by recombinant fructose-1,6-bisphosphate aldolase and cystatin derived from Schistosoma japonicum in a murine model. Parasitol Res 2019; 119:203-214. [PMID: 31845020 DOI: 10.1007/s00436-019-06511-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023]
Abstract
Infection with helminth parasites or the administration of their antigens can prevent or attenuate autoimmune diseases. To date, the specific molecules that prime the amelioration are only limited. In this study, recombinant Schistosoma japonicum cystatin (rSjcystatin) and fructose-1,6-bisphosphate aldolase (rSjFBPA) were administered to female NOD mice via intraperitoneal (i.p.) injection to characterize the immunological response by the recombinant proteins. We have shown that the administration of rSjcystatin or rSjFBPA significantly reduced the diabetes incidence and ameliorated the severity of type 1 diabetes mellitus (T1DM). Disease attenuation was associated with suppressed interferon-gamma (IFN-γ) production in autoreactive T cells and with a switch to the production of Th2 cytokines. Following rSjcystatin or rSjFBPA injection, regulatory T cells (Tregs) were remarkably increased, which was accompanied by increased expression of interleukin-10 (IL-10) and transforming growth factor beta (TGF-β). Our study suggests that helminth-derived proteins may be useful in strategies to limit pathology by promoting the Th2 response and upregulating Tregs during the inflammatory tissue-damage process in T1DM.
Collapse
|
34
|
Osada Y, Morita K, Tahara S, Ishihara T, Wu Z, Nagano I, Maekawa Y, Nakae S, Sudo K, Kanazawa T. Th2 signals are not essential for the anti-arthritic effects of Trichinella spiralis in mice. Parasite Immunol 2019; 42:e12677. [PMID: 31605645 DOI: 10.1111/pim.12677] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/01/2019] [Accepted: 10/09/2019] [Indexed: 12/31/2022]
Abstract
AIMS Many parasitic helminths are known to alter host immune responses and consequently affect the progression of autoimmune and allergic diseases. The parasitic nematode Trichinella sp has been reported to suppress several experimental diseases in rodents, including experimental autoimmune encephalomyelitis, type 1 diabetes, colitis, airway inflammation and autoimmune arthritis. We tried to clarify requirement of Th2 cytokines in the anti-arthritic effects of Trichinella spiralis (Ts) against collagen-induced arthritis (CIA). METHODS AND RESULTS We infected Ts and then induced CIA in STAT6KO DBA/1 mice, comparing the disease progression with that in wild-type (WT) DBA/1 mice, Ts significantly mitigated arthritis in WT mice, in addition to the impairment of anti-type II collagen (IIC) IgG production in a subclass-independent manner. The genetic absence of STAT6 in the mice did not abrogate the anti-arthritic effects of Ts. Alteration of splenic cytokines was not related to the anti-arthritic effects of the parasite. Moreover, lack of IL-10 did not abrogate the anti-arthritic effects of Ts. CONCLUSION Our results suggest that the anti-arthritic effects of Ts do not require host Th2 signals.
Collapse
Affiliation(s)
- Yoshio Osada
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kentaro Morita
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Sayaka Tahara
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tsubasa Ishihara
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Zhiliang Wu
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Isao Nagano
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yoichi Maekawa
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan.,Domain of Integrated Life Systems, Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Gifu, Japan
| | - Susumu Nakae
- Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Katsuko Sudo
- Animal Research Center, Tokyo Medical University, Tokyo, Japan
| | - Tamotsu Kanazawa
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
35
|
Sun X, Li Y, Naqvi MAUH, Naqvi SZ, Chu W, Xu L, Song X, Li X, Yan R. Succinate Coenzyme A Ligase Beta-Like Protein from Trichinella spiralis Suppresses the Immune Functions of Rat PBMCs in Vitro and Inhibits the Secretions of Interleukin-17 in Vivo. Vaccines (Basel) 2019; 7:vaccines7040167. [PMID: 31684056 PMCID: PMC6963543 DOI: 10.3390/vaccines7040167] [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: 10/16/2019] [Accepted: 10/29/2019] [Indexed: 12/16/2022] Open
Abstract
Succinate Coenzyme A ligase beta-like protein (SUCLA-β) is a subunit of Succinyl-coenzyme A synthetase, which is involved in substrate synergism, unusual kinetic reaction in which the presence of SUCLA-β for one partial reaction stimulates another partial reaction. Trichinella spiralis is a parasitic nematode, which may hinder the development of autoimmune diseases. Immunomodulatory effects of SUCLA-β from Trichinella spiralis in the parasite-host interaction are unidentified. In this study the gene encoding T. spiralis SUCLA-β was cloned and expressed. Binding activities of recombinant T. spiralis SUCLA-β (rTs-SUCLA-β) to rat peripheral blood mononuclear cells (PBMCs) were checked by immunofluorescence assay (IFA) and the immuno-regulatory effects of rTs-SUCLA-β on cell migration, cell proliferation, nitric oxide (NO) production and apoptosis were observed by co-incubation of rTs-SUCLA-β with rat PBMCs in vitro, while cytokine secretions in rTs-SUCLA-β treated rats were evaluated in vivo. Furthermore, phagocytosis of monocytes was detected by flow cytometry and effects of rTs-SUCLA-β-induced protective immunity on T. spiralis adult worms and muscle larva were evaluated in rats. The IFA results revealed that rTs-SUCLA-β could bind to rat PBMCs. Treatment of PBMCs with rTs-SUCLA-β significantly decreased the monocyte phagocytosis, cell migration and cell proliferation, while NO production and apoptosis of PBMCs were unaffected. Results of the in vivo study showed that the IL-17 secretion decreased significantly after rTs-SUCLA-β administration in rats, while no significant effects were observed on the secretions of IFN-γ, IL-9, TGF-β and IL-4. Moreover, significant reduction of T. spiralis muscle larvae burden and significant increase in anti-rTs-SUCLA-β immunoglobulin level of IgG, IgG1 and IgG2a was observed in rTs-SUCLA-β-administered rats. The results indicated that rTs-SUCLA-β may be a potential target for controlling T. spiralis infection by suppressing the immune functions of the rat PBMCs and by reducing the parasite burden. Additionally it may also contribute to the treatment of autoimmune diseases and graft rejection by suppressing IL-17 immune response in the host.
Collapse
Affiliation(s)
- Xiaoke Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yin Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Muhammad Ali-Ul-Husnain Naqvi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Sana Zahra Naqvi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Wen Chu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Lixin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Xiaokai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Xiangrui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Ruofeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
36
|
Therapeutic applicability of helminths in autoimmune diseases - literature overview. GASTROENTEROLOGY REVIEW 2019; 14:168-172. [PMID: 31649786 PMCID: PMC6807663 DOI: 10.5114/pg.2019.88164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/14/2019] [Indexed: 11/17/2022]
Abstract
This paper presents an overview of published studies conducted on helminths – parasites of the human gastrointestinal tract. Making use of their ability for immunomodulation may lead to the introduction of effective therapies for autoimmune diseases. This paper presents chronologically attempts to treat autoimmune diseases not only of the gastrointestinal tract, but also of the nervous and endocrine systems, which have been undertaken for decades. The overview of analysed reports demonstrates that as medical knowledge on the cells and mediators participating actively in inflammatory processes accumulates, clinical trials focus on ever more specific areas concerning the pathomechanisms of autoimmune diseases. The outcomes of clinical trials conducted both on animals and humans give reasons to assume that the modification of the human intestinal microflora may be the key to fighting against these diseases.
Collapse
|
37
|
French T, Düsedau HP, Steffen J, Biswas A, Ahmed N, Hartmann S, Schüler T, Schott BH, Dunay IR. Neuronal impairment following chronic Toxoplasma gondii infection is aggravated by intestinal nematode challenge in an IFN-γ-dependent manner. J Neuroinflammation 2019; 16:159. [PMID: 31352901 PMCID: PMC6661741 DOI: 10.1186/s12974-019-1539-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Background It has become increasingly evident that the immune and nervous systems are closely intertwined, relying on one another during regular homeostatic conditions. Prolonged states of imbalance between neural and immune homeostasis, such as chronic neuroinflammation, are associated with a higher risk for neural damage. Toxoplasma gondii is a highly successful neurotropic parasite causing persistent subclinical neuroinflammation, which is associated with psychiatric and neurodegenerative disorders. Little is known, however, by what means neuroinflammation and the associated neural impairment can be modulated by peripheral inflammatory processes. Methods Expression of immune and synapse-associated genes was assessed via quantitative real-time PCR to investigate how T. gondii infection-induced chronic neuroinflammation and associated neuronal alterations can be reshaped by a subsequent acute intestinal nematode co-infection. Immune cell subsets were characterized via flow cytometry in the brain of infected mice. Sulfadiazine and interferon-γ-neutralizing antibody were applied to subdue neuroinflammation. Results Neuroinflammation induced by T. gondii infection of mice was associated with increased microglia activation, recruitment of immune cells into the brain exhibiting Th1 effector functions, and enhanced production of Th1 and pro-inflammatory molecules (IFN-γ, iNOS, IL-12, TNF, IL-6, and IL-1β) following co-infection with Heligmosomoides polygyrus. The accelerated cerebral Th1 immune response resulted in enhanced T. gondii removal but exacerbated the inflammation-related decrease of synapse-associated gene expression. Synaptic proteins EAAT2 and GABAAα1, which are involved in the excitation/inhibition balance in the CNS, were affected in particular. These synaptic alterations were partially recovered by reducing neuroinflammation indirectly via antiparasitic treatment and especially by application of IFN-γ-neutralizing antibody. Impaired iNOS expression following IFN-γ neutralization directly affected EAAT2 and GABAAα1 signaling, thus contributing to the microglial regulation of neurons. Besides, reduced CD36, TREM2, and C1qa gene expression points toward inflammation induced synaptic pruning as a fundamental mechanism. Conclusion Our results suggest that neuroimmune responses following chronic T. gondii infection can be modulated by acute enteric nematode co-infection. While consecutive co-infection promotes parasite elimination in the CNS, it also adversely affects gene expression of synaptic proteins, via an IFN-γ-dependent manner. Electronic supplementary material The online version of this article (10.1186/s12974-019-1539-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Timothy French
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Henning Peter Düsedau
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Johannes Steffen
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Aindrila Biswas
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Norus Ahmed
- Department of Veterinary Medicine, Institute of Immunology, Free University Berlin, Berlin, Germany
| | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Free University Berlin, Berlin, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Björn H Schott
- Leibniz Institute of Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University Medicine Göttingen, Göttingen, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
38
|
Ryan NM, Oghumu S. Role of mast cells in the generation of a T-helper type 2 dominated anti-helminthic immune response. Biosci Rep 2019; 39:BSR20181771. [PMID: 30670631 PMCID: PMC6379226 DOI: 10.1042/bsr20181771] [Citation(s) in RCA: 12] [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: 10/25/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
Mast cells are long-lived, innate immune cells of the myeloid lineage which are found in peripheral tissues located throughout the body, and positioned at the interface between the host and the environment. Mast cells are found in high concentrations during helminth infection. Using Kitw-sh mast cell deficient mice, a recently published study in Bioscience Reports by Gonzalez et al. (Biosci. Rep., 2018) focused on the role of mast cells in the immune response to infection by the helminth Hymenolepis diminuta The authors showed that mast cells play a role in the modulation of Th2 immune response characterized by a unique IL-4, IL-5 and IL-13 cytokine profile, as well as subsequent robust worm expulsion during H. diminuta infection. Unlike WT mice which expelled H. diminuta at day 10, Kitw-sh deficient mice displayed delayed worm expulsion (day 14 post infection). Further, a possible role for mast cells in the basal expression of cytokines IL-25, IL-33 and thymic stromal lymphopoietin was described. Deletion of neutrophils in Kitw-sh deficient mice enhanced H. diminuta expulsion, which was accompanied by splenomegaly. However, interactions between mast cells and other innate and adaptive immune cells during helminth infections are yet to be fully clarified. We conclude that the elucidation of mechanisms underlying mast cell interactions with cells of the innate and adaptive immune system during infection by helminths can potentially uncover novel therapeutic applications against inflammatory, autoimmune and neoplastic diseases.
Collapse
Affiliation(s)
- Nathan M Ryan
- Department of Pathology, College of Medicine, Ohio State University Wexner Medical Center, Columbus, OH, U.S.A
| | - Steve Oghumu
- Department of Pathology, College of Medicine, Ohio State University Wexner Medical Center, Columbus, OH, U.S.A.
| |
Collapse
|
39
|
Tang CL, Zou JN, Zhang RH, Liu ZM, Mao CL. Helminths protect against type 1 diabetes: effects and mechanisms. Parasitol Res 2019; 118:1087-1094. [PMID: 30758662 DOI: 10.1007/s00436-019-06247-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 02/01/2019] [Indexed: 02/07/2023]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which cells of the immune system destroy pancreatic β cells, which secrete insulin. The high prevalence of T1D in developed societies may be explained by environmental changes, including lower exposure to helminths. Indeed, infection by helminths such as Schistosoma, Filaria, and Heligmosomoides polygyrus and their by-products has been reported to ameliorate or prevent the development of T1D in human and animal models. Helminths can trigger distinct immune regulatory pathways, often involving adaptive immune cells that include T helper 2 (Th2) cells and regulatory T cells (Tregs) and innate immune cells that include dendritic cells, macrophages, and invariant natural killer T cells, which may act synergistically to induce Tregs in a Toll-like receptor-dependent manner. Cytokines such as interleukin (IL)-4, IL-10, and transforming growth factor (TGF)-β also play an important role in protection from T1D. Herein, we provide a comprehensive review of the effects and mechanisms underlying protection against T1D by helminths.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Jie-Ning Zou
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Rong-Hui Zhang
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Zhi-Ming Liu
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China.
| | - Cun-Lan Mao
- Department of Obstetrics and Gynecology, People's Hospital of Songzi City, Songzi, 434200, Hubei, China.
| |
Collapse
|
40
|
L4 stage Heligmosomoides polygyrus prevents the maturation of dendritic JAWS II cells. Exp Parasitol 2019; 196:12-21. [DOI: 10.1016/j.exppara.2018.10.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/24/2018] [Accepted: 10/30/2018] [Indexed: 12/29/2022]
|
41
|
Saberi R, Sharif M, Sarvi S, Aghayan SA, Hosseini SA, Anvari D, Nayeri Chegeni T, Hosseininejad Z, Daryani A. Is Toxoplasma gondii playing a positive role in multiple sclerosis risk? A systematic review and meta-analysis. J Neuroimmunol 2018; 322:57-62. [DOI: 10.1016/j.jneuroim.2018.06.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 06/01/2018] [Accepted: 06/13/2018] [Indexed: 12/19/2022]
|
42
|
Cheng Y, Zhu X, Wang X, Zhuang Q, Huyan X, Sun X, Huang J, Zhan B, Zhu X. Trichinella spiralis Infection Mitigates Collagen-Induced Arthritis via Programmed Death 1-Mediated Immunomodulation. Front Immunol 2018; 9:1566. [PMID: 30093899 PMCID: PMC6070611 DOI: 10.3389/fimmu.2018.01566] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Helminth infection induces Th2-biased immune responses and inhibitory/regulatory pathways that minimize excessive inflammation to facilitate the chronic infection of helminth in the host and in the meantime, prevent host hypersensitivity from autoimmune or atopic diseases. However, the detailed molecular mechanisms behind modulation on inflammatory diseases are yet to be clarified. Programmed death 1 (PD-1) is one of the important inhibitory receptors involved in the balance of host immune responses during chronic infection. Here, we used the murine model to examine the role of PD-1 in CD4+ T cells in the effects of Trichinella spiralis infection on collagen-induced arthritis (CIA). Mice infected with T. spiralis demonstrated higher expression of PD-1 in the spleen CD4+ T cells than those without infection. Mice infected with T. spiralis 2 weeks prior to being immunized with type II collagen displayed lower arthritis incidence and significantly attenuated pathology of CIA compared with those of uninfected mice. The therapeutic effect of T. spiralis infection on CIA was reversed by blocking PD-1 with anti-PD-1 antibody, associated with enhanced Th1/Th17 pro-inflammatory responses and reduced Th2 responses. The role of PD-1 in regulating CD4+ T cell differentiation and proliferation during T. spiralis infection was further examined in PD-1 knockout (PD-1-/-) C57BL/6 J mice. Interestingly, T. spiralis-induced alteration of attenuated Th1 and enhanced Th2/regulatory T cell differentiation in wild-type (WT) mice was effectively diminished in PD-1-/- mice characterized by recovered Th1 cytokine levels, reduced levels of Th2 and regulatory cytokines and CD4+CD25+Foxp3+ cells. Moreover, T. spiralis-induced CD4+ T cell proliferation suppression in WT mice was partially restored in PD-1-/- mice. This study introduces the first evidence that PD-1 plays a critical role in helminth infection-attenuated CIA in a mouse model by regulating the CD4+ T cell function, which may provide the new insights into the mechanisms of helminth-induced immunomodulation of host autoimmunity.
Collapse
Affiliation(s)
- Yuli Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xing Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaohuan Wang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qinghui Zhuang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xu Huyan
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ximeng Sun
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingjing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bin Zhan
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Sanya RE, Nkurunungi G, Andia Biraro I, Mpairwe H, Elliott AM. A life without worms. Trans R Soc Trop Med Hyg 2018; 111:3-11. [PMID: 28340138 PMCID: PMC5412073 DOI: 10.1093/trstmh/trx010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 02/27/2017] [Indexed: 12/14/2022] Open
Abstract
Worms have co-evolved with humans over millions of years. To survive, they manipulate host systems by modulating immune responses so that they cause (in the majority of hosts) relatively subtle harm. Anthelminthic treatment has been promoted as a measure for averting worm specific pathology and to mitigate subtle morbidities which may include effects on anaemia, growth, cognitive function and economic activity. With our changing environment marked by rapid population growth, urbanisation, better hygiene practices and anthelminthic treatment, there has been a decline in worm infections and other infectious diseases and a rise in non-communicable diseases such as allergy, diabetes and cardiovascular disease. This review reflects upon our age-old interaction with worms, and the broader ramifications of life without worms for vaccine responses and susceptibility to other infections, and for allergy-related and metabolic disease. We touch upon the controversy around the benefits of mass drug administration for the more-subtle morbidities that have been associated with worm infections and then focus our attention on broader, additional aspects of life without worms, which may be either beneficial or detrimental.
Collapse
Affiliation(s)
- Richard E Sanya
- MRC/UVRI Uganda Research Unit, Uganda Virus Research Institute, P.O. Box 49, Entebbe, Uganda.,College of Health Sciences, Makerere University, Kampala, Uganda
| | - Gyaviira Nkurunungi
- MRC/UVRI Uganda Research Unit, Uganda Virus Research Institute, P.O. Box 49, Entebbe, Uganda.,Department of Clinical Research, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | | | - Harriet Mpairwe
- MRC/UVRI Uganda Research Unit, Uganda Virus Research Institute, P.O. Box 49, Entebbe, Uganda
| | - Alison M Elliott
- MRC/UVRI Uganda Research Unit, Uganda Virus Research Institute, P.O. Box 49, Entebbe, Uganda.,Department of Clinical Research, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
44
|
Immunity to gastrointestinal nematode infections. Mucosal Immunol 2018; 11:304-315. [PMID: 29297502 DOI: 10.1038/mi.2017.113] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 11/20/2017] [Indexed: 02/06/2023]
Abstract
Numerous species of nematodes have evolved to inhabit the gastrointestinal tract of animals and humans, with over a billion of the world's population infected with at least one species. These large multicellular pathogens present a considerable and complex challenge to the host immune system given that individuals are continually exposed to infective stages, as well as the high prevalence in endemic areas. This review summarizes our current understanding of host-parasite interactions, detailing induction of protective immunity, mechanisms of resistance, and resolution of the response. It is clear from studies of well-defined laboratory model systems that these responses are dominated by innate and adaptive type 2 cytokine responses, regulating cellular and soluble effectors that serve to disrupt the niche in which the parasites live by strengthening the physical mucosal barrier and ultimately promoting tissue repair.
Collapse
|
45
|
Taenia crassiceps Antigens Control Experimental Type 1 Diabetes by Inducing Alternatively Activated Macrophages. Mediators Inflamm 2017; 2017:8074329. [PMID: 29249872 PMCID: PMC5698814 DOI: 10.1155/2017/8074329] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/19/2017] [Accepted: 08/29/2017] [Indexed: 01/13/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease caused by the selective destruction of the pancreatic β-cells, causing inability to produce insulin. Proinflammatory cytokines such as IL-1β, IL-6, TNF-α, IFN-γ, IL-12, IL-17, and NO can be released by CD4 and CD8+ lymphocytes as well as by classically activated macrophages (CAMϕs), which are important in the development of T1D. Helminth infections have been shown to prevent T1D, mainly through Th2-biased responses and increased recruitment of regulatory cell populations. Previously, we have shown that Taenia crassiceps infection in mice significantly reduces hyperglycemia, insulitis, and the incidence of T1D. In this study, we determined whether T. crassiceps-derived products such as soluble (TcS) or excreted/secreted (TcES) antigens might have a beneficial influence on the development of experimental T1D. Treatment with different doses before or after induction of T1D was analyzed. Mice that were pretreated with TcS were unable to develop T1D, whereas those receiving TcES early after T1D induction displayed significantly reduced insulitis and hyperglycemia along with increased recruitment of alternatively activated macrophages (AAMϕs) and myeloid-derived suppressor cells (MDSCs). Finally, we examined the modulatory role of AAMϕs on T1D by depleting macrophages with clodronate-loaded liposomes, demonstrating that AAMϕs are key cells in T1D regulation.
Collapse
|
46
|
Wu Z, Wang L, Tang Y, Sun X. Parasite-Derived Proteins for the Treatment of Allergies and Autoimmune Diseases. Front Microbiol 2017; 8:2164. [PMID: 29163443 PMCID: PMC5682104 DOI: 10.3389/fmicb.2017.02164] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 10/20/2017] [Indexed: 12/26/2022] Open
Abstract
The morbidity associated with atopic diseases and immune dysregulation disorders such as asthma, food allergies, multiple sclerosis, atopic dermatitis, type 1 diabetes mellitus, and inflammatory bowel disease has been increasing all around the world over the past few decades. Although the roles of non-biological environmental factors and genetic factors in the etiopathology have been particularly emphasized, they do not fully explain the increase; for example, genetic factors in a population change very gradually. Epidemiological investigation has revealed that the increase also parallels a decrease in infectious diseases, especially parasitic infections. Thus, the reduced prevalence of parasitic infections may be another important reason for immune dysregulation. Parasites have co-evolved with the human immune system for a long time. Some parasite-derived immune-evasion molecules have been verified to reduce the incidence and harmfulness of atopic diseases in humans by modulating the immune response. More importantly, some parasite-derived products have been shown to inhibit the progression of inflammatory diseases and consequently alleviate their symptoms. Thus, parasites, and especially their products, may have potential applications in the treatment of autoimmune diseases. In this review, the potential of parasite-derived products and their analogs for use in the treatment of atopic diseases and immune dysregulation is summarized.
Collapse
Affiliation(s)
- Zhenyu Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Lifu Wang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Yanlai Tang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xi Sun
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| |
Collapse
|
47
|
Xu J, Liu Z, Zhan W, Jiang R, Yang C, Zhan H, Xiong Y. Recombinant TsP53 modulates intestinal epithelial barrier integrity via upregulation of ZO‑1 in LPS‑induced septic mice. Mol Med Rep 2017; 17:1212-1218. [PMID: 29115466 DOI: 10.3892/mmr.2017.7946] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 10/18/2017] [Indexed: 11/05/2022] Open
Abstract
Tight junctions (TJs) are a critical component in maintaining the intestinal mucosal barrier function and gastrointestinal health of animals. Gut barrier dysfunction contributes to the initiation and development of sepsis which induces an uncontrollable systemic inflammatory response and gives rise to life‑threatening clinical conditions. Excretory‑secretary antigens from Trichimella spiralis (T. spiralis) have been reported to protect from sepsis in a mouse model, however the mechanism remains to be elucidated. Mice were treated with recombinant T. spiralis 53‑kDa glycoprotein (rTsP53) at 2 or 6 h following lipopolysaccharide (LPS) injection. Survival rate, serum systemic inflammation, Chiu's score, D‑lactic acid (D‑LA) and diamine oxidase (DAO) as intestinal injured biomarkers, bacterial translocation and growth in peritoneal fluid (PF) and mesenteric lymph nodes (MLN), intestinal tight junction structure and protein zona occludens (ZO)‑1 expression were investigated. In LPS‑induced septic mice, rTsP53 was demonstrated to protract the survival and inhibit serum systemic inflammatory response, and then, allayed morphological alteration, decreased the release of D‑LA and DAO from intestines. Furthermore, LPS‑induced intestinal permeability, bacterial translocation and growth in PF, MLN and vital organs were significantly suppressed by rTsP53 treatment. Notably, rTsP53 treatment markedly improved the intestinal tight junction damaged in sepsis via promoting ZO‑1 expression. These results demonstrated that rTsP53 ameliorated LPS‑induced intestinal injury and is a potential protective agent for treatment of sepsis.
Collapse
Affiliation(s)
- Jia Xu
- Department of Emergency Medicine, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhihao Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wei Zhan
- Department of Emergency Medicine, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510360, P.R. China
| | - Ren Jiang
- Department of Emergency Medicine, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chunhua Yang
- Department of Emergency Medicine, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hong Zhan
- Department of Emergency Medicine, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yan Xiong
- Department of Emergency Medicine, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
48
|
The hygiene hypothesis in autoimmunity: the role of pathogens and commensals. Nat Rev Immunol 2017; 18:105-120. [PMID: 29034905 DOI: 10.1038/nri.2017.111] [Citation(s) in RCA: 319] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The incidence of autoimmune diseases has been steadily rising. Concomitantly, the incidence of most infectious diseases has declined. This observation gave rise to the hygiene hypothesis, which postulates that a reduction in the frequency of infections contributes directly to the increase in the frequency of autoimmune and allergic diseases. This hypothesis is supported by robust epidemiological data, but the underlying mechanisms are unclear. Pathogens are known to be important, as autoimmune disease is prevented in various experimental models by infection with different bacteria, viruses and parasites. Gut commensal bacteria also play an important role: dysbiosis of the gut flora is observed in patients with autoimmune diseases, although the causal relationship with the occurrence of autoimmune diseases has not been established. Both pathogens and commensals act by stimulating immunoregulatory pathways. Here, I discuss the importance of innate immune receptors, in particular Toll-like receptors, in mediating the protective effect of pathogens and commensals on autoimmunity.
Collapse
|
49
|
Junginger J, Raue K, Wolf K, Janecek E, Stein VM, Tipold A, Günzel-Apel AR, Strube C, Hewicker-Trautwein M. Zoonotic intestinal helminths interact with the canine immune system by modulating T cell responses and preventing dendritic cell maturation. Sci Rep 2017; 7:10310. [PMID: 28871165 PMCID: PMC5583179 DOI: 10.1038/s41598-017-10677-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 08/14/2017] [Indexed: 12/30/2022] Open
Abstract
Parasite co-evolution alongside the mammalian immune system gave rise to several modulatory strategies by which they prevent exaggerated pathology and facilitate a longer worm survival. As little is known about the immunoregulatory potential of the zoonotic canine parasites Ancylostoma caninum and Toxocara canis in the natural host, the present study aimed to investigate whether their larval excretory-secretory (ES) products can modulate the canine immune system. We demonstrated TcES to increase the frequency of CD4+ Foxp3high T cells, while both AcES and TcES were associated with elevated Helios expression in Foxp3high lymphocytes. ES products were further capable of inducing IL-10 production by lymphocytes, which was mainly attributed to CD8+ T cells. ES treatment of PBMCs prior to mitogen stimulation inhibited polyclonal proliferation of CD4+ and CD8+ T cells. Moreover, monocyte-derived ES-pulsed dendritic cells reduced upregulation of MHC-II and CD80 in response to lipopolysaccharide. The data showed that regulation of the canine immune system by A. caninum and T. canis larvae comprises the modification of antigen-specific and polyclonal T cell responses and dendritic cell maturation.
Collapse
Affiliation(s)
- Johannes Junginger
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, D-30559, Hannover, Germany
| | - Katharina Raue
- Institute for Parasitology, Center for Infection Medicine, University of Veterinary Medicine, Bünteweg 17, D-30559, Hannover, Germany
| | - Karola Wolf
- Unit of Reproductive Medicine of Clinics, University of Veterinary Medicine, Bünteweg 15, D-30559, Hannover, Germany.,Small Animal Clinic, University of Veterinary Medicine, Bünteweg 9, D-30559, Hannover, Germany
| | - Elisabeth Janecek
- Institute for Parasitology, Center for Infection Medicine, University of Veterinary Medicine, Bünteweg 17, D-30559, Hannover, Germany
| | - Veronika M Stein
- Small Animal Clinic, University of Veterinary Medicine, Bünteweg 9, D-30559, Hannover, Germany.,Vetsuisse Faculty, University of Bern, Länggassstrasse 128, CH-3012, Bern, Switzerland
| | - Andrea Tipold
- Small Animal Clinic, University of Veterinary Medicine, Bünteweg 9, D-30559, Hannover, Germany
| | - Anne-Rose Günzel-Apel
- Unit of Reproductive Medicine of Clinics, University of Veterinary Medicine, Bünteweg 15, D-30559, Hannover, Germany.,Small Animal Clinic, University of Veterinary Medicine, Bünteweg 9, D-30559, Hannover, Germany
| | - Christina Strube
- Institute for Parasitology, Center for Infection Medicine, University of Veterinary Medicine, Bünteweg 17, D-30559, Hannover, Germany
| | - Marion Hewicker-Trautwein
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, D-30559, Hannover, Germany.
| |
Collapse
|
50
|
Wang M, Wu L, Weng R, Zheng W, Wu Z, Lv Z. Therapeutic potential of helminths in autoimmune diseases: helminth-derived immune-regulators and immune balance. Parasitol Res 2017; 116:2065-2074. [PMID: 28664463 DOI: 10.1007/s00436-017-5544-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 06/21/2017] [Indexed: 12/22/2022]
Abstract
Helminths have accompanied human throughout history by releasing immune-evasion molecules that could counteract an aberrant immune response within the host. In the past decades, helminth infections are becoming less prevalent possibly due to the developed sanitation. Meanwhile, the incidence of autoimmune diseases is increasing, which cannot be exclusively explained by the changes of susceptibility genes. While the hygiene hypothesis casts light on the problem. The infections of helminths are believed to interact with and regulate human immunity with the byproduct of suppressing the autoimmune diseases. Thus, helminths are potential to treat or cure the autoimmune diseases. The therapeutic progresses and possible immune suppression mechanisms are illustrated in the review. The helminths that are studied most intensively include Heligmosomoides polygyrus, Hymenolepis diminuta, Schistosoma mansoni, Trichinella spiralis, and Trichuris suis. Special attentions are paid on the booming animal models and clinical trials that are to detect the efficiency of immune-modulating helminth-derived molecules on autoimmune diseases. These trials provide us with a prosperous clinical perspective, but the precise mechanism of the down-regulatory immune response remains to be clarified. More efforts are needed to be dedicated until these parasite-derived immune modulators could be used in clinic to treat or cure the autoimmune diseases under a standard management.
Collapse
Affiliation(s)
- Meng Wang
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Linxiang Wu
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Rennan Weng
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Weihong Zheng
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Zhongdao Wu
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, China
| | - Zhiyue Lv
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China. .,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China. .,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, China.
| |
Collapse
|