1
|
Peck Y, Pickering D, Mobli M, Liddell MJ, Wilson DT, Ruscher R, Ryan S, Buitrago G, McHugh C, Love NC, Pinlac T, Haertlein M, Kron MA, Loukas A, Daly NL. Solution structure of the N-terminal extension domain of a Schistosoma japonicum asparaginyl-tRNA synthetase. J Biomol Struct Dyn 2024; 42:7934-7944. [PMID: 37572327 DOI: 10.1080/07391102.2023.2241918] [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: 01/11/2023] [Accepted: 07/24/2023] [Indexed: 08/14/2023]
Abstract
Several secreted proteins from helminths (parasitic worms) have been shown to have immunomodulatory activities. Asparaginyl-tRNA synthetases are abundantly secreted in the filarial nematode Brugia malayi (BmAsnRS) and the parasitic flatworm Schistosoma japonicum (SjAsnRS), indicating a possible immune function. The suggestion is supported by BmAsnRS alleviating disease symptoms in a T-cell transfer mouse model of colitis. This immunomodulatory function is potentially related to an N-terminal extension domain present in eukaryotic AsnRS proteins but few structure/function studies have been done on this domain. Here we have determined the three-dimensional solution structure of the N-terminal extension domain of SjAsnRS. A protein containing the 114 N-terminal amino acids of SjAsnRS was recombinantly expressed with isotopic labelling to allow structure determination using 3D NMR spectroscopy, and analysis of dynamics using NMR relaxation experiments. Structural comparisons of the N-terminal extension domain of SjAsnRS with filarial and human homologues highlight a high degree of variability in the β-hairpin region of these eukaryotic N-AsnRS proteins, but similarities in the disorder of the C-terminal regions. Limitations in PrDOS-based intrinsically disordered region (IDR) model predictions were also evident in this comparison. Empirical structural data such as that presented in our study for N-SjAsnRS will enhance the prediction of sequence-homology based structure modelling and prediction of IDRs in the future.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yoshimi Peck
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Darren Pickering
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Mehdi Mobli
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
| | - Michael J Liddell
- College of Science and Engineering, James Cook University, Cairns, QLD, Australia
| | - David T Wilson
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Roland Ruscher
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Stephanie Ryan
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Geraldine Buitrago
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Connor McHugh
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | | | - Theresa Pinlac
- Department of Biochemistry, University of the Philippines, Manila, Philippines
| | | | - Michael A Kron
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Norelle L Daly
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
2
|
Brito RMDM, de Melo MF, Fernandes JV, Valverde JG, Matta Guedes PM, de Araújo JMG, Nascimento MSL. Acute Chikungunya Virus Infection Triggers a Diverse Range of T Helper Lymphocyte Profiles. Viruses 2024; 16:1387. [PMID: 39339863 PMCID: PMC11437511 DOI: 10.3390/v16091387] [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: 07/17/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Chikungunya virus (CHIKV) is an arbovirus causing acute febrile illness with severe joint pain, often leading to chronic arthralgia. This study investigated the adaptive immune responses during the early stages of symptomatic acute CHIKV infection, focusing on the transcription factors and cytokines linked to Th1, Th2, Th17, and Treg cells. Thirty-six individuals were enrolled: nine healthy controls and 27 CHIKV-positive patients confirmed by qRT-PCR. Blood samples were analyzed for the mRNA expression of transcription factors (Tbet, GATA3, FoxP3, STAT3, RORγt) and cytokines (IFN-γ, IL-4, IL-17, IL-22, TGF-β, IL-10). The results showed the significant upregulation of Tbet, GATA3, FoxP3, STAT3, and RORγt in CHIKV-positive patients, with RORγt displaying the highest increase. Correspondingly, cytokines IFN-γ, IL-4, IL-17, and IL-22 were upregulated, while TGF-β was downregulated. Principal component analysis (PCA) confirmed the distinct immune profiles between CHIKV-positive and healthy individuals. A correlation analysis indicated that higher Tbet expression correlated with a lower viral load, whereas FoxP3 and TGF-β were associated with higher viral loads. Our study sheds light on the intricate immune responses during acute CHIKV infection, characterized by a mixed Th1, Th2, Th17, and Treg response profile. These results emphasize the complex interplay between different adaptive immune responses and how they may contribute to the pathogenesis of Chikungunya fever.
Collapse
Affiliation(s)
| | - Marília Farias de Melo
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - José Veríssimo Fernandes
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Joanna Gardel Valverde
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Paulo Marcos Matta Guedes
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Josélio Maria Galvão de Araújo
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Manuela Sales Lima Nascimento
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| |
Collapse
|
3
|
Sikder S, Pierce D, Sarkar ER, McHugh C, Quinlan KGR, Giacomin P, Loukas A. Regulation of host metabolic health by parasitic helminths. Trends Parasitol 2024; 40:386-400. [PMID: 38609741 DOI: 10.1016/j.pt.2024.03.006] [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: 02/12/2024] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024]
Abstract
Obesity is a worldwide pandemic and major risk factor for the development of metabolic syndrome (MetS) and type 2 diabetes (T2D). T2D requires lifelong medical support to limit complications and is defined by impaired glucose tolerance, insulin resistance (IR), and chronic low-level systemic inflammation initiating from adipose tissue. The current preventative strategies include a healthy diet, controlled physical activity, and medication targeting hyperglycemia, with underexplored underlying inflammation. Studies suggest a protective role for helminth infection in the prevention of T2D. The mechanisms may involve induction of modified type 2 and regulatory immune responses that suppress inflammation and promote insulin sensitivity. In this review, the roles of helminths in counteracting MetS, and prospects for harnessing these protective mechanisms for the development of novel anti-diabetes drugs are discussed.
Collapse
Affiliation(s)
- Suchandan Sikder
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia.
| | - Doris Pierce
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia
| | - Eti R Sarkar
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, Queensland 4878, Australia
| | - Connor McHugh
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, Queensland 4878, Australia
| | - Kate G R Quinlan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Paul Giacomin
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; Macrobiome Therapeutics Pty Ltd, Cairns, Queensland 4878, Australia
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; Macrobiome Therapeutics Pty Ltd, Cairns, Queensland 4878, Australia
| |
Collapse
|
4
|
Gazzinelli-Guimaraes PH, Dulek B, Swanson P, Lack J, Roederer M, Nutman TB. Single-cell molecular signature of pathogenic T helper subsets in type 2-associated disorders in humans. JCI Insight 2024; 9:e177720. [PMID: 38587077 PMCID: PMC11128205 DOI: 10.1172/jci.insight.177720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/14/2024] [Indexed: 04/09/2024] Open
Abstract
To unravel the heterogeneity and molecular signature of effector memory Th2 cells (Tem2), we analyzed 23 individuals' PBMCs of filaria-infected (Filaria+) and 24 healthy volunteers (Filaria-), with or without coincident house dust mite (HDM) allergic sensitization. Flow cytometry revealed 3 CD4+ Tem subsets - CCR4+CCR6+CRTH2- Tem17, CCR4+CCR6-CRTH2+ Tem2, and CCR6+CCR4+CRTH2+ Tem17.2 - markedly enriched in Filaria+ individuals. These subsets were sorted and analyzed by multiomic single-cell RNA immunoprofiling. SingleR-annotated Th2 cells from Tem2 and Tem17.2 cell subsets had features of pathogenic Th2 effector cells based on their transcriptional signatures, with downregulated CD27 and elevated expression levels of ITGA4, IL17RB, HPGDS, KLRB1, PTGDR2, IL9R, IL4, IL5, and IL13 genes. When the Filaria+ individuals were subdivided based on their allergic status, Tem2 cells in HDM+Filaria+ individuals showed an overall reduction in TCR diversity, suggesting the occurrence of antigen-driven clonal expansion. Moreover, HDM+Filaria+ individuals showed not only an expansion in the frequency of both Tem2 and Tem17.2 cell subsets, but also a change in their molecular program by overexpressing GATA3, IL17RB, CLRF2, and KLRB1, as well as increased antigen-induced IL-4, IL-5, and IL-13 production, suggesting that aeroallergens reshape the transcriptional and functional programming of Th2 cell subsets in human filarial infection toward a pathogenic immunophenotype.
Collapse
Affiliation(s)
| | | | - Phillip Swanson
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
5
|
Toychiev A, Gafner N, Belotserkovets V, Sekler D, Tashpulatova S, Osipova S. Impact of Ascaris lumbricoides infection on the development of chronic pulmonary aspergillosis in patients with COPD. Trop Doct 2024; 54:149-156. [PMID: 38291709 DOI: 10.1177/00494755241226488] [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] [Indexed: 02/01/2024]
Abstract
The aetiopathogenesis of chronic obstructive pulmonary disease (COPD) remains unclear. The aim of our study was to determine the possible influence of Ascaris lumbricoides on the development of chronic pulmonary aspergillosis (CPA) in patients with COPD. The prevalence of A. lumbricoides in patients with COPD with CPA (19.05%) was significantly higher than that in those without (9.20%) and controls (4.9%) (p < 0.05). Trends in levels of Interleukin-1β and of tumour necrosis factor α suggest ascariasis increases susceptibility to Aspergillus sp. in patients with COPD and can be considered an additional risk factor for CPA.
Collapse
Affiliation(s)
- Abdurakhim Toychiev
- Postdoctoral Researcher, Department of Immunology of Parasitic and Fungal Diseases, Republican Specialized Research and Practical Medical Center of Epidemiology, Microbiology, Infectious and Parasitic Diseases, Tashkent, Uzbekistan
| | - Natalya Gafner
- Pulmonologist, Department of Therapy, Republican Specialized Scientific and Practical Medical Center of Tuberculosis and Pulmonology, Tashkent, Uzbekistan
| | - Vera Belotserkovets
- Pulmonologist, Department of Therapy, Republican Specialized Scientific and Practical Medical Center of Tuberculosis and Pulmonology, Tashkent, Uzbekistan
| | - Dildora Sekler
- Senior Researcher, Department of Immunology of Parasitic and Fungal Diseases, Republican Specialized Research and Practical Medical Center of Epidemiology, Microbiology, Infectious and Parasitic Diseases, Tashkent, Uzbekistan
| | - Shakhnoza Tashpulatova
- Associate Professor, Department of Infectious and Child Infectious Diseases, Tashkent Medical Academy, Tashkent, Uzbekistan
| | - Svetlana Osipova
- Principal Investigator, Department of Immunology of Parasitic and Fungal Diseases, Republican Specialized Research and Practical Medical Center of Epidemiology, Microbiology, Infectious and Parasitic Diseases, Tashkent, Uzbekistan
| |
Collapse
|
6
|
Mishra R, Tetarbe S, Bendre PS, Shah I. Difficult to treat liver abscesses in children - Do not forget parasitic infections. Trop Doct 2024; 54:172-175. [PMID: 38311934 DOI: 10.1177/00494755241227470] [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] [Indexed: 02/06/2024]
Abstract
Liver abscess (LA) is a significant health concern worldwide, particularly in tropical regions such as India, and is usually pyogenic or amoebic in origin. In rare cases it can be caused by parasites. We present two children with difficult-to-treat LAs, revealing underlying parasitic infections as the causative agents, implicated by eosinophilia, elevated immunoglobulin-E levels and exposure to domestic animals. In the first case, disseminated echinococcosis was diagnosed through imaging, serology and histopathology. The second case showed a relationship between LAs and Toxocara infection, evidenced by microscopic stool examination of a household cat.
Collapse
Affiliation(s)
- Ruchi Mishra
- Hepatology, B.J. Wadia Hospital for Children and Nawrosjee Wadia Maternity Hospital, Mumbai, India
| | - Shivangi Tetarbe
- Hepatology, B.J. Wadia Hospital for Children and Nawrosjee Wadia Maternity Hospital, Mumbai, India
| | - Pradnya Suhas Bendre
- Department of Paediatric Surgery, B.J. Wadia Hospital for Children, Mumbai, India
| | - Ira Shah
- Paediatric Infectious Diseases and Paediatric GI, Hepatology, B.J. Wadia Hospital for Children and Nawrosjee Wadia Maternity Hospital, Mumbai, India
| |
Collapse
|
7
|
Zheng S, Sun L, Huang L, Xie Y, Ding X. Toxocara canis infection in multiple types of animals: ophthalmological and pathological observations. Parasit Vectors 2024; 17:85. [PMID: 38395932 PMCID: PMC10885396 DOI: 10.1186/s13071-023-06070-y] [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: 09/06/2023] [Accepted: 11/27/2023] [Indexed: 02/25/2024] Open
Abstract
Human ocular toxocariasis (OT), caused by pet roundworm Toxocara canis (Nematoda Ascaridoidea), is a worldwide ocular parasitic infection that poses a severe threat to eyesight, especially in school-aged children. However, the infection process and pathological mechanism of Toxocara are difficult to study in the human body. This study was designed to explore long-term ocular manifestations in different rodents infected with Toxocara canis, uncovering the specific pathological mechanism and migration pathway of larvae after infection. The three types of experimental animals we selected were C57BL/6 mice, Mongolian gerbils and Brown Norway rats. Mice were randomly divided into five groups and infected orally with 1000, 2000, 4000, 8000 and 10,000 T. canis eggs; gerbils were randomly divided into four groups and infected orally with 1000, 2000, 4000 and 10,000 T. canis eggs; rats were randomly divided into three groups and infected orally with 2000, 6000 and 10,000 T. canis eggs. Their ocular changes were closely observed and recorded for at least 2 months. We also enucleated the eyeballs of some animals to perform pathological sectioning and hematoxylin-eosin staining. After 3 dpi (days post-infection), hemorrhagic lesions, mechanical injury of the retina and larval migration could be observed in some infected animals. The ocular infection and mortality rates tended to be stable at 7 dpi. Larval tissue, structure disorder and inflammation could be observed in the pathological sections. In conclusion, the mice infected with 2000 T. canis eggs and gerbils infected with 1000, 2000 and 4000 T. canis eggs showing obvious ocular lesions and lower mortality rates could provide a basis for long-term observation.
Collapse
Affiliation(s)
- Shuxin Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 7 Jinsui Road, Guangzhou, 510060, China
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 7 Jinsui Road, Guangzhou, 510060, China
| | - Limei Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 7 Jinsui Road, Guangzhou, 510060, China
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 7 Jinsui Road, Guangzhou, 510060, China
| | - Li Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 7 Jinsui Road, Guangzhou, 510060, China
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 7 Jinsui Road, Guangzhou, 510060, China
| | - Yue Xie
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, China
| | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 7 Jinsui Road, Guangzhou, 510060, China.
- Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 7 Jinsui Road, Guangzhou, 510060, China.
| |
Collapse
|
8
|
Grezzi L, Martínez YE, Barrios AA, Díaz Á, Casaravilla C. Characterization of the immunosuppressive environment induced by larval Echinococcus granulosus during chronic experimental infection. Infect Immun 2024; 92:e0027623. [PMID: 38174942 PMCID: PMC10863420 DOI: 10.1128/iai.00276-23] [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: 07/17/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
The larval stage of Echinococcus granulosus causes the chronic infection known as cystic echinococcosis, deploying strong inhibitory mechanisms on host immune responses. Using experimental intraperitoneal infection in C57BL/6 mice, we carried out an in-depth analysis of the local changes in macrophage populations associated with chronic infection. In addition, we analyzed T cells and relevant soluble mediators. Infected animals showed an increase in local cell numbers, mostly accounted for by eosinophils, T cells, and macrophages. Within macrophage populations, the largest increases in cell numbers corresponded to resident large peritoneal macrophages (LPM). Monocyte recruitment appeared to be active, as judged by the increased number of monocytes and cells in the process of differentiation towards LPM, including small (SPM) and converting peritoneal macrophages (CPM). In contrast, we found no evidence of macrophage proliferation. Infection induced the expression of M2 markers in SPM, CPM, and LPM. It also enhanced the expression of the co-inhibitor PD-L1 in LPM, SPM, and CPM and induced the co-inhibitor PD-L2 in SPM and CPM. Therefore, local macrophages acquire M2-like phenotypes with probable suppressive capacities. Regarding T cells, infection induced an increase in the percentage of CD4+ cells that are PD-1+, which represent a potential target of suppression by PD-L1+/PD-L2+ macrophages. In possible agreement, CD4+ T cells from infected animals showed blunted proliferative responses to in vitro stimulation with anti-CD3. Further evidence of immune suppression in the parasite vicinity arose from the observation of an expansion in FoxP3+ CD4+ regulatory T cells and increases in the local concentrations of the anti-inflammatory cytokines TGF-β and IL-1Ra.
Collapse
Affiliation(s)
- Leticia Grezzi
- Laboratorio de Inmunología, Instituto de Química Biológica, Facultad de Ciencias/Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
- Área Inmunología, Departamento de Biociencias, Facultad de Química/Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | - Yamila E. Martínez
- Laboratorio de Inmunología, Instituto de Química Biológica, Facultad de Ciencias/Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
- Área Inmunología, Departamento de Biociencias, Facultad de Química/Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | - Anabella A. Barrios
- Área Inmunología, Departamento de Biociencias, Facultad de Química/Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | - Álvaro Díaz
- Área Inmunología, Departamento de Biociencias, Facultad de Química/Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | - Cecilia Casaravilla
- Laboratorio de Inmunología, Instituto de Química Biológica, Facultad de Ciencias/Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
9
|
Taghipour A, Javanmard E, Rahimi HM, Abdoli A, Matin S, Haghbin M, Olfatifar M, Mirjalali H, Zali MR. Prevalence of intestinal parasitic infections in patients with diabetes: a systematic review and meta-analysis. Int Health 2024; 16:23-34. [PMID: 37052134 PMCID: PMC10759288 DOI: 10.1093/inthealth/ihad027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/03/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023] Open
Abstract
Patients with diabetes are at an increased risk of intestinal parasitic infections (IPIs). We evaluated the pooled prevalence and OR of IPIs in patients with diabetes through a systematic review and meta-analysis. A systematic search was performed using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) protocol for studies reporting IPIs in patients with diabetes through 1 August 2022. The collected data were analyzed using comprehensive meta-analysis software version 2. Thirteen case-control studies and nine cross-sectional studies were included in this study. The overall prevalence of IPIs in patients with diabetes was calculated to be 24.4% (95% CI 18.8 to 31%). Considering the case-control design, the prevalence of IPIs in case (25.7%; 95% CI 18.4 to 34.5%) was higher than controls (15.5%; 95% CI 8.4 to 26.9%) and a significant correlation was observed (OR, 1.80; 95% CI 1.08 to 2.97%). Moreover, a significant correlation was seen in the prevalence of Cryptosporidium spp. (OR, 3.30%; 95% CI 1.86 to 5.86%), Blastocystis sp. (OR, 1.57%; 95% CI 1.11 to 2.22%) and hookworm (OR, 6.09%; 95% CI 1.11 to 33.41%) in the cases group. The present results revealed a higher prevalence of IPIs in patients with diabetes than in controls. Therefore, the results of this study suggest a proper health education program to preventing measures for the acquisition of IPIs in patients with diabetes.
Collapse
Affiliation(s)
- Ali Taghipour
- Zoonoses Research Center, Jahrom University of Medical Sciences, Jahrom 74148-46199, Iran
| | - Ehsan Javanmard
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Hanieh Mohammad Rahimi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 4739-19395, Iran
| | - Amir Abdoli
- Zoonoses Research Center, Jahrom University of Medical Sciences, Jahrom 74148-46199, Iran
- Department of Medical Parasitology and Mycology, School of Medicine, Jahrom University of Medical Sciences, Jahrom 74148-46199, Iran
| | - Sara Matin
- Department of Pediatrics, Jahrom University of Medical Sciences, Jahrom 74148-46199, Iran
| | - Marzieh Haghbin
- Clinical Research Development Unit of Peymanieh Hospital, Faculty of Medicine, Jahrom University of Medical Sciences, Jahrom 3713649373, Iran
| | - Meysam Olfatifar
- Gastroenterology and Hepataology Diseases Research Center, Qom University of Medical Sciences, Qom 4739-19395, Iran
| | - Hamed Mirjalali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 4739-19395, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 4739-19395, Iran
| |
Collapse
|
10
|
Lekki-Jóźwiak J, Bąska P. The Roles of Various Immune Cell Populations in Immune Response against Helminths. Int J Mol Sci 2023; 25:420. [PMID: 38203591 PMCID: PMC10778651 DOI: 10.3390/ijms25010420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Helminths are multicellular parasites that are a substantial problem for both human and veterinary medicine. According to estimates, 1.5 billion people suffer from their infection, resulting in decreased life quality and burdens for healthcare systems. On the other hand, these infections may alleviate autoimmune diseases and allergy symptoms. The immune system is programmed to combat infections; nevertheless, its effector mechanisms may result in immunopathologies and exacerbate clinical symptoms. This review summarizes the role of the immune response against worms, with an emphasis on the Th2 response, which is a hallmark of helminth infections. We characterize non-immune cells (enteric tuft cells-ETCs) responsible for detecting parasites, as well as the role of hematopoietic-derived cells (macrophages, basophils, eosinophils, neutrophils, innate lymphoid cells group 2-ILC2s, mast cells, T cells, and B cells) in initiating and sustaining the immune response, as well as the functions they play in granulomas. The aim of this paper is to review the existing knowledge regarding the immune response against helminths, to attempt to decipher the interactions between cells engaged in the response, and to indicate the gaps in the current knowledge.
Collapse
Affiliation(s)
- Janina Lekki-Jóźwiak
- Division of Parasitology and Parasitic Diseases, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland;
| | - Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| |
Collapse
|
11
|
Yang L, Zhou R, Wang C, Xie X, Zhou S, Yin F. Host-parasite interactions: a study on the pathogenicity of different Mesanophrys sp. densities and hemocytes-mediated parasitic resistance of swimming crabs (Portunus trituberculatus). Parasitol Res 2023; 123:13. [PMID: 38060025 DOI: 10.1007/s00436-023-08046-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Mesanophrys sp. is a parasitic ciliate that invades and destroys the hemocytes of the swimming crab (Portunus trituberculatus). In the present study, we employed an in vitro model to elucidate how Mesanophrys sp. destroys crab hemocytes. We also evaluated the relationship between the parasite's density, the destruction rate of the hemocytes, and the rapid proliferation pattern of parasites in host crabs. We found that the survival rate and cell integrity of crab hemocytes decreased with an increase in Mesanophrys sp. density, depicting a negative correlation between hemocyte viability and parasite density. Further analyses revealed that crab hemocytes could resist destruction by a low density (10 ind/mL) of Mesanophrys sp. for a long time (60 h). Mesanophrys sp. and its culture medium (containing the ciliate secretions) destroy the host hemocytes. The natural population growth rate of Mesanophrys sp. decreased with an increase in the parasite density, but the Mesanophrys sp. density did not affect the generation time of the parasites. In summary, Mesanophrys sp. can destroy crab hemocytes, and the degree of destruction is directly proportional to the parasite density. The resistance of crab hemocytes to Mesanophrys sp. decreased gradually with an increase in the parasite density.
Collapse
Affiliation(s)
- Lujia Yang
- School of Marine Sciences, National Demonstration Center for Experimental (Aquaculture) Education, Ningbo University, 169 South Qixing Road, Ningbo, 315832, People's Republic of China
| | - Ruiling Zhou
- School of Marine Sciences, National Demonstration Center for Experimental (Aquaculture) Education, Ningbo University, 169 South Qixing Road, Ningbo, 315832, People's Republic of China
| | - Chunlin Wang
- School of Marine Sciences, National Demonstration Center for Experimental (Aquaculture) Education, Ningbo University, 169 South Qixing Road, Ningbo, 315832, People's Republic of China
| | - Xiao Xie
- School of Marine Sciences, National Demonstration Center for Experimental (Aquaculture) Education, Ningbo University, 169 South Qixing Road, Ningbo, 315832, People's Republic of China
| | - Suming Zhou
- School of Marine Sciences, National Demonstration Center for Experimental (Aquaculture) Education, Ningbo University, 169 South Qixing Road, Ningbo, 315832, People's Republic of China.
| | - Fei Yin
- School of Marine Sciences, National Demonstration Center for Experimental (Aquaculture) Education, Ningbo University, 169 South Qixing Road, Ningbo, 315832, People's Republic of China.
| |
Collapse
|
12
|
Nyangiri OA, Mulindwa J, Namulondo J, Kitibwa A, Nassuuna J, Elliott A, Kimuda MP, Boobo A, Nerima B, Adriko M, Dunton NJ, Madhan GK, Kristiansen M, Casacuberta-Partal M, Noyes H, Matovu E. Variants of IL6, IL10, FCN2, RNASE3, IL12B and IL17B loci are associated with Schistosoma mansoni worm burden in the Albert Nile region of Uganda. PLoS Negl Trop Dis 2023; 17:e0011796. [PMID: 38033168 PMCID: PMC10715658 DOI: 10.1371/journal.pntd.0011796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 12/12/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Individuals genetically susceptible to high schistosomiasis worm burden may contribute disproportionately to transmission and could be prioritized for control. Identifying genes involved may guide development of therapy. METHODOLOGY/PRINCIPAL FINDINGS A cohort of 606 children aged 10-15 years were recruited in the Albert Nile region of Uganda and assessed for Schistosoma mansoni worm burden using the Up-Converting Particle Lateral Flow (UCP-LF) test detecting circulating anodic antigen (CAA), point-of-care Circulating Cathodic Antigen (POC-CCA) and Kato-Katz tests. Whole genome genotyping was conducted on 326 children comprising the top and bottom 25% of worm burden. Linear models were fitted to identify variants associated with worm burden in preselected candidate genes. Expression quantitative trait locus (eQTL) analysis was conducted for candidate genes with UCP-LF worm burden included as a covariate. Single Nucleotide Polymorphism loci associated with UCP-LF CAA included IL6 rs2066992 (OR = 0.43, p = 0.0006) and rs7793163 (OR = 2.0, p = 0.0007); IL21 SNP kgp513476 (OR 1.79, p = 0.0025) and IL17B SNP kgp708159 (OR = 0.35, p = 0.0028). A haplotype in the IL10 locus was associated with lower worm burden (OR = 0.53, p = 0.015) and overlapped SNPs rs1800896, rs1800871 and rs1800872. Significant haplotypes (p<0.05, overlapping significant SNP) associated with worm burden were observed in IL6 and the Th17 pathway IL12B and IL17B genes. There were significant eQTL in the IL6, IL5, IL21, IL25 and IFNG regions. CONCLUSIONS Variants associated with S. mansoni worm burden were in IL6, FCN2, RNASE3, IL10, IL12B and IL17B gene loci. However only eQTL associations remained significant after Bonferroni correction. In summary, immune balance, pathogen recognition and Th17 pathways may play a role in modulating Schistosoma worm burden. Individuals carrying risk variants may be targeted first in allocation of control efforts to reduce the burden of schistosomiasis in the community.
Collapse
Affiliation(s)
- Oscar Asanya Nyangiri
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Julius Mulindwa
- Department of Biochemistry and Sports Sciences, College of Natural Sciences, Makerere University, Kampala, Uganda
| | - Joyce Namulondo
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Anna Kitibwa
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Jacent Nassuuna
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Alison Elliott
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Magambo Phillip Kimuda
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Alex Boobo
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Barbara Nerima
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Moses Adriko
- Vector Borne & NTD Control Division, Ministry of Health, Uganda
| | - Nathan J. Dunton
- UCL Genomics core facility, University College London, London, United Kingdom
| | | | - Mark Kristiansen
- UCL Genomics core facility, University College London, London, United Kingdom
| | | | - Harry Noyes
- Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Enock Matovu
- Department of Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | | |
Collapse
|
13
|
Yan SW, Zhang R, Guo X, Wang BN, Long SR, Liu RD, Wang ZQ, Cui J. Trichinella spiralis dipeptidyl peptidase 1 suppressed macrophage cytotoxicity by promoting M2 polarization via the STAT6/PPARγ pathway. Vet Res 2023; 54:77. [PMID: 37705099 PMCID: PMC10500742 DOI: 10.1186/s13567-023-01209-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/31/2023] [Indexed: 09/15/2023] Open
Abstract
Trichinella spiralis dipeptidyl peptidase 1 (TsDPP1), or cysteine cathepsin C, is a secretory protein that is highly expressed during the infective larvae and adult worm stages in the intestines. The aim of this study was to investigate the mechanism by which recombinant TsDPP1 (rTsDPP1) activates macrophages M2 polarization and decreases macrophage cytotoxicity to kill newborn larvae via ADCC. RAW264.7 macrophages and murine peritoneal macrophages were used in this study. The results of the immunofluorescence test (IFT) and confocal microscopy showed that rTsDPP1 specifically bound to macrophages, and the binding site was localized on the cell membrane. rTsDPP1 activated macrophage M2 polarization, as demonstrated by high expression levels of Arg1 (M2 marker) and M2-related genes (IL-10, TGF-β, CD206 and Arg1) and high numbers of CD206+ macrophages. Furthermore, the expression levels of p-STAT6, STAT6 and PPARγ were obviously increased in rTsDPP1-treated macrophages, which were evidently abrogated by using a STAT6 inhibitor (AS1517499) and PPARγ antagonist (GW9662). The results indicated that rTsDPP1 promoted macrophage M2 polarization through the STAT6/PPARγ pathway. Griess reaction results revealed that rTsDPP1 suppressed LPS-induced NO production in macrophages. qPCR and flow cytometry results showed that rTsDPP1 downregulated the expression of FcγR I (CD64) in macrophages. The ability of ADCC to kill newborn larvae was significantly decreased in rTsDPP1-treated macrophages, but AS1517499 and GW9662 restored its killing capacity. Our results demonstrated that rTsDPP1 induced macrophage M2 polarization, upregulated the expression of anti-inflammatory cytokines, and inhibited macrophage-mediated ADCC via activation of the STAT6/PPARγ pathway, which is beneficial to the parasitism and immune evasion of this nematode.
Collapse
Affiliation(s)
- Shu Wei Yan
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Ru Zhang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Xin Guo
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Bo Ning Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China.
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
14
|
Landry RL, Embers ME. The Probable Infectious Origin of Multiple Sclerosis. NEUROSCI 2023; 4:211-234. [PMID: 39483197 PMCID: PMC11523707 DOI: 10.3390/neurosci4030019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 11/03/2024] Open
Abstract
Multiple sclerosis (MS) is an immune inflammatory disease that causes demyelination of the white matter of the central nervous system. It is generally accepted that the etiology of MS is multifactorial and believed to be a complex interplay between genetic susceptibility, environmental factors, and infectious agents. While the exact cause of MS is still unknown, increasing evidence suggests that disease development is the result of interactions between genetically susceptible individuals and the environment that lead to immune dysregulation and CNS inflammation. Genetic factors are not sufficient on their own to cause MS, and environmental factors such as viral infections, smoking, and vitamin D deficiency also play important roles in disease development. Several pathogens have been implicated in the etiology of MS, including Epstein-Barr virus, human herpesvirus 6, varicella-zoster virus, cytomegalovirus, Helicobacter pylori, Chlamydia pneumoniae, and Borrelia burgdorferi. Although vastly different, viruses and bacteria can manipulate host gene expression, causing immune dysregulation, myelin destruction, and neuroinflammation. This review emphasizes the pathogenic triggers that should be considered in MS progression.
Collapse
Affiliation(s)
- Remi L Landry
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, LA 70433, USA
| | - Monica E Embers
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, LA 70433, USA
| |
Collapse
|
15
|
Sheng ZA, Wu CL, Wang DY, Zhong SH, Yang X, Rao GS, Peng H, Feng SW, Li J, Huang WY, Luo HL. Proteomic analysis of exosome-like vesicles from Fasciola gigantica adult worm provides support for new vaccine targets against fascioliasis. Parasit Vectors 2023; 16:62. [PMID: 36765398 PMCID: PMC9921414 DOI: 10.1186/s13071-023-05659-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/09/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) released by helminths play an important role in parasite-host communication. However, little is known about the characteristics and contents of the EVs of Fasciola gigantica, a parasitic flatworm that causes tropical fascioliasis. A better understanding of EVs released by F. gigantica will help elucidate the mechanism of F. gigantica-host interaction and facilitate the search for new vaccine candidates for the control and treatment of fascioliasis. METHODS Two different populations of EVs (15k EVs and 100k EVs) were purified from adult F. gigantica culture media by ultracentrifugation. The morphology and size of the purified EVs were determined by transmission electron microscopy (TEM) and by the Zetasizer Nano ZSP high performance particle characterization system. With the aim of identifying diagnostic markers or potential vaccine candidates, proteins within the isolated 100k EVs were analyzed using mass spectrometry-based proteomics (LC-MS/MS). Mice were then vaccinated with excretory/secretory products (ESPs; depleted of EVs), 15k EVs, 100k EVs and recombinant F. gigantica heat shock protein 70 (rFg-HSP70) combined with alum adjuvant followed by challenge infection with F. gigantica metacercariae. Fluke recovery and antibody levels were used as measures of vaccine protection. RESULTS TEM analysis and nanoparticle tracking analysis indicated the successful isolation of two subpopulations of EVs (15k EVs and 100k EVs) from adult F. gigantica culture supernatants using differential centrifugation. A total of 755 proteins were identified in the 100k EVs. Exosome biogenesis or vesicle trafficking proteins, ESCRT (endosomal sorting complex required for transport) pathway proteins and exosome markers, heat shock proteins and 14-3-3 proteins were identified in the 100k EVs. These results indicate that the isolated 100k EVs were exosome-like vesicles. The functions of the identified proteins may be associated with immune regulation, immune evasion and virulence. Mice immunized with F. gigantica ESPs, 15k EVs, 100k EVs and rFg-HSP70 exhibited a reduction in fluke burden of 67.90%, 60.38%, 37.73% and 56.6%, respectively, compared with the adjuvant control group. The vaccination of mice with F. gigantica 100k EVs, 15k EVs, ESP and rFg-HSP70 induced significant production of specific immunoglobulins in sera, namely IgG, IgG1 and IgG2a. CONCLUSION The results of this study suggest that proteins within the exosome-like vesicles of F. gigantica have immunomodulatory, immune evasion and virulence functions. This knowledge may lead to new strategies for immunotherapy, vaccination and the diagnosis of fascioliasis.
Collapse
Affiliation(s)
- Zhao-An Sheng
- grid.256609.e0000 0001 2254 5798Guangxi Colleges and Universities Key Laboratory of Prevention and Control for Animal Disease, College of Animal Science and Technology, Guangxi University, Nanning, People’s Republic of China ,grid.449428.70000 0004 1797 7280Department of Pathogenic Biology, Jining Medical University, Shandong, People’s Republic of China
| | - Cui-Lan Wu
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, Guangxi People’s Republic of China ,Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, Guangxi People’s Republic of China
| | - Dong-Ying Wang
- grid.256609.e0000 0001 2254 5798Guangxi Colleges and Universities Key Laboratory of Prevention and Control for Animal Disease, College of Animal Science and Technology, Guangxi University, Nanning, People’s Republic of China
| | - Shu-Hong Zhong
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, Guangxi People’s Republic of China ,Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, Guangxi People’s Republic of China
| | - Xi Yang
- grid.256609.e0000 0001 2254 5798Guangxi Colleges and Universities Key Laboratory of Prevention and Control for Animal Disease, College of Animal Science and Technology, Guangxi University, Nanning, People’s Republic of China ,Yuxi Animal Disease Prevention and Control Center, Yuxi, People’s Republic of China
| | - Guo-Shun Rao
- grid.256609.e0000 0001 2254 5798Guangxi Colleges and Universities Key Laboratory of Prevention and Control for Animal Disease, College of Animal Science and Technology, Guangxi University, Nanning, People’s Republic of China
| | - Hao Peng
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, Guangxi People’s Republic of China ,Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, Guangxi People’s Republic of China
| | - Shi-Wen Feng
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, Guangxi People’s Republic of China ,Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, Guangxi People’s Republic of China
| | - Jun Li
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, Guangxi, People's Republic of China. .,Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, Guangxi, People's Republic of China.
| | - Wei-Yi Huang
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control for Animal Disease, College of Animal Science and Technology, Guangxi University, Nanning, People's Republic of China.
| | - Hong-Lin Luo
- Institute of Oncology, Guangxi Academy of Medical Sciences, Nanning, Guangxi, People's Republic of China. .,Guangxi Colleges and Universities Key Laboratory of Prevention and Control for Animal Disease, College of Animal Science and Technology, Guangxi University, Nanning, People's Republic of China.
| |
Collapse
|
16
|
Chin KL, Fonte L, Lim BH, Sarmiento ME, Acosta A. Immunomodulation resulting of helminth infection could be an opportunity for immunization against tuberculosis and mucosal pathogens. Front Immunol 2023; 14:1091352. [PMID: 37020538 PMCID: PMC10067736 DOI: 10.3389/fimmu.2023.1091352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/08/2023] [Indexed: 04/07/2023] Open
Affiliation(s)
- Kai Ling Chin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
- *Correspondence: Kai Ling Chin, ; Luis Fonte, ; Armando Acosta,
| | - Luis Fonte
- Department of Parasitology, Institute of Tropical Medicine “Pedro Kourí”, Havana, Cuba
- *Correspondence: Kai Ling Chin, ; Luis Fonte, ; Armando Acosta,
| | - Boon Huat Lim
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Maria E. Sarmiento
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Armando Acosta
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- *Correspondence: Kai Ling Chin, ; Luis Fonte, ; Armando Acosta,
| |
Collapse
|
17
|
Hadadianpour A, Daniel J, Zhang J, Spiller BW, Makaraviciute A, DeWitt ÅM, Walden HS, Hamilton RG, Peebles RS, Nutman TB, Smith SA. Human IgE mAbs identify major antigens of parasitic worm infection. J Allergy Clin Immunol 2022; 150:1525-1533. [PMID: 35760390 PMCID: PMC9742163 DOI: 10.1016/j.jaci.2022.05.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/14/2022] [Accepted: 05/17/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Much of our understanding of the targets of IgE comes from studies of allergy, though little is known about the natural immunogenic targets seen after parasitic worm infections. OBJECTIVE We used human monoclonal antibodies (mAbs) for an unbiased and comprehensive characterization of the immunodominant antigens targeted by IgE in conditions like allergy or helminth infection that are associated with elevated levels of IgE. METHODS Using human hybridoma technology to immortalize IgE encoding B-cells from peripheral blood of subjects with filarial infections and elevated IgE, we generated naturally occurring human IgE mAbs. B-cell cultures were screened in an unbiased manner for IgE production without regard to specificity. Isolated IgE mAbs were then tested for binding to Brugia malayi somatic extracts using ImmunoCAP, immunoblot, and ELISA. Immunoprecipitation followed by mass spectrometry proteomics was used to identify helminth antigens that were then expressed in Escherichia coli for IgE binding characterization. RESULTS We isolated 56 discrete IgE mAbs from 7 individuals with filarial infections. From these mAbs, we were able to definitively identify 19 filarial antigens. All IgE mAbs targeted filarial excreted/secretory proteins, including a family of previously uncharacterized proteins. Interestingly, the transthyretin-related antigens acted as the dominant inducer of the filaria-specific IgE antibody response. These filaria-specific IgE mAbs were potent inducers of anaphylaxis when passively administered to human FcεRI-expressing mice. CONCLUSIONS We generated human hybridomas secreting naturally occurring helminth-specific IgE mAbs from filarial-infected subjects. This work provides much-needed insight into the ontogeny of helminth-induced immune response and IgE antibody response.
Collapse
Affiliation(s)
- Azadeh Hadadianpour
- Departments of Medicine and Pathology, Nashville, Tenn; departments of Medicine Microbiology and Immunology, Nashville, Tenn
| | - Jacob Daniel
- Departments of Medicine and Pathology, Nashville, Tenn; departments of Medicine Microbiology and Immunology, Nashville, Tenn
| | - Jian Zhang
- Departments of Medicine and Pathology, Nashville, Tenn; departments of Medicine Microbiology and Immunology, Nashville, Tenn
| | - Benjamin W Spiller
- departments of Medicine Microbiology and Immunology, Nashville, Tenn; Pharmacology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tenn
| | | | - Åsa M DeWitt
- Special Allergen Service, Thermo Fisher Scientific, Uppsala, Sweden
| | - Heather S Walden
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, Fla
| | - Robert G Hamilton
- Dermatology, Allergy and Clinical Immunology Laboratory, Johns Hopkins University School of Medicine, Baltimore, Md
| | - R Stokes Peebles
- Departments of Medicine and Pathology, Nashville, Tenn; departments of Medicine Microbiology and Immunology, Nashville, Tenn
| | - Thomas B Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Scott A Smith
- Departments of Medicine and Pathology, Nashville, Tenn; departments of Medicine Microbiology and Immunology, Nashville, Tenn.
| |
Collapse
|
18
|
Hananeh WM, Radhi A, Mukbel RM, Ismail ZB. Effects of parasites coinfection with other pathogens on animal host: A literature review. Vet World 2022; 15:2414-2424. [DOI: 10.14202/vetworld.2022.2414-2424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
A parasite-host relationship is complicated and largely remained poorly understood, especially when mixed infections involving pathogenic bacteria and viruses are present in the same host. It has been found that most parasites are able to manipulate the host's immune responses to evade or overcome its defense systems. Several mechanisms have been postulated that may explain this phenomenon in different animal species. Recent evidence suggests that coinfections involving many parasitic species alter the host's vulnerability to other microorganisms, hinder diagnostic accuracy, and may negatively impact vaccination by altering the host's immune responsiveness. The objective of this review was to provide a comprehensive summary of the current understanding of how parasites interact with other pathogens in different animal species. A better understanding of this complex relationship will aid in the improvement efforts of disease diagnosis, treatment, and control measures such as novel and effective vaccines and therapeutics for infectious diseases.
Collapse
Affiliation(s)
- Wael M. Hananeh
- Department of Veterinary Pathology and Public Health, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box, 3030, Irbid 22110, Jordan
| | - Asya Radhi
- Department of Veterinary Pathology and Public Health, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box, 3030, Irbid 22110, Jordan
| | - Rami M. Mukbel
- Department of Basic Veterinary Medical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Zuhair Bani Ismail
- Department of Clinical Veterinary Medical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| |
Collapse
|
19
|
Gildner TE, Cepon-Robins TJ, Urlacher SS. Cumulative host energetic costs of soil-transmitted helminth infection. Trends Parasitol 2022; 38:629-641. [DOI: 10.1016/j.pt.2022.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/24/2022]
|
20
|
Tomalka JA, Suthar MS, Deeks SG, Sekaly RP. Fighting the SARS-CoV-2 pandemic requires a global approach to understanding the heterogeneity of vaccine responses. Nat Immunol 2022; 23:360-370. [PMID: 35210622 DOI: 10.1038/s41590-022-01130-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/05/2022] [Indexed: 11/09/2022]
Abstract
Host genetic and environmental factors including age, biological sex, diet, geographical location, microbiome composition and metabolites converge to influence innate and adaptive immune responses to vaccines. Failure to understand and account for these factors when investigating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine efficacy may impair the development of the next generation of vaccines. Most studies aimed at identifying mechanisms of vaccine-mediated immune protection have focused on adaptive immune responses. It is well established, however, that mobilization of the innate immune response is essential to the development of effective cellular and humoral immunity. A comprehensive understanding of the innate immune response and environmental factors that contribute to the development of broad and durable cellular and humoral immune responses to SARS-CoV-2 and other vaccines requires a holistic and unbiased approach. Along with optimization of the immunogen and vectors, the development of adjuvants based on our evolving understanding of how the innate immune system shapes vaccine responses will be essential. Defining the innate immune mechanisms underlying the establishment of long-lived plasma cells and memory T cells could lead to a universal vaccine for coronaviruses, a key biomedical priority.
Collapse
Affiliation(s)
- Jeffrey A Tomalka
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.,Department of Pediatrics, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven G Deeks
- Department of Medicine, University of California at San Francisco School of Medicine, San Francisco, CA, USA
| | - Rafick Pierre Sekaly
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA. .,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
21
|
Hardisty GR, Knipper JA, Fulton A, Hopkins J, Dutia BM, Taylor MD. Concurrent Infection With the Filarial Helminth Litomosoides sigmodontis Attenuates or Worsens Influenza A Virus Pathogenesis in a Stage-Dependent Manner. Front Immunol 2022; 12:819560. [PMID: 35140712 PMCID: PMC8818685 DOI: 10.3389/fimmu.2021.819560] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Filarial helminths infect approximately 120 million people worldwide initiating a type 2 immune response in the host. Influenza A viruses stimulate a virulent type 1 pro-inflammatory immune response that in some individuals can cause uncontrolled immunopathology and fatality. Although coinfection with filariasis and influenza is a common occurrence, the impact of filarial infection on respiratory viral infection is unknown. The aim of this study was to determine the impact of pre-existing filarial infection on concurrent infection with influenza A virus. A murine model of co-infection was established using the filarial helminth Litomosoides sigmodontis and the H1N1 (A/WSN/33) influenza A virus (IAV). Co-infection was performed at 3 different stages of L. sigmodontis infection (larval, juvenile adult, and patency), and the impact of co-infection was determined by IAV induced weight loss and clinical signs, quantification of viral titres, and helminth counts. Significant alterations of IAV pathogenesis, dependent upon stage of infection, was observed on co-infection with L. sigmodontis. Larval stage L. sigmodontis infection alleviated clinical signs of IAV co-infection, whilst more established juvenile adult infection also significantly delayed weight loss. Viral titres remained unaltered at either infection stage. In contrast, patent L. sigmdodontis infection led to a reversal of age-related resistance to IAV infection, significantly increasing weight loss and clinical signs of infection as well as increasing IAV titre. These data demonstrate that the progression of influenza infection can be ameliorated or worsened by pre-existing filarial infection, with the outcome dependent upon the stage of filarial infection.
Collapse
Affiliation(s)
- Gareth R. Hardisty
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanna A. Knipper
- Institute of Immunology and Infection Research, Ashworth Laboratories, University of Edinburgh, Edinburgh, United Kingdom
| | - Alison Fulton
- Institute of Immunology and Infection Research, Ashworth Laboratories, University of Edinburgh, Edinburgh, United Kingdom
| | - John Hopkins
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Matthew D. Taylor
- Institute of Immunology and Infection Research, Ashworth Laboratories, University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Matthew D. Taylor,
| |
Collapse
|
22
|
The Role of the Intestinal Epithelium in the "Weep and Sweep" Response during Gastro-Intestinal Helminth Infections. Animals (Basel) 2022; 12:ani12020175. [PMID: 35049796 PMCID: PMC8772803 DOI: 10.3390/ani12020175] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/25/2021] [Accepted: 01/10/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary The immune system actively combats intruders such as bacteria, viruses, fungi, and protozoan and metazoan parasites using leukocytes. During an infection white blood cells are activated to internalize bacteria or viruses and release a number of molecules to kill pathogens. Unfortunately, those mechanisms are ineffective against larger intruders like helminths, which are too large to be killed by a single immune cell. To eliminate gastro-intestinal helminths an integrated response involving the nervous, endocrine, and immune systems are used to expel the parasites. This is achieved through increased gut hydration and muscle contractions which detach worms from the gut and lead to release outside the body in a “weep and sweep” response. Epithelial cells of the intestine are significant players in this process, being responsible for detecting the presence of helminths in the gut and participating in the regulation of parasite expulsion. This paper describes the role of the gut epithelium in detecting and eliminating helminths from the intestine. Abstract Helminths are metazoan parasites infecting around 1.5 billion people all over the world. During coevolution with hosts, worms have developed numerous ways to trick and evade the host immune response, and because of their size, they cannot be internalized and killed by immune cells in the same way as bacteria or viruses. During infection, a substantial Th2 component to the immune response is evoked which helps restrain Th1-mediated tissue damage. Although an enhanced Th2 response is often not enough to kill the parasite and terminate an infection in itself, when tightly coordinated with the nervous, endocrine, and motor systems it can dislodge parasites from tissues and expel them from the gut. A significant role in this “weep and seep” response is attributed to intestinal epithelial cells (IEC). This review highlights the role of various IEC lineages (enterocytes, tuft cells, Paneth cells, microfold cells, goblet cells, and intestine stem cells) during the course of helminth infections and summarizes their roles in regulating gut architecture and permeability, and muscle contractions and interactions with the immune and nervous system.
Collapse
|
23
|
Ieşanu MI, Cliveti R, Anghel M, Stoicescu MM, Boboc C, Ioan A, Galoş F. Parasite-Induced Th2 Polarization—An Unusual Cause of Paediatric Hepatic Abscess. Medicina (B Aires) 2021; 57:medicina57121322. [PMID: 34946267 PMCID: PMC8705542 DOI: 10.3390/medicina57121322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Liver abscess (LA) is a serious infectious disease, but is relatively rare in the paediatric population, especially in developed countries. Mostly, hepatic abscesses are pyogenic, caused by Staphylococcus aureus, while in extremely rare cases can be caused by parasites, such as Ascaris lumbricoides. Antimicrobial therapy and percutaneous drainage are the treatments of choice, lowering the mortality caused by this infection. We report a case of a 3-year-old girl admitted to the hospital for abdominal pain and a low-grade fever, with abdominal ultrasonography revealing a hepatic lesion. Initial laboratory tests showed moderate anaemia, thrombocytosis, eosinophilia, high inflammatory markers, and normal liver function. A computed tomography scan revealed two liver abscesses located subdiaphragmatically, and a high immunoglobulin E (IgE) value (22,300 U/mL). After excluding other possible etiologies, the patient was tested for parasitic infections. IgE for Ascaris lumbricoides came slightly higher. In addition to empirical antibiotic treatment, the patient received albendazole and made an uneventful recovery, with the full remission of the abscesses and without the need for drainage. In certain cases, parasites such as Ascaris lumbricoides are capable of inducing a T helper 2 (Th2) dominated immune response, predisposing the host to eosinophilia, hyperIgE, and increased susceptibility to bacterial infections. Early diagnosis and treatment in these cases may lead to less invasive therapy options in order to obtain a full recovery. To the best of our knowledge, this is the only reported case in the literature of a paediatric patient with parasite-induced liver abscesses, with extremely high IgE values, minimal symptomatology, that made a fast, full recovery without the need of drainage.
Collapse
Affiliation(s)
- Mara-Ioana Ieşanu
- Marie Curie Emergency Children’s Hospital, 041451 Bucharest, Romania; (R.C.); (M.A.); (M.-M.S.); (C.B.); (A.I.); (F.G.)
- Correspondence:
| | - Ramona Cliveti
- Marie Curie Emergency Children’s Hospital, 041451 Bucharest, Romania; (R.C.); (M.A.); (M.-M.S.); (C.B.); (A.I.); (F.G.)
| | - Mălina Anghel
- Marie Curie Emergency Children’s Hospital, 041451 Bucharest, Romania; (R.C.); (M.A.); (M.-M.S.); (C.B.); (A.I.); (F.G.)
| | - Mihai-Mirel Stoicescu
- Marie Curie Emergency Children’s Hospital, 041451 Bucharest, Romania; (R.C.); (M.A.); (M.-M.S.); (C.B.); (A.I.); (F.G.)
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Cătălin Boboc
- Marie Curie Emergency Children’s Hospital, 041451 Bucharest, Romania; (R.C.); (M.A.); (M.-M.S.); (C.B.); (A.I.); (F.G.)
| | - Andreea Ioan
- Marie Curie Emergency Children’s Hospital, 041451 Bucharest, Romania; (R.C.); (M.A.); (M.-M.S.); (C.B.); (A.I.); (F.G.)
| | - Felicia Galoş
- Marie Curie Emergency Children’s Hospital, 041451 Bucharest, Romania; (R.C.); (M.A.); (M.-M.S.); (C.B.); (A.I.); (F.G.)
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
24
|
Chaney HL, Grose LF, LaBarbara JM, Sirk AW, Blancke AM, Sánchez JM, Passaro C, Lonergan P, Mathew DJ. Galectin-1 Confers Endometrial Gene Expression and Protein Related to Maternal-Conceptus Immune Tolerance in Cattle. Biol Reprod 2021; 106:487-502. [PMID: 34792096 DOI: 10.1093/biolre/ioab215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/25/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
Conceptus secretory factors include galectins, a family of carbohydrate binding proteins that elicit cell adhesion and immune suppression by interacting with intracellular and extracellular glycans. In rodents, galectin-1 (LGALS1) promotes maternal-fetal immune tolerance in the decidua through expansion of tolerogenic CD11c+ dendritic cells, increased anti-inflammatory IL-10, and activation of FOXP3+ regulatory T cells (Treg). This study characterized galectin expression in early ruminant conceptuses and endometrium. We also tested the effect of recombinant bovine LGALS1 (rbLGALS1) and progesterone (P4) on endometrial expression of genes and protein related to maternal-fetal immune tolerance in cattle. Elongating bovine and ovine conceptuses expressed several galectins, particularly, LGALS1, LGALS3 and LGALS8. Within bovine endometrium, expression of LGALS3, LGALS7 and LGALS9 was greater on Day 16 of pregnancy compared to the estrous cycle. Within ovine endometrium, LGALS7 was greater during pregnancy compared to the estrous cycle and endometrium of pregnant sheep tended to have greater LGALS9 and LGALS15. Expression of endometrial LGALS4 was less during pregnancy in sheep. Treating bovine endometrium with rbLGALS1 increased endometrial expression of CD11c, IL-10 and FOXP3, within 24 h. Specifically, within caruncular endometrium, both rbLGALS1 and P4 increased FOXP3, suggesting that both ligands may promote Treg expansion. Using IHC, FOXP3+ cells with a leukocyte phenotype were localized to the bovine uterine stratum compactum near the uterine surface and increased in response to rbLGALS1. We hypothesize that galectins have important functions during establishment of pregnancy in ruminants and bovine conceptus LGALS1 and luteal P4 confer mechanisms of maternal-conceptus immune tolerance in cattle.
Collapse
Affiliation(s)
- Heather L Chaney
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, USA
| | - Lindsay F Grose
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, USA
| | - Jeanna M LaBarbara
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, USA
| | - Adam W Sirk
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, USA
| | - Alyssa M Blancke
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, USA
| | - Jose M Sánchez
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| | - Claudia Passaro
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| | - Patrick Lonergan
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| | - Daniel J Mathew
- Department of Animal Science, University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
25
|
Macrophage migration inhibitory factor in Nodding syndrome. PLoS Negl Trop Dis 2021; 15:e0009821. [PMID: 34662363 PMCID: PMC8553141 DOI: 10.1371/journal.pntd.0009821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/28/2021] [Accepted: 09/18/2021] [Indexed: 11/19/2022] Open
Abstract
Nodding syndrome (NS) is a catastrophic and enigmatic childhood epilepsy, accompanied by multiple neurological impairments and neuroinflammation. Of all the infectious, environmental and psychological factors associated with NS, the major culprit is Onchocerca Volvulus (Ov)-a parasitic worm transmitted to human by blackflies. NS seems to be an 'Autoimmune Epilepsy' in light of the recent findings of deleterious autoimmune antibodies to Glutamate receptors and to Leiomodin-I in NS patients. Moreover, we recently found immunogenetic fingerprints in HLA peptide-binding grooves associate with protection or susceptibility to NS. Macrophage migration inhibitory factor (MIF) is an immune-regulatory cytokine playing a central role in modulating innate and adaptive immunity. MIF is also involved in various pathologies: infectious, autoimmune and neurodegenerative diseases, epilepsy and others. Herein, two functional polymorphisms in the MIF gene, a -794 CATT5-8 microsatellite repeat and a -173 G/C single-nucleotide polymorphism, were assessed in 49 NS patients and 51 healthy controls from South Sudan. We also measured MIF plasma levels in established NS patients and healthy controls. We discovered that the frequency of the high-expression MIF -173C containing genotype was significantly lower in NS patients compared to healthy controls. Interestingly however, MIF plasma levels were significantly elevated in NS patients than in healthy controls. We further demonstrated that the HLA protective and susceptibility associations are dominant over the MIF association with NS. Our findings suggest that MIF might have a dual role in NS. Genetically controlled high-expression MIF genotype is associated with disease protection. However, elevated MIF in the plasma may contribute to the detrimental autoimmunity, neuroinflammation and epilepsy.
Collapse
|
26
|
Kwarteng A, Asiedu E, Koranteng KK, Asiedu SO. Highlighting the Relevance of CD8 + T Cells in Filarial Infections. Front Immunol 2021; 12:714052. [PMID: 34603287 PMCID: PMC8481813 DOI: 10.3389/fimmu.2021.714052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/31/2021] [Indexed: 01/06/2023] Open
Abstract
The T cell immune responses in filarial infections are primarily mediated by CD4+ T cells and type 2-associated cytokines. Emerging evidence indicates that CD8+ T cell responses are important for anti-filarial immunity, however, could be suppressed in co-infections. This review summarizes what we know so far about the activities of CD8+ T cell responses in filarial infections, co-infections, and the associations with the development of filarial pathologies.
Collapse
Affiliation(s)
- Alexander Kwarteng
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Ebenezer Asiedu
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Kelvin Kwaku Koranteng
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Samuel Opoku Asiedu
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| |
Collapse
|
27
|
Vieira-Santos F, Leal-Silva T, de Lima Silva Padrão L, Ruas ACL, Nogueira DS, Kraemer L, Oliveira FMS, Caliari MV, Russo RC, Fujiwara RT, Bueno LL. Concomitant experimental coinfection by Plasmodium berghei NK65-NY and Ascaris suum downregulates the Ascaris-specific immune response and potentiates Ascaris-associated lung pathology. Malar J 2021; 20:296. [PMID: 34210332 PMCID: PMC8248286 DOI: 10.1186/s12936-021-03824-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 06/17/2021] [Indexed: 12/22/2022] Open
Abstract
Background Ascariasis and malaria are highly prevalent parasitic diseases in tropical regions and often have overlapping endemic areas, contributing to high morbidity and mortality rates in areas with poor sanitary conditions. Several studies have previously aimed to correlate the effects of Ascaris-Plasmodium coinfections but have obtained contradictory and inconclusive results. Therefore, the present study aimed to investigate parasitological and immunopathological aspects of the lung during murine experimental concomitant coinfection by Plasmodium berghei and Ascaris suum during larvae ascariasis. Methods C57BL/6J mice were inoculated with 1 × 104P. berghei strain NK65-NY-infected red blood cells (iRBCs) intraperitoneally and/or 2500 embryonated eggs of A. suum by oral gavage. P. berghei parasitaemia, morbidity and the survival rate were assessed. On the seventh day postinfection (dpi), A. suum lung burden analysis; bronchoalveolar lavage (BAL); histopathology; NAG, MPO and EPO activity measurements; haematological analysis; and respiratory mechanics analysis were performed. The concentrations of interleukin (IL)-1β, IL-12/IL-23p40, IL-6, IL-4, IL-33, IL-13, IL-5, IL-10, IL-17A, IFN-γ, TNF and TGF-β were assayed by sandwich ELISA. Results Animals coinfected with P. berghei and A. suum show decreased production of type 1, 2, and 17 and regulatory cytokines; low leukocyte recruitment in the tissue; increased cellularity in the circulation; and low levels of NAG, MPO and EPO activity that lead to an increase in larvae migration, as shown by the decrease in larvae recovered in the lung parenchyma and increase in larvae recovered in the airway. This situation leads to severe airway haemorrhage and, consequently, an impairment respiratory function that leads to high morbidity and early mortality. Conclusions This study demonstrates that the Ascaris-Plasmodium interaction is harmful to the host and suggests that this coinfection may potentiate Ascaris-associated pathology by dampening the Ascaris-specific immune response, resulting in the early death of affected animals. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03824-w.
Collapse
Affiliation(s)
- Flaviane Vieira-Santos
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thaís Leal-Silva
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiza de Lima Silva Padrão
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Cristina Loiola Ruas
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Denise Silva Nogueira
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Kraemer
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Marcus Silva Oliveira
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Vidigal Caliari
- Laboratory of Protozooses, Institute of Biological Sciences, Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Institute of Biological Sciences, Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
28
|
Avni D, Avni O. Extracellular Vesicles: Schistosomal Long-Range Precise Weapon to Manipulate the Immune Response. Front Cell Infect Microbiol 2021; 11:649480. [PMID: 33869080 PMCID: PMC8044974 DOI: 10.3389/fcimb.2021.649480] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Schistosomiasis (Bilharziasis), a neglected tropical disease that affects more than 240 million people around the world, is caused by infection with the helminth parasite Schistosoma. As part of their secretome, schistosomes release extracellular vesicles (EVs) that modulate the host immune response. The EV-harbored miRNAs upregulate the innate immune response of the M1 pathway and downregulate the differentiation toward the adaptive Th2 immunity. A schistosomal egg-derived miRNA increases the percentage of regulatory T cells. This schistosomal-inducible immunoediting process generates ultimately a parasitic friendly environment that is applied carefully as restrained Th2 response is crucial for the host survival and successful excretion of the eggs. Evidence indicates a selective targeting of schistosomal EVs, however, the underlying mechanisms are unclear yet. The effects of the schistosomes on the host immune system is in accordance with the hygiene hypothesis, attributing the dramatic increase in recent decades in allergy and other diseases associated with imbalanced immune response, to the reduced exposure to infectious agents that co-evolved with humans during evolution. Deciphering the bioactive cargo, function, and selective targeting of the parasite-secreted EVs may facilitate the development of novel tools for diagnostics and delivered therapy to schistosomiasis, as well as to immune-associated disorders.
Collapse
Affiliation(s)
- Dror Avni
- Laboratory of Molecular Cell Biology, Sheba Medical Center, Tel Hashomer, Israel.,Laboratory for the Study of Tropical Diseases, Sheba Medical Center, Tel Hashomer, Israel.,Department of Medicine C, Sheba Medical Center, Tel Hashomer, Israel
| | - Orly Avni
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
29
|
Mewamba EM, Nyangiri OA, Noyes HA, Egesa M, Matovu E, Simo G. The Genetics of Human Schistosomiasis Infection Intensity and Liver Disease: A Review. Front Immunol 2021; 12:613468. [PMID: 33659002 PMCID: PMC7917240 DOI: 10.3389/fimmu.2021.613468] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Schistosomiasis remains the fourth most prevalent parasitic disease affecting over 200 million people worldwide. Control efforts have focussed on the disruption of the life cycle targeting the parasite, vector and human host. Parasite burdens are highly skewed, and the majority of eggs are shed into the environment by a minority of the infected population. Most morbidity results from hepatic fibrosis leading to portal hypertension and is not well-correlated with worm burden. Genetics as well as environmental factors may play a role in these skewed distributions and understanding the genetic risk factors for intensity of infection and morbidity may help improve control measures. In this review, we focus on how genetic factors may influence parasite load, hepatic fibrosis and portal hypertension. We found 28 studies on the genetics of human infection and 20 studies on the genetics of pathology in humans. S. mansoni and S. haematobium infection intensity have been showed to be controlled by a major quantitative trait locus SM1, on chromosome 5q31-q33 containing several genes involved in the Th2 immune response, and three other loci of smaller effect on chromosomes 1, 6, and 7. The most common pathology associated with schistosomiasis is hepatic and portal vein fibroses and the SM2 quantitative trait locus on chromosome six has been linked to intensity of fibrosis. Although there has been an emphasis on Th2 cytokines in candidate gene studies, we found that four of the five QTL regions contain Th17 pathway genes that have been included in schistosomiasis studies: IL17B and IL12B in SM1, IL17A and IL17F in 6p21-q2, IL6R in 1p21-q23 and IL22RA2 in SM2. The Th17 pathway is known to be involved in response to schistosome infection and hepatic fibrosis but variants in this pathway have not been tested for any effect on the regulation of these phenotypes. These should be priorities for future studies.
Collapse
Affiliation(s)
- Estelle M. Mewamba
- Molecular Parasitology and Entomology Unit, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Oscar A. Nyangiri
- College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Harry A. Noyes
- Centre for Genomic Research, School of Biological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Moses Egesa
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Enock Matovu
- College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Gustave Simo
- Molecular Parasitology and Entomology Unit, Faculty of Science, University of Dschang, Dschang, Cameroon
| |
Collapse
|
30
|
Nishimoto A, Wohlgemuth N, Rosch J, Schultz-Cherry S, Cortez V, Rowe HM. Transkingdom Interactions Important for the Pathogenesis of Human Viruses. J Infect Dis 2020; 223:S201-S208. [PMID: 33330907 DOI: 10.1093/infdis/jiaa735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The bacterial, fungal, and helminthic species that comprise the microbiome of the mammalian host have profound effects on health and disease. Pathogenic viruses must contend with the microbiome during infection and likely have evolved to exploit or evade the microbiome. Both direct interactions between the virions and the microbiota and immunomodulation and tissue remodeling caused by the microbiome alter viral pathogenesis in either host- or virus-beneficial ways. Recent insights from in vitro and murine models of viral pathogenesis have highlighted synergistic and antagonistic, direct and indirect interactions between the microbiome and pathogenic viruses. This review will focus on the transkingdom interactions between human gastrointestinal and respiratory viruses and the constituent microbiome of those tissues.
Collapse
Affiliation(s)
- Andrew Nishimoto
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Nicholas Wohlgemuth
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jason Rosch
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Valerie Cortez
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Hannah M Rowe
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
31
|
Chang JE, Choi MS. A Molecular Perspective on the Potential Benefits of Metformin for the Treatment of Inflammatory Skin Disorders. Int J Mol Sci 2020; 21:ijms21238960. [PMID: 33255783 PMCID: PMC7728327 DOI: 10.3390/ijms21238960] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Due to its anti-hyperglycemic effect, metformin is the first-line medication for the treatment of type 2 diabetes, particularly in people who are obese. However, metformin is a drug with a very wide range of pharmacological properties and reports of its therapeutic effect on diseases including inflammation and cancer are increasing. Numerous research groups have reported that metformin has beneficial effects on a variety of inflammatory skin disorders including psoriasis, acanthosis nigricans, acne, hidradenitis suppurativa, and allergic contact dermatitis. According to these reports, in addition to the well-known action of metformin, that is, its anti-hyperglycemic effect, NF-kB inhibition and the resulting alteration to the cytokine network may be the potential targets of metformin. Its anti-hyperandrogenism effect has also been confirmed as the major action of metformin in some inflammatory skin diseases. Moreover, novel regulatory mechanisms, including autophagy and antioxidant processes, have been suggested as promising mechanisms of action for metformin in inflammatory skin disorders.
Collapse
Affiliation(s)
- Ji-Eun Chang
- Lab of Pharmaceutics, College of Pharmacy, Dongduk Women’s University, Seoul 02748, Korea;
| | - Min Sik Choi
- Lab of Pharmacology, College of Pharmacy, Dongduk Women’s University, Seoul 02748, Korea
- Correspondence:
| |
Collapse
|
32
|
Mayer JU, Brown SL, MacDonald AS, Milling SW. Defined Intestinal Regions Are Drained by Specific Lymph Nodes That Mount Distinct Th1 and Th2 Responses Against Schistosoma mansoni Eggs. Front Immunol 2020; 11:592325. [PMID: 33193437 PMCID: PMC7644866 DOI: 10.3389/fimmu.2020.592325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/01/2020] [Indexed: 12/04/2022] Open
Abstract
The balance of type 1 and type 2 immune responses plays a crucial role in anti-helminth immunity and can either support chronic infection or drive type 2 mediated expulsion of the parasite. Helminth antigens and secreted molecules directly influence this balance and induce a favorable immunological environment for the parasite’s survival. However, less is known if the site of infection also influences the balance of type 1 and type 2 immunity. Here, we report that tissue-specific immune responses are mounted against helminth antigens, which elicited strong IL-4 responses when injected into the skin, while the same antigen, delivered into the intestinal subserosa, induced increased IFN-γ and reduced Th2 responses. Immune responses in individual mesenteric lymph nodes that drain defined regions of the intestine furthermore displayed a site-specific pattern of type 1 and type 2 immunity after Schistosoma mansoni or Heligmosomoides polygyrus infection. S. mansoni egg-specific Th2 responses were detectable in all mesenteric lymph nodes but Th1 responses were only present in those draining the colon, while H. polygyrus infection elicited mixed Th1 and Th2 responses in the lymph nodes associated with the site of infection. Similar site-specific type 1 and type 2 immune responses were observed in the draining lymph nodes after the controlled delivery of S. mansoni eggs into different segments of the small and large intestine using microsurgical techniques. Different subsets of intestinal dendritic cells were hereby responsible for the uptake and priming of Th1 and Th2 responses against helminth antigens. Migratory CD11b+CD103− and especially CD11b+CD103+ DC2s transported S. mansoni egg antigens to the draining lymph nodes to induce Th1 and Th2 responses, while CD103+ DC1s induced only IFN-γ responses. In contrast, H. polygyrus antigens were predominantly transported by CD11b+CD103− DC2s and CD103+ DC1s and all DC subsets induced similar Th1 but weaker Th2 responses, compared to S. mansoni egg antigens. The development of adaptive anti-helminth immune responses is therefore influenced by the antigen itself, the uptake and priming characteristics of antigen-positive dendritic cell subsets and the site of infection, which shape the level of Th1 and Th2 responses in order to create a favorable immunological environment for the parasite.
Collapse
Affiliation(s)
- Johannes U Mayer
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom.,Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sheila L Brown
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Simon W Milling
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
33
|
|
34
|
Epiphanies of well-known and newly discovered macromolecular carbohydrates – A review. Int J Biol Macromol 2020; 156:51-66. [DOI: 10.1016/j.ijbiomac.2020.04.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/08/2020] [Accepted: 04/03/2020] [Indexed: 12/16/2022]
|
35
|
Effects of TLR agonists on immune responses in Trichinella spiralis infected mice. Parasitol Res 2020; 119:2505-2510. [PMID: 32535733 PMCID: PMC7292931 DOI: 10.1007/s00436-020-06747-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023]
Abstract
Human trichinellosis is acquired by eating raw or undercooked meats carrying muscle larvae of Trichinella spp. Toll-like receptors (TLRs) are essential components of the innate immune system. However, little is known about the potential application of TLR agonists for immunotherapy against Trichinella spiralis (T. spiralis) infection. Here, we evaluated the effects of four TLR agonists (i.e., TLR3, TLR4, TLR8, and TLR9 agonists) on T. spiralis infection in mice. The reduction rate of worm burden showed that TLR3 agonist poly(I:C) significantly reduced T. spiralis infection rather than TLR4, TLR8, and TLR9 agonists (p < 0.05). Moreover, TLR3 showed a continuous high-level of expression during 6–35 days post infection (dpi). The levels of interferon-gamma (IFN-γ), interleukin (IL)-2, and IL-6 increased significantly in mice serum compared with control group after treatment with TLR3 agonist at 0, 3, 6, 9, 12, 15, 18, 21, 28, and 35 dpi (p < 0.05). A significant decreasing trend was also detected in levels of IL-10 and IL-4 after treatment with TLR3 agonist compared with control group at 0, 3, 6, 9, 12, 15, 18, 21, 28, and 35 dpi (p < 0.05). Overall, this study suggested that TLR3-targeted therapies might be effective on worm burden reduction by regulation of the cytokine levels in the mice infected with T. spiralis.
Collapse
|
36
|
Ruiz-Manzano RA, Palacios-Arreola MI, Hernández-Cervantes R, Del Río-Araiza VH, Nava-Castro KE, Ostoa-Saloma P, Muñoz-Cruz S, Morales-Montor J. Potential Novel Risk Factor for Breast Cancer: Toxocara canis Infection Increases Tumor Size Due to Modulation of the Tumor Immune Microenvironment. Front Oncol 2020; 10:736. [PMID: 32547942 PMCID: PMC7272683 DOI: 10.3389/fonc.2020.00736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Worldwide, breast cancer is the most important type of cancer in women with regard to incidence and prevalence. Several risk factors interact to increase the probability of breast cancer development. Biological environmental contaminants such as infectious agents play a significant role in tumor development, and helminths have been recognized as cancer enhancers or inducers due to their ability to regulate the host immune response. Toxocara canis is a zoonotic and cosmopolite nematode with immuno-regulatory abilities. T. canis infection has been related to T helper type-2 cell (Th2 or type 2) and regulatory responses. Type 2 and regulatory immune responses may favor the development of comorbidities that are usually controlled or eliminated through a type 1 response such as cancer. The aim of this study was to determine whether T. canis infection alters mammary tumor growth through modulation of the immune response. Infected mice developed larger tumors. Tumor immune cell milieu analysis revealed that infection reduced the proportions of CD8+ lymphocytes and increased the proportions of F4/80+ macrophages and CD19+ B cells. These changes were accompanied by a type 2 local response represented by increased amounts of IL-4 and VEGF and a regulatory microenvironment associated with higher IL-10 levels. Thus, this study demonstrates that T. canis infection enhances tumor development and suggests that this is through modulation of the tumor immune microenvironment.
Collapse
Affiliation(s)
- Rocío Alejandra Ruiz-Manzano
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Margarita Isabel Palacios-Arreola
- Departamento de Genotoxicología y Mutagénesis Ambiental, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Rosalía Hernández-Cervantes
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Víctor Hugo Del Río-Araiza
- Laboratorio de Inmunología y Biología Molecular de Parásitos, Facultad de Medicina Veterinaria y Zootecnia, Departamento de Parasitología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Karen Elizabeth Nava-Castro
- Departamento de Genotoxicología y Mutagénesis Ambiental, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Pedro Ostoa-Saloma
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Samira Muñoz-Cruz
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
37
|
Myeloid-derived suppressor cells exert immunosuppressive function on the T helper 2 in mice infected with Echinococcus granulosus. Exp Parasitol 2020; 215:107917. [PMID: 32446699 DOI: 10.1016/j.exppara.2020.107917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 12/24/2019] [Accepted: 05/14/2020] [Indexed: 01/15/2023]
Abstract
Cystic echinococcosis (CE) is a worldwide hazardous zoonotic parasitosis caused by Echinococcus granulosus. CE development involves complex immunological mechanisms, including participation of multiple immune cells and effector molecules. Myeloid-derived suppressor cells (MDSCs) are known to be involved in chronic and acute inflammatory conditions. In this study, we aimed to characterize the immune function of MDSCs in CE to improve the understanding, prevention and treatment of CE. Our results indicated that MDSCs overexpressing Ly6C and Ly6G inhibit the formation and activity of T helper 2 cells in a NO-dependent manner during E. granulosus infection.
Collapse
|
38
|
Fuenmayor-Boscán A, Hernández-Rincón I, Arismendi-Morillo G, Mengual E, Rivero Z, Romero G, Lizarzábal M, Álvarez-Mon M. Changes in the severity of gastric mucosal inflammation associated with Helicobacter pylori in humans coinfected with intestinal helminths. Indian J Gastroenterol 2020; 39:186-195. [PMID: 32436176 DOI: 10.1007/s12664-020-01023-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 02/04/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Though a few studies in animal models suggest that intestinal helminths (IH) favorably affect evolution of gastritis associated with Helicobacter pylori (H. pylori) the studies supporting this concept in humans are only a few and are based on serological data. METHODS To evaluate the possible influence of IH on the human gastric mucosa, three groups of Venezuelan adults with gastropathy (endoscopically diagnosed) were studied: H. pylori-/IH- (n = 17), H. pylori+/IH- (n = 18), and H. pylori+/IH+ (n = 11). Histological analysis (hematoxylin-eosin) and immunohistochemical staining (peroxidase) for cytokines interleukin-1beta (IL-1β), tumor necrosis factor alpha (TNF-α), gamma interferon (IFN-γ), and interleukin 4 (IL-4) were undertaken in gastric antral biopsies. RESULTS Expression of the four cytokines was detected in all individuals in varying degrees, but proinflammatory cytokines were expressed in a higher degree in the H. pylori+/IH- group, mainly IL-1β (Th1-dominant immune response), associated with a higher degree of both histological inflammation and gastric cancer risk index (GCRI), as compared to the H. pylori-/IH- group. In contrast, an increased expression of IL-4 and a reduced expression of proinflammatory cytokines (Th2-dominant response), plus the tendency to a lower degree of mononuclear infiltration, mucosal atrophy in gastric corpus, and GCRI, were evidenced in the coinfected group. CONCLUSIONS The findings of the present study is perhaps the first histological evidence of a possible modulatory effect of IH on the gastric mucosal inflammatory response due to H. pylori infection in humans.
Collapse
Affiliation(s)
- Alisbeth Fuenmayor-Boscán
- Departamento de Microbiología, Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela. .,Laboratorio de Bacteriología, Edificio Ciencia y Salud, planta baja, Avenida 18 con calle 65, Apartado Postal 15165, Maracaibo, Venezuela.
| | - Ileana Hernández-Rincón
- Instituto de Investigaciones Biológicas "Doctores Orlando Castejón y Haydée V. Castejón", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Gabriel Arismendi-Morillo
- Instituto de Investigaciones Biológicas "Doctores Orlando Castejón y Haydée V. Castejón", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Edgardo Mengual
- Instituto de Investigaciones Biológicas "Doctores Orlando Castejón y Haydée V. Castejón", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Zulbey Rivero
- Departamento de Microbiología, Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Gisela Romero
- Postgrado de Gastroenterología, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Maribel Lizarzábal
- Postgrado de Gastroenterología, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Melchor Álvarez-Mon
- Laboratorio de Enfermedades del Sistema Inmune y Oncológicas, Departamento de Medicina, Universidad de Alcalá, Madrid, Spain
| |
Collapse
|
39
|
Kotraiah V, Phares TW, Browne CD, Pannucci J, Mansour M, Noe AR, Tucker KD, Christen JM, Reed C, MacKay A, Weir GM, Rajagopalan R, Stanford MM, Chung CS, Ayala A, Huang J, Tsuji M, Gutierrez GM. Novel Peptide-Based PD1 Immunomodulators Demonstrate Efficacy in Infectious Disease Vaccines and Therapeutics. Front Immunol 2020; 11:264. [PMID: 32210956 PMCID: PMC7068811 DOI: 10.3389/fimmu.2020.00264] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/31/2020] [Indexed: 12/31/2022] Open
Abstract
Many pathogens use the same immune evasion mechanisms as cancer cells. Patients with chronic infections have elevated levels of checkpoint receptors (e.g., programed cell death 1, PD1) on T cells. Monoclonal antibody (mAb)-based inhibitors to checkpoint receptors have also been shown to enhance T-cell responses in models of chronic infection. Therefore, inhibitors have the potential to act as a vaccine “adjuvant” by facilitating the expansion of vaccine antigen-specific T-cell repertoires. Here, we report the discovery and characterization of a peptide-based class of PD1 checkpoint inhibitors, which have a potent adaptive immunity adjuvant capability for vaccines against infectious diseases. Briefly, after identifying peptides that bind to the recombinant human PD1, we screened for in vitro efficacy in reporter assays and human peripheral blood mononuclear cells (PBMC) readouts. We first found the baseline in vivo performance of the peptides in a standard mouse oncology model that demonstrated equivalent efficacy compared to mAbs against the PD1 checkpoint. Subsequently, two strategies were used to demonstrate the utility of our peptides in infectious disease indications: (1) as a therapeutic in a bacteria-induced lethal sepsis model in which our peptides were found to increase survival with enhanced bacterial clearance and increased macrophage function; and (2) as an adjuvant in combination with a prophylactic malaria vaccine in which our peptides increased T-cell immunogenicity and the protective efficacy of the vaccine. Therefore, our peptides are promising as both a therapeutic agent and a vaccine adjuvant for infectious disease with a potentially safer and more cost-effective target product profile compared to mAbs. These findings are essential for deploying a new immunomodulatory regimen in infectious disease primary and clinical care settings.
Collapse
Affiliation(s)
- Vinayaka Kotraiah
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | - Timothy W Phares
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | | | - James Pannucci
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | - Marc Mansour
- MM Scientific Consultants, Inc., Halifax, NS, Canada
| | - Amy R Noe
- Leidos Life Sciences, Leidos Inc., Frederick, MD, United States
| | | | | | - Charles Reed
- Inovio Pharmaceuticals, Plymouth Meeting, PA, United States
| | | | | | | | | | | | - Alfred Ayala
- Lifespan-Rhode Island Hospital, Providence, RI, United States
| | - Jing Huang
- The Aaron Diamond AIDS Research Center, New York, NY, United States
| | - Moriya Tsuji
- The Aaron Diamond AIDS Research Center, New York, NY, United States
| | - Gabriel M Gutierrez
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| |
Collapse
|
40
|
Mazur-Melewska K, Mania A, Sluzewski W, Figlerowicz M. Clinical pathology of larval toxocariasis. ADVANCES IN PARASITOLOGY 2020; 109:153-163. [PMID: 32381195 DOI: 10.1016/bs.apar.2020.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Human toxocariasis is a worldwide helminth zoonosis caused by roundworms of the genus Toxocara. Toxocariasis is the most common helminth infection in many countries. Disease caused by Toxocara can be classified into five clinical forms: generalised, neurological, ocular, covert, and asymptomatic. The clinical pathology of toxocariasis largely depends on the form of infection, the intensity of the infection, the larvae localisation, and the age of the host. Because histological and molecular examination of toxocariasis is limited by difficulties in obtaining material to analyse, clinical diagnosis is often based on nonspecific tests, such as the detection of eosinophilia and hyperimmunoglobulinemia E (Hyper-IgE). Specific analysis uses an initial ELISA test to detect anti-Toxocara IgG and requires confirmation for all positive results via Western blot. This strategy does not distinguish between actual and recent infections, making the monitoring of disease a challenge for clinicians. Additional research will be required to distinguish active disease from the presence of recent infection.
Collapse
Affiliation(s)
- Katarzyna Mazur-Melewska
- Department of Infectious Diseases and Child Neurology, Karol Marcinkowski University of Medical Sciences, Poznań, Poland.
| | - Anna Mania
- Department of Infectious Diseases and Child Neurology, Karol Marcinkowski University of Medical Sciences, Poznań, Poland
| | - Wojciech Sluzewski
- Department of Infectious Diseases and Child Neurology, Karol Marcinkowski University of Medical Sciences, Poznań, Poland
| | - Magdalena Figlerowicz
- Department of Infectious Diseases and Child Neurology, Karol Marcinkowski University of Medical Sciences, Poznań, Poland
| |
Collapse
|
41
|
Olia A, Shimokawa C, Imai T, Suzue K, Hisaeda H. Suppression of systemic lupus erythematosus in NZBWF1 mice infected with Hymenolepis microstoma. Parasitol Int 2020; 76:102057. [PMID: 31954872 DOI: 10.1016/j.parint.2020.102057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/15/2022]
Abstract
Intestinal helminths induce immune suppressive responses thought to regulate inflammatory diseases including allergies and autoimmune diseases. This study was designed to evaluate whether helminthic infections suppress the natural development of systemic lupus erythematosus (SLE) in NZBWF1 mice. Infection of NZBWF1 SLE-prone mice with two nematodes failed to establish long-lasting settlement. However, the Hymenolepis microstoma (Hm) rodent tapeworm successfully established long-term parasitization of NZBWF1 mice and was used to evaluate the suppressive effects of helminth infection. Ten-month-old NZBWF1 mice developed symptoms including autoantibody generation, proteinuria, glomerular histopathology, and splenomegaly, but mice infected with Hm at 2 months of age did not show any clinical signs. Furthermore, infection with Hm reduced lymphocyte activation and increased regulatory T cells in the spleen and mesenteric lymph nodes. These results indicate that infection with Hm protects NZBWF1 mice from naturally developing SLE and suggest that pathological immunity is attenuated, presumably because of the induction of regulatory T cells.
Collapse
Affiliation(s)
- Alex Olia
- Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Chikako Shimokawa
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takashi Imai
- Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Kazutomo Suzue
- Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Hajime Hisaeda
- Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan.
| |
Collapse
|
42
|
Meningher T, Barsheshet Y, Ofir‐Birin Y, Gold D, Brant B, Dekel E, Sidi Y, Schwartz E, Regev‐Rudzki N, Avni O, Avni D. Schistosomal extracellular vesicle-enclosed miRNAs modulate host T helper cell differentiation. EMBO Rep 2020; 21:e47882. [PMID: 31825165 PMCID: PMC6944914 DOI: 10.15252/embr.201947882] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 11/08/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022] Open
Abstract
During the chronic stage of Schistosoma infection, the female lays fertile eggs, triggering a strong anti-parasitic type 2 helper T-cell (Th2) immune response. It is unclear how this Th2 response gradually declines even though the worms live for years and continue to produce eggs. Here, we show that Schistosoma mansoni downregulates Th2 differentiation in an antigen-presenting cell-independent manner, by modulating the Th2-specific transcriptional program. Adult schistosomes secrete miRNA-harboring extracellular vesicles that are internalized by Th cells in vitro. Schistosomal miRNAs are found also in T helper cells isolated from Peyer's patches and mesenteric lymph nodes of infected mice. In T helper cells, the schistosomal miR-10 targets MAP3K7 and consequently downmodulates NF-κB activity, a critical transcription factor for Th2 differentiation and function. Our results explain, at least partially, how schistosomes tune down the Th2 response, and provide further insight into the reciprocal geographic distribution between high prevalence of parasitic infections and immune disorders such as allergy. Furthermore, this worm-host crosstalk mechanism can be harnessed to develop diagnostic and therapeutic approaches for human schistosomiasis and Th2-associated diseases.
Collapse
Affiliation(s)
- Tal Meningher
- Laboratory of Molecular Cell BiologyCenter for Cancer Research and Department of Medicine CSheba Medical CenterTel HashomerIsrael
- Molecular Laboratory for the Study of Tropical DiseasesSheba Medical CenterTel HashomerIsrael
| | | | - Yifat Ofir‐Birin
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Daniel Gold
- Department of Clinical Microbiology and ImmunologyFaculty of MedicineSackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Boris Brant
- Azrieli Faculty of MedicineBar Ilan UniversitySafedIsrael
| | - Elya Dekel
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Yechezkel Sidi
- Laboratory of Molecular Cell BiologyCenter for Cancer Research and Department of Medicine CSheba Medical CenterTel HashomerIsrael
- Faculty of MedicineSackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Eli Schwartz
- Molecular Laboratory for the Study of Tropical DiseasesSheba Medical CenterTel HashomerIsrael
- Faculty of MedicineSackler School of MedicineTel Aviv UniversityTel AvivIsrael
- The Center for Geographic MedicineSheba Medical CenterTel HashomerIsrael
| | - Neta Regev‐Rudzki
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Orly Avni
- Azrieli Faculty of MedicineBar Ilan UniversitySafedIsrael
| | - Dror Avni
- Laboratory of Molecular Cell BiologyCenter for Cancer Research and Department of Medicine CSheba Medical CenterTel HashomerIsrael
- Molecular Laboratory for the Study of Tropical DiseasesSheba Medical CenterTel HashomerIsrael
| |
Collapse
|
43
|
Ruiz-Manzano RA, Hernández-Cervantes R, Del Río-Araiza VH, Palacios-Arreola MI, Nava-Castro KE, Morales-Montor J. Immune response to chronic Toxocara canis infection in a mice model. Parasite Immunol 2019; 41:e12672. [PMID: 31557337 DOI: 10.1111/pim.12672] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
AIMS The zoonotic nematode Toxocara canis causes larva migrans syndrome that induces an immune response characterized by the production of antibodies and eosinophilia. A Th2 polarization has been associated with the infection, but there are still details of the cellular and humoral immune response that need to be described. Thus, the aim of this study was to describe the systemic host immune response to T canis chronic infection in a mouse model. METHODS AND RESULTS BALB/c mice were inoculated once with 500 T canis embryonated eggs, per os. After 49 days, the amounts of larval found in brain and muscle tissues were statistically two and four times higher, respectively, than the amounts found in lung, liver, kidney or heart tissues. Splenic proportions of F4/80+ cells, as well as B, cytotoxic T and CD4+ Foxp3+ lymphocytes, were statistically higher (P ≤ .05, P ≤ .01, P ≤ .001 and P ≤ .001, respectively) as compared with control mice. In lymph nodes, some of these proportions changed, with the exception of F4/80+ cells. IgG1 levels in infected mice sera were increased. IL-4, IL-10 and VEGF levels were statistically higher in spleen (P ≤ .05, all) and sera (P ≤ .01, P ≤ .05 and P ≤ .05, respectively) in the infected mice. Also, in infected animals, IL-5 serum levels were increased (P ≤ .01). CONCLUSION These results suggest that T canis chronic infection in BALB/c mice results in a type 2 response with an incipient regulatory response.
Collapse
Affiliation(s)
- Rocío Alejandra Ruiz-Manzano
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Rosalía Hernández-Cervantes
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Víctor Hugo Del Río-Araiza
- Departamento de Parasitología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Margarita Isabel Palacios-Arreola
- Departamento de Genotoxicología, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Karen Elizabeth Nava-Castro
- Departamento de Genotoxicología, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
44
|
Coronado S, Zakzuk J, Regino R, Ahumada V, Benedetti I, Angelina A, Palomares O, Caraballo L. Ascaris lumbricoides Cystatin Prevents Development of Allergic Airway Inflammation in a Mouse Model. Front Immunol 2019; 10:2280. [PMID: 31611876 PMCID: PMC6777510 DOI: 10.3389/fimmu.2019.02280] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022] Open
Abstract
Severe helminth infections are negatively associated to allergic diseases like asthma; therefore, the immunomodulatory properties of parasite-derived components have been analyzed, raising the possibility of their use as anti-inflammatory molecules. We evaluated the immunomodulatory properties of Ascaris lumbricoides recombinant cysteine protease inhibitor (rAl-CPI) in a mouse model of allergic airway inflammation induced by the house dust mite (HDM) Blomia tropicalis and its effects on human monocyte-derived dendritic cells (HmoDCs). The B. tropicalis sensitized/challenged mice developed extensive cellular airway inflammatory response, which was significantly reduced upon treatment with rAl-CPI prior to B. tropicalis sensitization, affecting particularly the perivascular/peribronchial infiltrate cells, eosinophils/neutrophils, and goblet cells. A significant decrease of Th2 cytokines, total, and specific IgE antibodies was observed in rAl-CPI treated mice. The antibody response was biased to IgG, mainly IgG2a. Administration of rAl-CPI-alone and rAl-CPI before mite sensitization were associated with a significant increase of regulatory T cells (Tregs) in spleen and elevated IL-10 levels in BAL and splenocytes culture supernatants, which was partially affected by anti-IL10 receptor use. In vitro, rAl-CPI showed a modulatory effect on HmoDCs, lowering the expression of HLA-DR, CD83, and CD86, while inducing IL-10 and IL-6 production. This suggests an inhibition of HmoDC maturation and a possible link with the inhibition of the allergic response observed in the murine model.
Collapse
Affiliation(s)
- Sandra Coronado
- Institute for Immunological Research, Universidad de Cartagena, Cartagena, Colombia
| | - Josefina Zakzuk
- Institute for Immunological Research, Universidad de Cartagena, Cartagena, Colombia
| | - Ronald Regino
- Institute for Immunological Research, Universidad de Cartagena, Cartagena, Colombia
| | - Velky Ahumada
- Institute for Immunological Research, Universidad de Cartagena, Cartagena, Colombia
| | - Ines Benedetti
- Faculty of Medicine, Universidad de Cartagena, Cartagena, Colombia
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, Chemistry School, Complutense University of Madrid, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, Chemistry School, Complutense University of Madrid, Madrid, Spain
| | - Luis Caraballo
- Institute for Immunological Research, Universidad de Cartagena, Cartagena, Colombia
| |
Collapse
|
45
|
Gazzinelli-Guimaraes PH, de Queiroz Prado R, Ricciardi A, Bonne-Année S, Sciurba J, Karmele EP, Fujiwara RT, Nutman TB. Allergen presensitization drives an eosinophil-dependent arrest in lung-specific helminth development. J Clin Invest 2019; 129:3686-3701. [PMID: 31380805 DOI: 10.1172/jci127963] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
This study investigates the relationship between helminth infection and allergic sensitization by assessing the influence of preexisting allergy on the outcome of helminth infections, rather than the more traditional approach in which the helminth infection precedes the onset of allergy. Here we used a murine model of house dust mite-induced (HDM-induced) allergic inflammation followed by Ascaris infection to demonstrate that allergic sensitization drives an eosinophil-rich pulmonary type 2 immune response (Th2 cells, M2 macrophages, type 2 innate lymphoid cells, IL-33, IL-4, IL-13, and mucus) that directly hinders larval development and reduces markedly the parasite burden in the lungs. This effect is dependent on the presence of eosinophils, as eosinophil-deficient mice were unable to limit parasite development or numbers. In vivo administration of neutralizing antibodies against CD4 prior to HDM sensitization significantly reduced eosinophils in the lungs, resulting in the reversal of the HDM-induced Ascaris larval killing. Our data suggest that HDM allergic sensitization drives a response that mimics a primary Ascaris infection, such that CD4+ Th2-mediated eosinophil-dependent helminth larval killing in the lung tissue occurs. This study provides insight into the mechanisms underlying tissue-specific responses that drive a protective response against the early stages of the helminths prior to their establishing long-lasting infections in the host.
Collapse
Affiliation(s)
- Pedro H Gazzinelli-Guimaraes
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Rafael de Queiroz Prado
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Alessandra Ricciardi
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Sandra Bonne-Année
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Joshua Sciurba
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Erik P Karmele
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA.,Institute for Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Ricardo T Fujiwara
- Department of Parasitology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thomas B Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
46
|
Piecyk A, Ritter M, Kalbe M. The right response at the right time: Exploring helminth immune modulation in sticklebacks by experimental coinfection. Mol Ecol 2019; 28:2668-2680. [PMID: 30993799 PMCID: PMC6852435 DOI: 10.1111/mec.15106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 02/06/2023]
Abstract
Parasites are one of the strongest selective agents in nature. They select for hosts that evolve counter‐adaptive strategies to cope with infection. Helminth parasites are special because they can modulate their hosts’ immune responses. This phenomenon is important in epidemiological contexts, where coinfections may be affected. How different types of hosts and helminths interact with each other is insufficiently investigated. We used the three‐spined stickleback (Gasterosteus aculeatus) – Schistocephalus solidus model to study mechanisms and temporal components of helminth immune modulation. Sticklebacks from two contrasting populations with either high resistance (HR) or low resistance (LR) against S. solidus, were individually exposed to S. solidus strains with characteristically high growth (HG) or low growth (LG) in G. aculeatus. We determined the susceptibility to another parasite, the eye fluke Diplostomum pseudospathaceum, and the expression of 23 key immune genes at three time points after S. solidus infection. D. pseudospathaceum infection rates and the gene expression responses depended on host and S. solidus type and changed over time. Whereas the effect of S. solidus type was not significant after three weeks, T regulatory responses and complement components were upregulated at later time points if hosts were infected with HG S. solidus. HR hosts showed a well orchestrated immune response, which was absent in LR hosts. Our results emphasize the role of regulatory T cells and the timing of specific immune responses during helminth infections. This study elucidates the importance to consider different coevolutionary trajectories and ecologies when studying host‐parasite interactions.
Collapse
Affiliation(s)
- Agnes Piecyk
- Evolutionary Ecology, Max Planck Institute for Evolutionary Biology, Plön, Germany.,Marine Evolutionary Ecology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Kiel, Germany
| | - Marc Ritter
- Evolutionary Ecology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Martin Kalbe
- Evolutionary Ecology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| |
Collapse
|
47
|
Kumar S, Jeong Y, Ashraf MU, Bae YS. Dendritic Cell-Mediated Th2 Immunity and Immune Disorders. Int J Mol Sci 2019; 20:ijms20092159. [PMID: 31052382 PMCID: PMC6539046 DOI: 10.3390/ijms20092159] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/29/2019] [Accepted: 04/29/2019] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DCs) are the professional antigen-presenting cells that recognize and present antigens to naïve T cells to induce antigen-specific adaptive immunity. Among the T-cell subsets, T helper type 2 (Th2) cells produce the humoral immune responses required for protection against helminthic disease by activating B cells. DCs induce a Th2 immune response at a certain immune environment. Basophil, eosinophil, mast cells, and type 2 innate lymphoid cells also induce Th2 immunity. However, in the case of DCs, controversy remains regarding which subsets of DCs induce Th2 immunity, which genes in DCs are directly or indirectly involved in inducing Th2 immunity, and the detailed mechanisms underlying induction, regulation, or maintenance of the DC-mediated Th2 immunity against allergic environments and parasite infection. A recent study has shown that a genetic defect in DCs causes an enhanced Th2 immunity leading to severe atopic dermatitis. We summarize the Th2 immune-inducing DC subsets, the genetic and environmental factors involved in DC-mediated Th2 immunity, and current therapeutic approaches for Th2-mediated immune disorders. This review is to provide an improved understanding of DC-mediated Th2 immunity and Th1/Th2 immune balancing, leading to control over their adverse consequences.
Collapse
Affiliation(s)
- Sunil Kumar
- Science Research Center (SRC) for Immune Research on Non-Lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do 16419, Korea.
| | - Yideul Jeong
- Science Research Center (SRC) for Immune Research on Non-Lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do 16419, Korea.
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do 16419, Korea.
| | - Muhammad Umer Ashraf
- Science Research Center (SRC) for Immune Research on Non-Lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do 16419, Korea.
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do 16419, Korea.
| | - Yong-Soo Bae
- Science Research Center (SRC) for Immune Research on Non-Lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do 16419, Korea.
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do 16419, Korea.
| |
Collapse
|
48
|
Yasuda K, Nakanishi K. Host responses to intestinal nematodes. Int Immunol 2019; 30:93-102. [PMID: 29346656 DOI: 10.1093/intimm/dxy002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/10/2018] [Indexed: 12/19/2022] Open
Abstract
Helminth infection remains common in developing countries, where residents who suffer from the consequences of such infections can develop serious physical and mental disorders and often persist in the face of serious economic problems. Intestinal nematode infection induces the development of Th2-type immune responses including the B-cell IgE response; additionally, this infection induces an increase in the numbers and activation of various types of effector cells, such as mast cells, eosinophils and basophils, as well as the induction of goblet cell hyperplasia, anti-microbial peptide production and smooth-muscle contraction, all of which contribute to expel nematodes. Innate immunity is important in efforts to eliminate helminth infection; cytokines, including IL-25, IL-33 and thymic stromal lymphopoietin, which are products of epithelial cells and mast cells, induce Th2 cells and group 2 innate lymphoid cells to proliferate and produce Th2 cytokines. Nematodes also facilitate chronic infection by suppression of immune reactions through an increased number of Treg cells. Immunosuppression by parasite infection may ultimately be beneficial for the host animals; indeed, a negative correlation has been found between parasite infection and the prevalence of inflammatory disease in humans.
Collapse
Affiliation(s)
- Koubun Yasuda
- Department of Immunology, Hyogo College of Medicine, Hyogo, Japan
| | - Kenji Nakanishi
- Department of Immunology, Hyogo College of Medicine, Hyogo, Japan
| |
Collapse
|
49
|
Zamora V, Rodero M, Andreu-Ballester JC, Mendez S, Cuéllar C. Induction of tolerogenic properties by Anisakis larval antigens on murine dendritic cells. Parasite Immunol 2019; 41:e12616. [PMID: 30719721 DOI: 10.1111/pim.12616] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 01/24/2019] [Accepted: 01/29/2019] [Indexed: 12/17/2022]
Abstract
AIMS The objective of this work is to investigate whether Anisakis simplex larval antigens present immunomodulatory properties by the induction of tolerogenic dendritic cells (DCs) from two strains of mice (BALB/c and C57BL/6J). METHODS AND RESULTS We used mouse bone marrow-derived DCs. We determined their antigen-presenting ability by expression of membrane markers (MHC I and MHC II, CD80, CD86) and intracellular expression levels of IL-10 and IL-12 cytokines. We also analysed whether stimulation with A simplex larval antigens is enhanced by the co-administration of the TLR4 and TLR9 agonists [LPS E coli 026B6 and CpG (ODN1826), respectively]. Two differential types of responses were found in the two mouse strains studied: the BALB/c strain showed an acute and inflammatory response, whereas the C57BL/6J mice developed a more discrete and resistant response. This suggests the coexistence of two opposing responses generated by A simplex larval antigens and confirms that the host genetic basis plays a role in the development of a Th2 or Treg response. CONCLUSION The study of the mechanisms by which Anisakis manipulates the immune response through anti-inflammatory molecules is of interest not only for the direct application on the development of anthelmintic strategies, but also for the development of new anti-inflammatory products.
Collapse
Affiliation(s)
- Vega Zamora
- Facultad de Farmacia, Departamento de Microbiología y Parasitología, Universidad Complutense, Madrid, Spain
| | - Marta Rodero
- Facultad de Farmacia, Departamento de Microbiología y Parasitología, Universidad Complutense, Madrid, Spain
| | | | - Susana Mendez
- Microbiology Review Branch, DHHS/NIH/NIAID/DEA/SRP, Rockville, Maryland
| | - Carmen Cuéllar
- Facultad de Farmacia, Departamento de Microbiología y Parasitología, Universidad Complutense, Madrid, Spain
| |
Collapse
|
50
|
Coelho CH, Gazzinelli-Guimaraes PH, Howard J, Barnafo E, Alani NAH, Muratova O, McCormack A, Kelnhofer E, Urban JF, Narum DL, Anderson C, Langhorne J, Nutman TB, Duffy PE. Chronic helminth infection does not impair immune response to malaria transmission blocking vaccine Pfs230D1-EPA/Alhydrogel® in mice. Vaccine 2019; 37:1038-1045. [PMID: 30685251 PMCID: PMC6382667 DOI: 10.1016/j.vaccine.2019.01.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 12/20/2022]
Abstract
Pfs230 is a candidate malaria transmission blocking vaccine against P. falciparum. Pfs230 vaccine is being tested in areas where malaria and helminth infections are co-endemic. Chronic helminth infection induces a marked increase in systemic Th2 and regulatory cytokine levels in mice. Chronic H. polygyrus bakeri infection does not alter Pfs230 vaccine specific-antibody levels. Functional activity of Pfs230 vaccine was not impaired by chronic helminth infection in mice.
Introduction Malaria transmission blocking vaccines (TBV) are innovative approaches that aim to induce immunity in humans against Plasmodium during mosquito stage, neutralizing the capacity of the infected vectors to transmit malaria. Pfs230D1-EPA/Alhydrogel®, a promising protein-protein conjugate malaria TBV, is currently being tested in human clinical trials in areas where P. falciparum malaria is coendemic with helminth parasites. Helminths are complex metazoans that share the master capacity to downregulate the host immune response towards themselves and also to bystander antigens, including vaccines. However, it is not known whether the activity of a protein-based malaria TBV may be affected by a chronic helminth infection. Methods Using an experimental murine model for a chronic helminth infection (Heligmosomoides polygyrus bakeri - Hpb), we evaluated whether prior infection alters the activity of Pfs230D1-EPA/Alhydrogel® TBV in mice. Results After establishment of a chronic infection, characterized by a marked increase of parasite antigen-specific IgG1, IgA and IgE antibody responses, concomitant with an increase of systemic IL-10, IL-5 and IL-6 levels, the Hpb-infected mice were immunized with Pfs230D1-EPA/Alhydrogel® and the vaccine-specific immune response was compared with that in non-infected immunized mice. TBV immunizations induced an elevated vaccine specific-antibody response, however Pfs230D1 specific-IgG levels were similar between infected and uninfected mice at days 15, 25 and 35 post-vaccination. Absolute numbers of Pfs230D1-activated B cells generated in response to the vaccine were also similar among the vaccinated groups. Finally, vaccine activity assessed by reduction of oocyst number in P. falciparum infected mosquitoes was similar between Hpb-infected and immunized mice with non-infected immunized mice. Conclusion Pfs230D1-EPA/Alhydrogel® efficacy is not impaired by a chronic helminth infection in mice.
Collapse
Affiliation(s)
- Camila H Coelho
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Jennifer Howard
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Emma Barnafo
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Nada A H Alani
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Olga Muratova
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Ashley McCormack
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Emily Kelnhofer
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Joseph F Urban
- US Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomic and Immunology Laboratory, Beltsville, MD, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Charles Anderson
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Thomas B Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|