1
|
Anwar S, Alhumaydhi FA, Rahmani AH, Kumar V, Alrumaihi F. A Review on Risk Factors, Traditional Diagnostic Techniques, and Biomarkers for Pneumonia Prognostication and Management in Diabetic Patients. Diseases 2024; 12:310. [PMID: 39727640 DOI: 10.3390/diseases12120310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/20/2024] [Accepted: 11/30/2024] [Indexed: 12/28/2024] Open
Abstract
People of all ages can contract pneumonia, and it can cause mild to severe disease and even death. In addition to being a major cause of death for elderly people and those with prior medical conditions such as diabetes, it isthe world's biggest infectious cause of death for children. Diabetes mellitus is a metabolic condition with a high glucose level and is a leading cause of lower limb amputation, heart attacks, strokes, blindness, and renal failure. Hyperglycemia is known to impair neutrophil activity, damage antioxidant status, and weaken the humoral immune system. Therefore, diabetic patients are more susceptible to pneumonia than people without diabetes and linked fatalities. The absence of quick, precise, simple, and affordable ways to identify the etiologic agents of community-acquired pneumonia has made diagnostic studies' usefulness contentious. Improvements in biological markers and molecular testing techniques have significantly increased the ability to diagnose pneumonia and other related respiratory infections. Identifying the risk factors for developing severe pneumonia and early testing in diabetic patients might lead to a significant decrease in the mortality of diabetic patients with pneumonia. In this regard, various risk factors, traditional testing techniques, and pathomechanisms are discussed in this review. Further, biomarkers and next-generation sequencing are briefly summarized. Finding biomarkers with the ability to distinguish between bacterial and viral pneumonia could be crucial because identifying the precise pathogen would stop the unnecessary use of antibiotics and effectively save the patient's life.
Collapse
Affiliation(s)
- Shehwaz Anwar
- Department of Medical Laboratory Technology, College of Nursing and Paramedical Sciences, Bareilly 243302, Uttar Pradesh, India
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Vikalp Kumar
- Department of Medical Laboratory Technology, College of Nursing and Paramedical Sciences, Bareilly 243302, Uttar Pradesh, India
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
2
|
Amorim CCO, Nogueira DS, Gazzinelli-Guimarães AC, Leal-Silva T, Barbosa FS, Oliveira FMS, Kraemer LR, de Almeida RM, Souza JLN, Dias Magalhães LM, Russo RC, Caliari MV, Gaze S, Bueno LL, Fujiwara RT. Dose-response effects of multiple Ascaris suum exposures and their impact on lung protection during larval ascariasis. PLoS Negl Trop Dis 2024; 18:e0012678. [PMID: 39621794 PMCID: PMC11637409 DOI: 10.1371/journal.pntd.0012678] [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: 07/01/2024] [Revised: 12/12/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Human ascariasis is the most prevalent geohelminthiasis worldwide, affecting approximately 446 million individuals. In regions with endemic prevalence, the majority of infected adults are frequently exposed to the parasite and tend to have a low parasite load. Further studies are necessary to provide more evidence on the dynamics of infection and to elucidate the possible mechanisms involved in regulating protection, especially during the acute phase, also known as larval ascariasis. The aim of this study is to compare the impact of lung function between single and multiple infections in a murine model. METHODS We infected BALB/c mice considering the frequency of exposures: single-exposure-SI; twice-exposures-RE 2x and thrice-exposures-RE 3x, and considering the doses of infection: 25 eggs-RE 25; 250 eggs-RE 250 and 2,500 eggs-RE 2500, followed by infection challenge with 2,500 eggs. From this, we evaluated: parasite burden in lungs, cellular and humoral response, histopathological and physiological alterations in lungs. RESULTS The main results showed a reduction of parasite burden in the reinfected groups compared to the single-infected group, with protection increasing with higher exposure and dose. Furthermore, the RE 250 group exhibited a decrease of parasite burden close to RE 2500, but with less tissue damage, displaying the most favorable prognosis among the reinfected groups. CONCLUSION Our research indicates a dose-dependent relationship between antibody production and the intensity of the immune response required to regulate the parasite burden.
Collapse
Affiliation(s)
- Chiara Cássia Oliveira Amorim
- Laboratory of Immunobiology and Parasite Control, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | | | - Ana Clara Gazzinelli-Guimarães
- Laboratory of Immunobiology and Parasite Control, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Thais Leal-Silva
- Laboratory of Immunobiology and Parasite Control, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Fernando Sérgio Barbosa
- Laboratory of Immunobiology and Parasite Control, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Fabrício Marcus Silva Oliveira
- Laboratory of Immunobiology and Parasite Control, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Lucas Rocha Kraemer
- Laboratory of Immunobiology and Parasite Control, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Raquel Martins de Almeida
- Laboratory of Immunobiology and Parasite Control, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Jorge Lucas Nascimento Souza
- Laboratory of Immunobiology and Parasite Control, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Luisa Mourão Dias Magalhães
- Laboratory of Interactions in ImmunoParasitology, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Marcelo Vidigal Caliari
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Soraya Gaze
- Cellular and Molecular Immunology Group, René Rachou Institute, Oswaldo Cruz Foundation–FIOCRUZ, Minas Gerais, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunobiology and Parasite Control, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunobiology and Parasite Control, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| |
Collapse
|
3
|
Antar SA, ElMahdy MK, Darwish AG. Examining the contribution of Notch signaling to lung disease development. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6337-6349. [PMID: 38652281 DOI: 10.1007/s00210-024-03105-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
Notch pathway is a widely observed signaling system that holds pivotal functions in regulating various developmental cellular functions and operations. The Notch signaling mechanism is crucial for lung homeostasis, damage, and restoration. Based on increasing evidence, the Notch pathway has been identified, as critical for fibrosis and subsequently, the development of chronic fibroproliferative conditions in various organs and tissues. Recent research indicates that deregulation of Notch signaling correlates with the pathogenesis of significant pulmonary conditions, particularly chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, asthma, pulmonary arterial hypertension (PAH), lung carcinoma, and pulmonary abnormalities in some hereditary disorders. In various cellular and tissue environments, and across both physiological and pathological conditions, multiple consequences of Notch activation have been observed. Studies have ascertained that the Notch signaling cascade exhibits close associations with various other signaling systems. This study provides an updated overview of Notch signaling's role, especially its link to fibrosis and its potential therapeutic implications. This study sheds light on the latest findings regarding the mechanisms and outcomes of irregular or lacking Notch activity in the onset and development of pulmonary diseases. As our insight into this signaling mechanism suggests that modulating Notch signaling might hold potential as a valuable additional therapeutic approach in upcoming research.
Collapse
Affiliation(s)
- Samar A Antar
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA, 24016, USA.
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt.
| | - Mohamed Kh ElMahdy
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| | - Ahmed G Darwish
- Center for Viticulture and Small Fruit Research, College of Agriculture and Food Sciences, Florida A&M University, Tallahassee, FL, 32308, USA
| |
Collapse
|
4
|
Baral B, Saini V, Kandpal M, Kundu P, Dixit AK, Parmar HS, Meena AK, Trivedi P, Jha HC. The interplay of co-infections in shaping COVID-19 severity: Expanding the scope beyond SARS-CoV-2. J Infect Public Health 2024; 17:102486. [PMID: 39002466 DOI: 10.1016/j.jiph.2024.102486] [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: 12/10/2023] [Revised: 06/17/2024] [Accepted: 06/23/2024] [Indexed: 07/15/2024] Open
Abstract
High mortality has been reported in severe cases of COVID-19. Emerging reports suggested that the severity is not only due to SARS-CoV-2 infection, but also due to coinfections by other pathogens exhibiting symptoms like COVID-19. During the COVID-19 pandemic, simultaneous respiratory coinfections with various viral (Retroviridae, Flaviviridae, Orthomyxoviridae, and Picoviridae) and bacterial (Mycobacteriaceae, Mycoplasmataceae, Enterobacteriaceae and Helicobacteraceae) families have been observed. These pathogens intensify disease severity by potentially augmenting SARSCoV-2 replication, inflammation, and modulation of signaling pathways. Coinfection emerges as a critical determinant of COVID-19 severity, principally instigated by heightened pro-inflammatory cytokine levels, as cytokine storm. Thereby, in co-infection scenario, the severity is also driven by the modulation of inflammatory signaling pathways by both pathogens possibly associated with interleukin, interferon, and cell death exacerbating the severity. In the current review, we attempt to understand the role of co- infections by other pathogens and their involvement in the severity of COVID-19.
Collapse
Affiliation(s)
- Budhadev Baral
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Vaishali Saini
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Meenakshi Kandpal
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Pratik Kundu
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Amit Kumar Dixit
- Central Ayurveda Research Institute, 4-CN Block, Sector -V, Bidhannagar, Kolkata 700 091, India
| | - Hamendra Singh Parmar
- School of Biotechnology, Devi Ahilya Vishwavidyalaya, Takshashila Campus, Indore, Madhya Pradesh 452001, India
| | - Ajay Kumar Meena
- Regional Ayurveda Research Institute, Gwalior, Amkhoh, Gwalior, Madhya Pradesh 474001, India
| | - Pankaj Trivedi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India; Centre for Rural Development and Technology, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India.
| |
Collapse
|
5
|
de Almeida Lopes C, Wang J, Liffner B, Absalon S, Gazzinelli-Guimaraes PH. Ascaris Mouse Model Protocols: Advancing Research on Larval Ascariasis Biology. Curr Protoc 2024; 4:e1074. [PMID: 38923794 PMCID: PMC11215937 DOI: 10.1002/cpz1.1074] [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] [Indexed: 06/28/2024]
Abstract
Ascariasis, caused by both Ascaris lumbricoides and Ascaris suum, is the most prevalent parasitic disease worldwide, affecting both human and porcine populations. However, due to the difficulties of assessing the early events of infection in humans, most studies of human ascariasis have been restricted to the chronic intestinal phase. Therefore, the Ascaris mouse model has become a fundamental tool for investigating the immunobiology and pathogenesis of the early infection stage referred to as larval ascariasis because of the model's practicality and ability to replicate the natural processes involved. The Ascaris mouse model has been widely used to explore factors such as infection resistance/susceptibility, liver inflammation, lung immune-mediated pathology, and co-infections and, notably, as a pivotal element in preclinical vaccine trials. Exploring the immunobiology of larval ascariasis may offer new insights into disease development and provide a substantial understanding of key components that trigger a protective immune response. This article focuses on creating a comprehensive guide for conducting Ascaris experimental infections in the laboratory as a foundation for future research efforts. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Acquisition and embryonation of Ascaris suum eggs from adult females Alternate Protocol: Cleaning and purification of Ascaris suum from female A. suum uteri Basic Protocol 2: Preparation of Ascaris suum eggs and murine infection Basic Protocol 3: Measurement of larval burden and Ascaris-larva-induced pathogenesis Basic Protocol 4: In vitro hatching and purification of Ascaris L3 larvae Support Protocol: Preparation of crude antigen from Ascaris infectious stages Basic Protocol 5: Ultrastructure-expansion microscopy (U-ExM) of Ascaris suum larval stages.
Collapse
Affiliation(s)
- Camila de Almeida Lopes
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
- Laboratory of Immunobiology and Parasites Control, Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jianbin Wang
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee
| | - Benjamin Liffner
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sabrina Absalon
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Pedro H Gazzinelli-Guimaraes
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia
| |
Collapse
|
6
|
Cheng DH, Jiang TG, Zeng WB, Li TM, Jing YD, Li ZQ, Guo YH, Zhang Y. Identification and coregulation pattern analysis of long noncoding RNAs in the mouse brain after Angiostrongylus cantonensis infection. Parasit Vectors 2024; 17:205. [PMID: 38715092 PMCID: PMC11077716 DOI: 10.1186/s13071-024-06278-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Angiostrongyliasis is a highly dangerous infectious disease. Angiostrongylus cantonensis larvae migrate to the mouse brain and cause symptoms, such as brain swelling and bleeding. Noncoding RNAs (ncRNAs) are novel targets for the control of parasitic infections. However, the role of these molecules in A. cantonensis infection has not been fully clarified. METHODS In total, 32 BALB/c mice were randomly divided into four groups, and the infection groups were inoculated with 40 A. cantonensis larvae by gavage. Hematoxylin and eosin (H&E) staining and RNA library construction were performed on brain tissues from infected mice. Differential expression of long noncoding RNAs (lncRNAs) and mRNAs in brain tissues was identified by high-throughput sequencing. The pathways and functions of the differentially expressed lncRNAs were determined by Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses. The functions of the differentially expressed lncRNAs were further characterized by lncRNA‒microRNA (miRNA) target interactions. The potential host lncRNAs involved in larval infection of the brain were validated by quantitative real-time polymerase chain reaction (qRT‒PCR). RESULTS The pathological results showed that the degree of brain tissue damage increased with the duration of infection. The transcriptome results showed that 859 lncRNAs and 1895 mRNAs were differentially expressed compared with those in the control group, and several lncRNAs were highly expressed in the middle-late stages of mouse infection. GO and KEGG pathway analyses revealed that the differentially expressed target genes were enriched mainly in immune system processes and inflammatory response, among others, and several potential regulatory networks were constructed. CONCLUSIONS This study revealed the expression profiles of lncRNAs in the brains of mice after infection with A. cantonensis. The lncRNAs H19, F630028O10Rik, Lockd, AI662270, AU020206, and Mexis were shown to play important roles in the infection of mice with A. cantonensis infection.
Collapse
Affiliation(s)
- Dong-Hui Cheng
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (National Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases; WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, People's Republic of China
| | - Tian-Ge Jiang
- School of Global Health, National Center for Tropical Disease Research, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Wen-Bo Zeng
- School of Life Sciences, Fudan University, Shanghai, People's Republic of China
| | - Tian-Mei Li
- Dali Prefectural Institute of Research and Control On Schistosomiasis, Yunnan, People's Republic of China
| | - Yi-Dan Jing
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (National Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases; WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, People's Republic of China
| | - Zhong-Qiu Li
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (National Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases; WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, People's Republic of China
| | - Yun-Hai Guo
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (National Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases; WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, People's Republic of China
| | - Yi Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (National Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases; WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, People's Republic of China.
- School of Global Health, National Center for Tropical Disease Research, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| |
Collapse
|
7
|
Palkumbura PGAS, Mahakapuge TAN, Wijesundera RRMKK, Wijewardana V, Kangethe RT, Rajapakse RPVJ. Mucosal Immunity of Major Gastrointestinal Nematode Infections in Small Ruminants Can Be Harnessed to Develop New Prevention Strategies. Int J Mol Sci 2024; 25:1409. [PMID: 38338687 PMCID: PMC10855138 DOI: 10.3390/ijms25031409] [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: 12/01/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Gastrointestinal parasitic nematode (GIN) infections are the cause of severe losses to farmers in countries where small ruminants such as sheep and goat are the mainstay of livestock holdings. There is a need to develop effective and easy-to-administer anti-parasite vaccines in areas where anthelmintic resistance is rapidly rising due to the inefficient use of drugs currently available. In this review, we describe the most prevalent and economically significant group of GIN infections that infect small ruminants and the immune responses that occur in the host during infection with an emphasis on mucosal immunity. Furthermore, we outline the different prevention strategies that exist with a focus on whole and purified native parasite antigens as vaccine candidates and their possible oral-nasal administration as a part of an integrated parasite control toolbox in areas where drug resistance is on the rise.
Collapse
Affiliation(s)
- P. G. Ashani S. Palkumbura
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Kandy 20400, Sri Lanka
| | - Thilini A. N. Mahakapuge
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Kandy 20400, Sri Lanka
| | - R. R. M. K. Kavindra Wijesundera
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Kandy 20400, Sri Lanka
| | - Viskam Wijewardana
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, 2444 Seibersdorf, Austria
| | - Richard Thiga Kangethe
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, 2444 Seibersdorf, Austria
| | - R. P. V. Jayanthe Rajapakse
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Kandy 20400, Sri Lanka
| |
Collapse
|
8
|
Oliveira FMS, Kraemer L, Vieira-Santos F, Leal-Silva T, Gazzinelli-Guimarães AC, Lopes CA, Amorim CCO, Pinheiro GRG, Moura MS, Matias PHP, Barbosa FS, Caliari MV, Weatherhead JE, Bueno LL, Russo RC, Fujiwara RT. The long-lasting Ascaris suum antigens in the lungs shapes the tissue adaptation modifying the pulmonary architecture and immune response after infection in mice. Microb Pathog 2024; 186:106483. [PMID: 38092133 DOI: 10.1016/j.micpath.2023.106483] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/09/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023]
Abstract
Ascariasis is the most prevalent helminth affecting approximately 819 million people worldwide. The acute phase of Ascariasis is characterized by larval migration of Ascaris spp., through the intestinal wall, carried to the liver and lungs of the host by the circulatory system. Most of the larvae subsequently transverse the lung parenchyma leading to tissue injury, reaching the airways and pharynx, where they can be expectorated and swallowed back to the gastrointestinal tract, where they develop into adult worms. However, some larvae are trapped in the lung parenchyma inciting an inflammatory response that causes persistent pulmonary tissue damage long after the resolution of infection, which returns to tissue homeostasis. However, the mechanism by which chronic lung disease develops and resolves remains unknown. Here, using immunohistochemistry, we demonstrate that small fragments and larval antigens of Ascaris suum are deposited and retained chronically in the lung parenchyma of mice following a single Ascaris infection. Our results reveal that the prolonged presence of Ascaris larval antigens in the lung parenchyma contributes to the persistent immune stimulation inducing histopathological changes observed chronically following infection, and clearly demonstrate that larval antigens are related to all phases of tissue adaptation after infection: lung injury, chronic inflammation, resolution, and tissue remodeling, in parallel to increased specific humoral immunity and the recovery of lung function in mice. Additional insight is needed into the mechanisms of Ascaris antigen to induce chronic immune responses and resolution in the host lungs following larval migration.
Collapse
Affiliation(s)
- Fabrício M S Oliveira
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Cellular and Molecular Immunology Group, René Rachou Institute, Oswaldo Cruz Foundation - FIOCRUZ, Av. Augusto de Lima, 1.715, Belo Horizonte, Minas Gerais, Brazil
| | - Lucas Kraemer
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flaviane Vieira-Santos
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thaís Leal-Silva
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana C Gazzinelli-Guimarães
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila A Lopes
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Chiara C O Amorim
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Guilherme R G Pinheiro
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus S Moura
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pablo H P Matias
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Marcelo V Caliari
- Laboratory of Protozooses, Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jill E Weatherhead
- Department of Medicine, Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Lilian L Bueno
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo C Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo T Fujiwara
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
9
|
Okon EM, Okocha RC, Taiwo AB, Michael FB, Bolanle AM. Dynamics of co-infection in fish: A review of pathogen-host interaction and clinical outcome. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2023; 4:100096. [PMID: 37250211 PMCID: PMC10213192 DOI: 10.1016/j.fsirep.2023.100096] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 04/09/2023] [Accepted: 05/15/2023] [Indexed: 05/31/2023] Open
Abstract
Co-infections can affect the transmission of a pathogen within a population and the pathogen's virulence, ultimately affecting the disease's dynamics. In addition, co-infections can potentially affect the host's immunological responses, clinical outcomes, survival, and disease control efficacy. Co-infections significantly impact fish production and can change several fish diseases' progression and severity. However, the effect of co-infection has only recently garnered limited attention in aquatic animals such as fish, and there is currently a dearth of studies on this topic. This study, therefore, presents an in-depth summary of the dynamics of co-infection in fish. This study reviewed the co-infection of fish pathogens, the interaction of pathogens and fish, clinical outcomes and impacts on fish immune responses, and fish survival. Most studies described the prevalence of co-infections in fish, with various parameters influencing their outcomes. Bacterial co-infection increased fish mortality, ulcerative dermatitis, and intestinal haemorrhage. Viral co-infection resulted in osmoregulatory effects, increased mortality and cytopathic effect (CPE). More severe histological alterations and clinical symptoms were related to the co-infection of fish than in single-infected fish. In parasitic co-infection, there was increased mortality, high kidney swelling index, and severe necrotic alterations in the kidney, liver, and spleen. In other cases, there were more severe kidney lesions, cartilage destruction and displacement. There was a dearth of information on mitigating co-infections in fish. Therefore, further studies on the mitigation strategies of co-infections in fish will provide valuable insights into this research area. Also, more research on the immunology of co-infection specific to each fish pathogen class (bacteria, viruses, fungi, and parasites) is imperative. The findings from such studies would provide valuable information on the relationship between fish immune systems and targeted responses.
Collapse
Affiliation(s)
| | - Reuben Chukwuka Okocha
- Department of Animal Science, College of Agricultural Sciences, Landmark University, P.M.B. 1001 Omu-Aran, Kwara State, Nigeria
- Climate Action Research Group, Landmark University SDG 13, Nigeria
| | | | - Falana Babatunde Michael
- Department of Animal Science, College of Agricultural Sciences, Landmark University, P.M.B. 1001 Omu-Aran, Kwara State, Nigeria
- Life Below Water Research Group, Landmark University SDG 14, Nigeria
| | - Adeniran Moji Bolanle
- Department of Animal Science, College of Agricultural Sciences, Landmark University, P.M.B. 1001 Omu-Aran, Kwara State, Nigeria
| |
Collapse
|
10
|
Gazzinelli-Guimaraes PH, Golec DP, Karmele EP, Sciurba J, Bara-Garcia P, Hill T, Kang B, Bennuru S, Schwartzberg PL, Nutman TB. Eosinophil trafficking in allergen-mediated pulmonary inflammation relies on IL-13-driven CCL-11 and CCL-24 production by tissue fibroblasts and myeloid cells. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100131. [PMID: 37781651 PMCID: PMC10509988 DOI: 10.1016/j.jacig.2023.100131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/19/2023] [Accepted: 04/30/2023] [Indexed: 10/03/2023]
Abstract
Background The immunologic mechanisms underlying pulmonary type 2 inflammation, including the dynamics of eosinophil recruitment to the lungs, still need to be elucidated. Objective We sought to investigate how IL-13-producing TH2 effector cells trigger eosinophil migration in house dust mite (HDM)-driven allergic pulmonary inflammation. Methods Multiparameter and molecular profiling of murine lungs with HDM-induced allergy was investigated in the absence of IL-13 signaling by using IL-13Rα1-deficient mice and separately through adoptive transfer of CD4+ T cells from IL-5-deficient mice into TCRα-/- mice before allergic inflammation. Results We demonstrated through single-cell techniques that HDM-driven pulmonary inflammation displays a profile characterized by TH2 effector cell-induced IL-13-dominated eosinophilic inflammation. Using HDM-sensitized IL-13Rα1-/- mice, we found a marked reduction in the influx of eosinophils into the lungs along with a significant downregulation of both CCL-11 and CCL-24. We further found that eosinophil trafficking to the lung relies on production of IL-13-driven CCL-11 and CCL-24 by fibroblasts and Ly6C+ (so-called classical) monocytes. Moreover, this IL-13-mediated eotaxin-dependent eosinophil influx to the lung tissue required IL-5-induced eosinophilia. Finally, we demonstrated that this IL-13-driven eosinophil-dominated pulmonary inflammation was critical for limiting bystander lung transiting Ascaris parasites in a model of allergy and helminth interaction. Conclusion Our data suggest that IL-5-dependent allergen-specific TH2 effector cell response and subsequent signaling through the IL-13/IL-13Rα1 axis in fibroblasts and myeloid cells regulate the eotaxin-dependent recruitment of eosinophils to the lungs, with multiple downstream consequences, including bystander control of lung transiting parasitic helminths.
Collapse
Affiliation(s)
| | - Dominic P. Golec
- Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, MD
| | - Erik P. Karmele
- Laboratory of Molecular Immunology, NIAID, National Institutes of Health, Bethesda, MD
| | - Joshua Sciurba
- Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD
| | - Pablo Bara-Garcia
- Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD
| | - Tom Hill
- National Institute of Allergy and Infectious Diseases (NIAID) Collaborative Bioinformatics Resource, NIAID, National Institutes of Health, Bethesda, MD
| | - Byunghyun Kang
- Laboratory of Molecular Immunology, NIAID, National Institutes of Health, Bethesda, MD
| | - Sasisekhar Bennuru
- Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD
| | | | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD
| |
Collapse
|
11
|
Abstract
The biology of a cell, whether it is a unicellular organism or part of a multicellular network, is influenced by cell type, temporal changes in cell state, and the cell's environment. Spatial cues play a critical role in the regulation of microbial pathogenesis strategies. Information about where the pathogen is-in a tissue or in proximity to a host cell-regulates gene expression and the compartmentalization of gene products in the microbe and the host. Our understanding of host and pathogen identity has bloomed with the accessibility of transcriptomics and proteomics techniques. A missing piece of the puzzle has been our ability to evaluate global transcript and protein expression in the context of the subcellular niche, primary cell, or native tissue environment during infection. This barrier is now lower with the advent of new spatial omics techniques to understand how location regulates cellular functions. This review will discuss how recent advances in spatial proteomics and transcriptomics approaches can address outstanding questions in microbial pathogenesis.
Collapse
Affiliation(s)
- Samantha Lempke
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Dana May
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Sarah E. Ewald
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
12
|
You H, Jones MK, Gordon CA, Arganda AE, Cai P, Al-Wassiti H, Pouton CW, McManus DP. The mRNA Vaccine Technology Era and the Future Control of Parasitic Infections. Clin Microbiol Rev 2023; 36:e0024121. [PMID: 36625671 PMCID: PMC10035331 DOI: 10.1128/cmr.00241-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Despite intensive long-term efforts, with very few exceptions, the development of effective vaccines against parasitic infections has presented considerable challenges, given the complexity of parasite life cycles, the interplay between parasites and their hosts, and their capacity to escape the host immune system and to regulate host immune responses. For many parasitic diseases, conventional vaccine platforms have generally proven ill suited, considering the complex manufacturing processes involved and the costs they incur, the inability to posttranslationally modify cloned target antigens, and the absence of long-lasting protective immunity induced by these antigens. An effective antiparasite vaccine platform is required to assess the effectiveness of novel vaccine candidates at high throughput. By exploiting the approach that has recently been used successfully to produce highly protective COVID mRNA vaccines, we anticipate a new wave of research to advance the use of mRNA vaccines to prevent parasitic infections in the near future. This article considers the characteristics that are required to develop a potent antiparasite vaccine and provides a conceptual foundation to promote the development of parasite mRNA-based vaccines. We review the recent advances and challenges encountered in developing antiparasite vaccines and evaluate the potential of developing mRNA vaccines against parasites, including those causing diseases such as malaria and schistosomiasis, against which vaccines are currently suboptimal or not yet available.
Collapse
Affiliation(s)
- Hong You
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Malcolm K. Jones
- School of Veterinary Science, The University of Queensland, Brisbane, Australia
| | - Catherine A. Gordon
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Alexa E. Arganda
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Pengfei Cai
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Harry Al-Wassiti
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Colin W. Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Donald P. McManus
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
13
|
In silico design of a polypeptide as a vaccine candidate against ascariasis. Sci Rep 2023; 13:3504. [PMID: 36864139 PMCID: PMC9981566 DOI: 10.1038/s41598-023-30445-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Ascariasis is the most prevalent zoonotic helminthic disease worldwide, and is responsible for nutritional deficiencies, particularly hindering the physical and neurological development of children. The appearance of anthelmintic resistance in Ascaris is a risk for the target of eliminating ascariasis as a public health problem by 2030 set by the World Health Organisation. The development of a vaccine could be key to achieving this target. Here we have applied an in silico approach to design a multi-epitope polypeptide that contains T-cell and B-cell epitopes of reported novel potential vaccination targets, alongside epitopes from established vaccination candidates. An artificial toll-like receptor-4 (TLR4) adjuvant (RS09) was added to improve immunogenicity. The constructed peptide was found to be non-allergic, non-toxic, with adequate antigenic and physicochemical characteristics, such as solubility and potential expression in Escherichia coli. A tertiary structure of the polypeptide was used to predict the presence of discontinuous B-cell epitopes and to confirm the molecular binding stability with TLR2 and TLR4 molecules. Immune simulations predicted an increase in B-cell and T-cell immune response after injection. This polypeptide can now be validated experimentally and compared to other vaccine candidates to assess its possible impact in human health.
Collapse
|
14
|
Bioaccessibility and oral immunization efficacy of a chimeric protein vaccine against Ascaris suum. Microbes Infect 2023; 25:105042. [PMID: 36075515 DOI: 10.1016/j.micinf.2022.105042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/02/2022] [Accepted: 08/27/2022] [Indexed: 02/03/2023]
Abstract
Human ascariasis has been characterized as the most prevalent neglected tropical disease worldwide. There is an urgent need for search to alternative prevention and control methods for ascariasis. Here we aimed to establish a protocol of oral immunization with a previously described chimera protein capable of resist through digestion and induce mucous protection against Ascaris suum infection. Mice were oral immunized with seven doses with one day interval and challenged with A. suum ten days after the last dose. In vitro digestion showed that 64% of chimeric protein was bioaccessible for absorption after digestion. Immunized mice display 66,2% reduction of larval burden in lungs compared to control group. In conclusion we demonstrated that oral immunization with chimera protein protects the host against A. suum larval migration leading to less pronounced histopathological lesions.
Collapse
|
15
|
Ekobol N, Boonjaraspinyo S, Artchayasawat A, Boonmars T. Monks: A Population at Risk for Liver Fluke and Skin-Penetrating Helminths. Trop Med Infect Dis 2023; 8:tropicalmed8030135. [PMID: 36977136 PMCID: PMC10059027 DOI: 10.3390/tropicalmed8030135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/06/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023] Open
Abstract
Monks cannot cook received raw meat dishes and should walk barefoot while working. This population lacks both a survey of parasitic infection and a proper prevention and control policy. Five hundred and fourteen monks from the Ubolratana, Ban Haet, and Ban Phai Districts of Kh on Kaen Province were enrolled in this study. A stool container and questionnaire were collected from each study participant. Stool samples were processed by formalin ethyl acetate concentration and agar plate culture techniques. We then analyzed the results and risk factors to demonstrate associations. The prevalence of overall parasites, liver flukes, and skin-penetrating helminths were 28.8%, 11.1%, and 19.3%, respectively. Raw fish dish offerings were associated with opisthorchiasis (ORcrude 3.32; 95% CI 1.53–7.20). The risk factors for skin-penetrating helminths were older age (ORcrude 5.02; 95% CI 2.2–11.17), being a long-term ordinate (ORcrude 3.28; 95% CI 1.15–9.34), smoking (ORcrude 2.03; 95% CI 1.23–3.36), and chronic kidney disease with other underlying disease (ORcrude 20.7; 95% CI 2.54–190.1). The protective factors for skin-penetrating helminths were secular education above primary education (ORcrude 0.41; 95% CI 0.25–0.65) and having received health education about parasitic infection (ORcrude 0.47; 95% CI 0.28–0.80). Wearing shoes at times other than alms work does not show a protective effect against skin-penetrating helminths (ORcrude 0.86; 95% CI 0.51–1.46). These findings support the recommendation for a strict Rule of Discipline regarding raw meat ingestion and allowing shoes to be worn for protection against skin-penetrating helminths in high-risk situations.
Collapse
Affiliation(s)
- Nuttapon Ekobol
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sirintip Boonjaraspinyo
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
- Department of Community Medicine, Family Medicine and Occupational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Atchara Artchayasawat
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Thidarut Boonmars
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
- Correspondence: ; Tel.: +66-43-363-434
| |
Collapse
|
16
|
Thapa B, Parajuli RP, Dhakal P. Prevalence and burden of gastrointestinal parasites in stray cattle of the Kathmandu Valley. J Parasit Dis 2022; 46:845-853. [PMID: 36091270 PMCID: PMC9458840 DOI: 10.1007/s12639-022-01499-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 05/10/2022] [Indexed: 11/30/2022] Open
Abstract
Gastrointestinal parasites (GIPs) are common among cattle resulting severe infection. Prevalence of GIPs in stray street cattle may pose risk of dissemination of parasites of zoonotic importance. This study was conducted to determine the prevalence of GIPs in stray cattle of the Kathmandu valley. One hundred (n = 100) freshly voided dung samples were collected from eight sites. The samples were processed using the concentration method for microscopic examination, and modified McMaster technique for quantification of mean eggs/oocysts per gram of feces (EPG/OPG). Results revealed that 72% of the cattle were found positive for one or more species of GIPs and nine genera of GIPs were recorded (Eimeria, Ostertagia, Haemonchus, Trichostrongylus, Capillaria, Trichuris, Toxocara, Fasciola and Paramphistomum). The prevalence of parasitic infection was higher in males (73.68%) than in females (69.76%). The prevalence was found to be highest in adults (63.89%) followed by heifers or steers (27.78%) and calves (8.33%). Approximately, 76% of the cross breed and 65% of the local breed of cattle were positive for parasitic infection. The parasites differed both in prevalence and intensity. Eimeria sp. was the most prevalent (27%) species and had the highest intensity (858.02 OPG ± 63.46 SD). To our knowledge, this is the first research of its kind in relation to stray cattle in Nepal. Our findings reveal that there is a burden of helminth infections of zoonotic and socioeconomic importance in straycattle. Therefore, we recommend regular inspection of stray cattle and relevant preventive measures be put in place.
Collapse
Affiliation(s)
- Bigyan Thapa
- Central Department of Zoology (CDZ), Institute of Science and Technology (IoST), Tribhuvan University (TU), Kirtipur, Kathmandu, Nepal
| | - Rajendra Prasad Parajuli
- Central Department of Zoology (CDZ), Institute of Science and Technology (IoST), Tribhuvan University (TU), Kirtipur, Kathmandu, Nepal
| | - Pitambar Dhakal
- Central Department of Zoology (CDZ), Institute of Science and Technology (IoST), Tribhuvan University (TU), Kirtipur, Kathmandu, Nepal
| |
Collapse
|
17
|
Chang G, Li N, Wang Q, Ding J, Liu S, Hua L, Li S, Wang W. Dynamic transcriptome landscape of pulmonary tissues of rats infected with Paragonimus proliferus. Am J Transl Res 2022; 14:3395-3406. [PMID: 35702118 PMCID: PMC9185055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/30/2022] [Indexed: 06/15/2023]
Abstract
Paragonimiasis (pulmonary fluke disease) is a foodborne parasitic disease caused by trematode infections. Paragonimus proliferus is a characteristic Paragonimus species that was first identified in Yunnan Province of China. No direct evidence has yet proven that P. proliferus can infect humans. However, we previously found that P. proliferus infects and damages rat lung tissues via an unclear mechanism. Here, we infected Sprague Dawley rats with P. proliferus and sequenced their lung transcriptomes at various intervals thereafter. We detected P. proliferus on the surface of rat lung tissues at 7 days post infection. It colonized by attaching and secreting dsRNA and utilized nutrients from the lung tissues for mitosis and meiosis and the dynein arm of lung tissues to develop symmetrical organs. The rats generated different types of immune responses that differed according to the stage of infection. We then analyzed P. proliferus responses to these immune strategies and the genes expressed during each stage of infection. Our findings provide a foundation for developing medical treatments for P. proliferus infection.
Collapse
Affiliation(s)
- Guoji Chang
- Clinical-Medical Center of Infectious Disease of Yunnan Province, The Third People’s Hospital of KunmingKunming 650043, Yunnan, China
| | - Na Li
- Department of Pharmacy, The Third People’s Hospital of KunmingKunming 650043, Yunnan, China
| | - Qingqing Wang
- Clinical-Medical Center of Infectious Disease of Yunnan Province, The Third People’s Hospital of KunmingKunming 650043, Yunnan, China
| | - Jie Ding
- Clinical-Medical Center of Infectious Disease of Yunnan Province, The Third People’s Hospital of KunmingKunming 650043, Yunnan, China
| | - Siqi Liu
- Clinical-Medical Center of Infectious Disease of Yunnan Province, The Third People’s Hospital of KunmingKunming 650043, Yunnan, China
| | - Lijuan Hua
- Clinical-Medical Center of Infectious Disease of Yunnan Province, The Third People’s Hospital of KunmingKunming 650043, Yunnan, China
| | - Shenghao Li
- Clinical-Medical Center of Infectious Disease of Yunnan Province, The Third People’s Hospital of KunmingKunming 650043, Yunnan, China
| | - Wenlin Wang
- School of Basic Medicine, Kunming Medical UniversityKunming 650504, Yunnan, China
| |
Collapse
|
18
|
Long SR, Shang WX, Jiang M, Li JF, Liu RD, Wang ZQ, Sun H, Cui J. Preexisting Trichinella spiralis infection attenuates the severity of Pseudomonas aeruginosa-induced pneumonia. PLoS Negl Trop Dis 2022; 16:e0010395. [PMID: 35500031 PMCID: PMC9098000 DOI: 10.1371/journal.pntd.0010395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/12/2022] [Accepted: 04/05/2022] [Indexed: 11/19/2022] Open
Abstract
Background A range of helminth species involve the migration of developing larvae through the lung and establish chronic infections in the host that include potent immune regulatory effects. Trichinella spiralis is one of the most successful parasitic symbiotes. After released by intestinal female adult worms, newborn larvae of T. spiralis travel through the circulatory system to the lung and finally reach skeletal muscle cells. As unique inflammation modulator of intracellular parasitism, T. spiralis shows improved responses to autoimmune disease and viral pulmonary inflammation by exerting immunomodulatory effects on innate and adaptive immune cells. Methodology/Principal findings C57BL/6 mice were divided into four groups: uninfected; helminth- T. spiralis infected; P. aeruginosa infected; and co-infected. Mice infected with T. spiralis were incubated for 6 weeks, followed by P. aeruginosa intranasal inoculation. Bronchial alveolar lavage fluid, blood and lung samples were analyzed. We found that T. spiralis induced Th2 response in the mouse lung tissue, increased lung CD4+ T cells, GATA3, IL-4, IL-5 and IL-13 expression. Pre-existing T. spiralis infection decreased lung neutrophil recruitment, inflammatory mediator IL-1β and IL-6 expression and chemokine CXCL1 and CXCL2 release during P. aeruginosa- pneumonia. Furthermore, T. spiralis co-infected mice exhibited significantly more eosinophils at 6 hours following P. aeruginosa infection, ameliorated pulmonary inflammation and improved survival in P. aeruginosa pneumonia. Conclusions These findings indicate that a prior infection with T. spiralis ameliorates experimental pulmonary inflammation and improves survival in P. aeruginosa pneumonia through a Th2-type response with eosinophils. Helminth infections elicit type 2 immunity, which influences host immune responses to additional threats, such as allergens, metabolic disease and other pathogens. Pseudomonas aeruginosa is one of the most common gram-negative pathogens causing pneumonia in immunocompromised patients. The mortality rate of ventilator associated pneumonia caused by P. aeruginosa is higher than that due to other pathogens. Trichinella spiralis is a zoonotic nematode of intracellular parasitism that infects a wide range of vertebrate hosts, including humans. There is a lung migratory phase in the life cycle of T. spiralis. In this study, we found that T. spiralis induced Th2 response in the mouse lung tissue. T. spiralis co-infected mice exhibited significantly more eosinophils and less neutrophils at 6 hours following P. aeruginosa infection, ameliorated pulmonary inflammation and improved survival in P. aeruginosa pneumonia. These findings suggest a pre-existing chronic helminth with a lung migration phase infection promotes the survival of bacterial airway co-infected host.
Collapse
Affiliation(s)
- Shao Rong Long
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Wen Xuan Shang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
- Biology, School of Life Scence, Zhengzhou University, Zhengzhou, China
| | - Miao Jiang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Jing Fei Li
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Hualei Sun
- Department of Nutrition, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- * E-mail: (HS); (JC)
| | - Jing Cui
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
- * E-mail: (HS); (JC)
| |
Collapse
|
19
|
Kraemer L, McKay DM, Russo RC, Fujiwara RT. Chemokines and chemokine receptors: insights from human disease and experimental models of helminthiasis. Cytokine Growth Factor Rev 2022; 66:38-52. [DOI: 10.1016/j.cytogfr.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 11/03/2022]
|
20
|
Oyesola OO, Souza COS, Loke P. The Influence of Genetic and Environmental Factors and Their Interactions on Immune Response to Helminth Infections. Front Immunol 2022; 13:869163. [PMID: 35572520 PMCID: PMC9103684 DOI: 10.3389/fimmu.2022.869163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/04/2022] [Indexed: 12/20/2022] Open
Abstract
Helminth infection currently affect over 2 billion people worldwide, with those with the most pathologies and morbidities, living in regions with unequal and disproportionate access to effective healthcare solutions. Host genetics and environmental factors play critical roles in modulating and regulating immune responses following exposure to various pathogens and insults. However, the interplay of environment and genetic factors in influencing who gets infected and the establishment, persistence, and clearance of helminth parasites remains unclear. Inbred strains of mice have long been used to investigate the role of host genetic factors on pathogenesis and resistance to helminth infection in a laboratory setting. This review will discuss the use of ecological and environmental mouse models to study helminth infections and how this could be used in combination with host genetic variation to explore the relative contribution of these factors in influencing immune response to helminth infections. Improved understanding of interactions between genetics and the environment to helminth immune responses would be important for efforts to identify and develop new prophylactic and therapeutic options for the management of helminth infections and their pathogenesis.
Collapse
Affiliation(s)
- Oyebola O. Oyesola
- Laboratory of Parasitic Disease, National Institute of Allergy and Infectious Disease (NIAID), National Institute of Health, Bethesda, MD, United States
| | | | | |
Collapse
|
21
|
Andersen-Civil AIS, Myhill LJ, Büdeyri Gökgöz N, Engström MT, Mejer H, Zhu L, Zeller WE, Salminen JP, Krych L, Lauridsen C, Nielsen DS, Thamsborg SM, Williams AR. Dietary proanthocyanidins promote localized antioxidant responses in porcine pulmonary and gastrointestinal tissues during Ascaris suum-induced type 2 inflammation. FASEB J 2022; 36:e22256. [PMID: 35333423 DOI: 10.1096/fj.202101603rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/16/2022] [Accepted: 03/07/2022] [Indexed: 11/11/2022]
Abstract
Proanthocyanidins (PAC) are dietary polyphenols with putative anti-inflammatory and immunomodulatory effects. However, whether dietary PAC can regulate type-2 immune function and inflammation at mucosal surfaces remains unclear. Here, we investigated if diets supplemented with purified PAC modulated pulmonary and intestinal mucosal immune responses during infection with the helminth parasite Ascaris suum in pigs. A. suum infection induced a type-2 biased immune response in lung and intestinal tissues, characterized by pulmonary granulocytosis, increased Th2/Th1 T cell ratios in tracheal-bronchial lymph nodes, intestinal eosinophilia, and modulation of genes involved in mucosal barrier function and immunity. Whilst PAC had only minor effects on pulmonary immune responses, RNA-sequencing of intestinal tissues revealed that dietary PAC significantly enhanced transcriptional responses related to immune function and antioxidant responses in the gut of both naïve and A. suum-infected animals. A. suum infection and dietary PAC induced distinct changes in gut microbiota composition, primarily in the jejunum and colon, respectively. Notably, PAC consumption substantially increased the abundance of Limosilactobacillus reuteri. In vitro experiments with porcine macrophages and intestinal epithelial cells supported a role for both PAC polymers and PAC-derived microbial metabolites in regulating oxidative stress responses in host tissues. Thus, dietary PAC may have distinct beneficial effects on intestinal health during infection with mucosal pathogens, while having a limited activity to modulate naturally-induced type-2 pulmonary inflammation. Our results shed further light on the mechanisms underlying the health-promoting properties of PAC-rich foods, and may aid in the design of novel dietary supplements to regulate mucosal inflammatory responses in the gastrointestinal tract.
Collapse
Affiliation(s)
| | - Laura J Myhill
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Marica T Engström
- Natural Chemistry Research Group, Department of Chemistry, University of Turku, Turku, Finland
| | - Helena Mejer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ling Zhu
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Wayne E Zeller
- USDA-ARS, U.S. Dairy Forage Research Center, Madison, Wisconsin, USA
| | - Juha-Pekka Salminen
- Natural Chemistry Research Group, Department of Chemistry, University of Turku, Turku, Finland
| | - Lukasz Krych
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | | | - Dennis S Nielsen
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Stig M Thamsborg
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Andrew R Williams
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
22
|
Carvallo FR, Stevenson VB. Interstitial pneumonia and diffuse alveolar damage in domestic animals. Vet Pathol 2022; 59:586-601. [DOI: 10.1177/03009858221082228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Classification of pneumonia in animals has been controversial, and the most problematic pattern is interstitial pneumonia. This is true from the gross and histologic perspectives, and also from a mechanistic point of view. Multiple infectious and noninfectious diseases are associated with interstitial pneumonia, all of them converging in the release of inflammatory mediators that generate local damage and attract inflammatory cells that inevitably trigger a second wave of damage. Diffuse alveolar damage is one of the more frequently identified histologic types of interstitial pneumonia and involves injury to alveolar epithelial and/or endothelial cells, with 3 distinct stages. The first is the “exudative” stage, with alveolar edema and hyaline membranes. The second is the “proliferative” stage, with hyperplasia and reactive atypia of type II pneumocytes, infiltration of lymphocytes, plasma cells, and macrophages in the interstitium and early proliferation of fibroblasts. These stages are reversible and often nonfatal. If damage persists, there is a third “fibrosing” stage, characterized by fibrosis of the interstitium due to proliferation of fibroblasts/myofibroblasts, persistence of type II pneumocytes, segments of squamous metaplasia of alveolar epithelium, plus inflammation. Understanding the lesion patterns associated with interstitial pneumonias, their causes, and the underlying mechanisms aid in accurate diagnosis that involves an interdisciplinary collaborative approach involving pathologists, clinicians, and radiologists.
Collapse
Affiliation(s)
- Francisco R. Carvallo
- Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA
- Virginia Department of Agriculture and Consumer Services, Harrisonburg, VA
| | | |
Collapse
|
23
|
Kim SY, Barnes MA, Sureshchandra S, Menicucci AR, Patel JJ, Messaoudi I, Nair MG. CX3CR1-Expressing Myeloid Cells Regulate Host-Helminth Interaction and Lung Inflammation. Adv Biol (Weinh) 2022; 6:e2101078. [PMID: 35119218 PMCID: PMC8934291 DOI: 10.1002/adbi.202101078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/31/2021] [Indexed: 11/06/2022]
Abstract
Many helminth life cycles, including hookworm, involve a mandatory lung phase, where myeloid and granulocyte subsets interact with the helminth and respond to infection-induced lung injury. To evaluate these innate subsets in Nippostrongylus brasiliensis infection, reporter mice for myeloid cells (CX3CR1GFP ) and granulocytes (PGRPdsRED ) are employed. Nippostrongylus infection induces lung infiltration of reporter cells, including CX3CR1+ myeloid cells and PGRP+ eosinophils. Strikingly, CX3CR1GFP/GFP mice, which are deficient in CX3CR1, are protected from Nippostrongylus infection with reduced weight loss, lung leukocyte infiltration, and worm burden compared to CX3CR1+/+ mice. This protective effect is specific for CX3CR1 as CCR2-deficient mice do not exhibit reduced worm burdens. Nippostrongylus co-culture with lung Ly6C+ monocytes or CD11c+ cells demonstrates that CX3CR1GFP/GFP monocytes secrete more pro-inflammatory cytokines and actively bind the parasites causing reduced motility. RNA sequencing of Ly6C+ or CD11c+ cells shows Nippostrongylus-induced gene expression changes, particularly in monocytes, associated with inflammation, chemotaxis, and extracellular matrix remodeling pathways. Analysis reveals cytotoxic and adhesion molecules as potential effectors against the parasite, such as Gzma and Gzmb, which are elevated in CX3CR1GFP/GFP monocytes. These studies validate a dual innate cell reporter for lung helminth infection and demonstrate that CX3CR1 impairs monocyte-helminth interaction.
Collapse
Affiliation(s)
| | | | | | - Andrea R. Menicucci
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, California 92697-3900, United States
| | - Jay J. Patel
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California 92521, United States
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, California 92697-3900, United States
| | | |
Collapse
|
24
|
|
25
|
Gazzinelli-Guimarães AC, Gazzinelli-Guimarães P, Weatherhead JE. A historical and systematic overview of Ascaris vaccine development. Parasitology 2021; 148:1795-1805. [PMID: 35586777 PMCID: PMC9109942 DOI: 10.1017/s0031182021001347] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 11/06/2022]
Abstract
Ascariasis is the most prevalent helminth infection in the world and leads to significant, life-long morbidity, particularly in young children. Current efforts to control and eradicate ascariasis in endemic regions have been met with significant challenges including high-rates of re-infection and potential development of anthelminthic drug resistance. Vaccines against ascariasis are a key tool that could break the transmission cycle and lead to disease eradication globally. Evolution of the Ascaris vaccine pipeline has progressed, however no vaccine product has been brought to human clinical trials to date. Advancement in recombinant protein technology may provide the first step in generating an Ascaris vaccine as well as a pan-helminthic vaccine ready for human trials. However, several roadblocks remain and investment in new technologies will be important to develop a successful human Ascaris vaccine that is critically needed to prevent significant morbidity in Ascaris-endemic regions around the world.
Collapse
Affiliation(s)
| | | | - Jill E. Weatherhead
- Department of Medicine, Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Pediatric Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
26
|
Zhang C, Wang H, Li J, Hou X, Li L, Wang W, Shi Y, Li D, Li L, Zhao Z, Li L, Aji T, Lin R, Shao Y, Vuitton DA, Tian Z, Sun H, Wen H. Involvement of TIGIT in Natural Killer Cell Exhaustion and Immune Escape in Patients and Mouse Model With Liver Echinococcus multilocularis Infection. Hepatology 2021; 74:3376-3393. [PMID: 34192365 DOI: 10.1002/hep.32035] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Alveolar echinococcosis (AE) is a lethal helminthic liver disease caused by persistent infection with Echinococcus multilocularis. Although more attention has been paid to the immunotolerance of T cells caused by E. multilocularis infection, the role of natural killer (NK) cell, a critical player in liver immunity, is seldom studied. APPROACH AND RESULTS Here, we observed that NK cells from the blood and closed liver tissue (CLT) of AE patients expressed a higher level of inhibitory receptor TIGIT and were functionally exhausted with a lower expression of granzyme B, perforin, interferon-gamma (IFN-γ), and TNF-α. Addition of anti-TIGIT (T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain) monoclonal antibody into AE patients' peripheral blood mononuclear cell culture significantly enhanced the synthesis of IFN-γ and TNF-α by NK cells, indicating the reversion of exhausted NK cells by TIGIT blockade. In the mouse model of E. multilocularis infection, liver and splenic TIGIT+ NK cells progressively increased dependent of infection dosage and timing and were less activated and less degranulated with lower cytokine secretion. Furthermore, TIGIT deficiency or blockade in vivo inhibited liver metacestode growth, reduced liver injury, and increased the level of IFN-γ produced by liver NK cells. Interestingly, NK cells from mice with persistent chronic infection expressed a higher level of TIGIT compared to self-healing mice. To look further into the mechanisms, more regulatory CD56bright and murine CD49a+ NK cells with higher TIGIT expression existed in livers of AE patients and mice infected with E. multilocularis, respectively. They coexpressed higher surface programmed death ligand 1 and secreted more IL-10, two strong inducers to mediate the functional exhaustion of NK cells. CONCLUSIONS Our results indicate that inhibitory receptor TIGIT is involved in NK cell exhaustion and immune escape from E. multilocularis infection.
Collapse
Affiliation(s)
- Chuanshan Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, Xinjiang Medical University, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Hui Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, Xinjiang Medical University, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Jing Li
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Xinling Hou
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, Xinjiang Medical University, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Linghui Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, Xinjiang Medical University, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wei Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, Xinjiang Medical University, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Yang Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, Xinjiang Medical University, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Dewei Li
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Liang Li
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, WHO Collaborating Centre for Prevention and Case Management of Echinococcosis, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhibin Zhao
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Liang Li
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Tuerganaili Aji
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Renyong Lin
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, WHO Collaborating Centre for Prevention and Case Management of Echinococcosis, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yingmei Shao
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Dominique A Vuitton
- WHO Collaborating Centre for the Prevention and Treatment of Human Echinococcosis, Department of Parasitology, University Bourgogne Franche-Comté (EA 3181) and University Hospital, Besançon, France
| | - Zhigang Tian
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Haoyu Sun
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, Xinjiang Medical University, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
27
|
Mohammed SH, Jabbr AS, Ibrahim NK. Impact of parasitic infection with Ascaris lumbricoides on pulmonary function tests in asthmatic and non-asthmatic children. Respir Med Case Rep 2021; 34:101552. [PMID: 34820258 PMCID: PMC8600146 DOI: 10.1016/j.rmcr.2021.101552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 11/16/2022] Open
Abstract
Background There is strong evidence for a causal relationship between helminthes infection and allergic disease like asthma due to the similarity in the way they respond. This study aimed to investigate the effects of Ascaris infection on pulmonary function tests (PFTs) to reveal the relationship between ascariasis and asthma in children. Patients and methods This a randomized-control study conducted in Basrah City, Iraq, in which four groups of a total of 490 children were enrolled: Group1 included 120 normal children; Group 2 included 135 asthmatic children; Group 3 who were 150 Ascaris infected children and group G4 included 85 asthmatic and Ascaris infected. PFTs, IgE level, differential blood count and parasitic examination were done for all groups. Results Both group2 and 4, which included asthmatic children showed a significant decrease in PFT (P > 0.05),while the PFT of parasitic infected group was not affected. There were no significant changes in WBC, eosinophils and IgE between asthmatic and parasitic infected groups. Conclusion Ascaris infection could induce the inflammatory immune response in children, but couldn't cause a significant effect on pulmonary function tests in these children; The impairment in PFT was due to asthma disease and not correlated to ascariasis.
Collapse
Affiliation(s)
- Suha Haithem Mohammed
- Department of Clinical Laboratory Sciences, College of Pharmacy, University of Basrah, Iraq
| | - Azza Sajid Jabbr
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Basrah, Iraq
| | | |
Collapse
|
28
|
Muñoz C, Guevara L, Escamilla MI, Regino R, Acevedo N, Escamilla-Arrieta JM. Risk Factors Associated With Health Care Utilization in Preschool Recurrent Wheezers in a Tropical Environment. FRONTIERS IN ALLERGY 2021; 2:761492. [PMID: 35387009 PMCID: PMC8974872 DOI: 10.3389/falgy.2021.761492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/30/2021] [Indexed: 11/17/2022] Open
Abstract
Introduction: The severity of wheezing episodes is related with the need for health services, but the factors associated with health care utilization in preschool recurrent wheezers in underdeveloped regions are unclear. Objective: To evaluate the factors associated with health care utilization in preschool recurrent wheezers in Cartagena, Colombia. Methods: One hundred twenty-seven recurrent wheezers (age 2-6 years old) who were admitted to the emergency room (ER) due to wheezing in a Pediatric reference hospital in Cartagena were included. Children were evaluated by means of questionnaires and classified according to the number of ER visits, need for hospitalization and history of intensive care unit (ICU) admission due to wheezing within the last year. Total serum IgE and specific IgE to house dust mite allergens (HDM) were measured by ImmunoCAP® and allergen sensitization was evaluated by skin prick tests (SPT). Results: The maternal report of nocturnal cough without fever in their children increased the risk to have ≥5 ER visits in the last year due to wheezing. The use of montelukast was negatively associated with hospitalization, while a history of pneumonia and lack of tap water, increased the risk of hospitalization due to wheezing. A history of bronchiolitis, family history of asthma, cohabiting with two or more siblings, passive exposure to smoke and lack of sewage facilities increased the risk of ICU admission due to wheezing. The presence of atopy evaluated by SPT reactivity, total IgE levels or specific IgE to HDM were not associated with health care utilization. We also found that seroprevalence of positive IgE (≥0.35 kU/L) was 27% to B. tropicalis and 20.3% to D. pteronyssinus but the prevalence of positive IgE sensitization to these allergens was below 2% and 8% when evaluated by SPT, respectively. Conclusions: Poverty indicators are associated with ICU admission in a group of preschool recurrent wheezers and should be considered as aggravating factors for wheezing. These factors must be systematically assessed in the medical approach in underdeveloped regions in the tropics. Nocturnal cough without fever is a symptom associated with frequent ER visits while atopy was not associated with health care utilization in preschool recurrent wheezers.
Collapse
Affiliation(s)
- César Muñoz
- Department of Pediatrics, University of Cartagena and Pediatric Pulmonology Service, Hospital Infantil Napoleón Franco Pareja–La Casa del Niño, Cartagena, Colombia
| | - Lissette Guevara
- Department of Pediatrics, University of Cartagena and Pediatric Pulmonology Service, Hospital Infantil Napoleón Franco Pareja–La Casa del Niño, Cartagena, Colombia
| | - María-Isabel Escamilla
- Department of Pediatrics and Sleep Medicine, Fundación Neumológica Colombiana, Bogotá, Colombia
| | - Ronald Regino
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | - Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | - Jose Miguel Escamilla-Arrieta
- Department of Pediatrics, University of Cartagena and Pediatric Pulmonology Service, Hospital Infantil Napoleón Franco Pareja–La Casa del Niño, Cartagena, Colombia
| |
Collapse
|
29
|
Leal-Silva T, Vieira-Santos F, Oliveira FMS, Padrão LDLS, Kraemer L, da Paixão Matias PH, de Almeida Lopes C, Loiola Ruas AC, de Azevedo IC, Nogueira DS, Rachid MA, Caliari MV, Castro Russo R, Fujiwara RT, Bueno LL. Detrimental role of IL-33/ST2 pathway sustaining a chronic eosinophil-dependent Th2 inflammatory response, tissue damage and parasite burden during Toxocara canis infection in mice. PLoS Negl Trop Dis 2021; 15:e0009639. [PMID: 34324507 PMCID: PMC8354467 DOI: 10.1371/journal.pntd.0009639] [Citation(s) in RCA: 4] [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: 03/15/2021] [Revised: 08/10/2021] [Accepted: 07/09/2021] [Indexed: 12/23/2022] Open
Abstract
Toxocariasis is a neglected disease that affects people around the world. Humans become infected by accidental ingestion of eggs containing Toxocara canis infective larvae, which upon reaching the intestine, hatch, penetrate the mucosa and migrate to various tissues such as liver, lungs and brain. Studies have indicated that Th2 response is the main immune defense mechanism against toxocariasis, however, there are still few studies related to this response, mainly the IL-33/ST2 pathway. Some studies have reported an increase in IL-33 during helminth infections, including T. canis. By binding to its ST2 receptor, IL-33 stimulating the Th2 polarized immune cell and cytokine responses. Thus, we aimed to investigate the role of the IL-33/ST2 pathway in the context of T. canis larval migration and the immunological and pathophysiological aspects of the infection in the liver, lungs and brain from Wild-Type (WT) BALB/c background and genetically deficient mice for the ST2 receptor (ST2-/-). The most important findings revealed that the IL-33/ST2 pathway is involved in eosinophilia, hepatic and cerebral parasitic burden, and induces the formation of granulomas related to tissue damage and pulmonary dysfunction. However, ST2-/- mice, the immune response was skewed to Th1/Th17 type than Th2, that enhanced the control of parasite burden related to IgG2a levels, tissue macrophages infiltration and reduced lung dysfunction. Collectively, our results demonstrate that the Th2 immune response triggered by IL-33/ST2 pathway mediates susceptibility to T. canis, related to parasitic burden, eosinophilia and granuloma formation in which consequently contributes to tissue inflammation and injury. Toxocariasis is a neglected disease caused by Toxocara canis, which has 19% worldwide seroprevalence, and is associated with socioeconomic, geographic and environmental factors. Humans become infected by accidental ingestion of T. canis eggs present in contaminated food, water or soil. After ingestion, the larvae hatch in the intestine and can reach various tissues such as liver, lung and brain. Helminth infections usually trigger a Th2 immune response in the host, by releasing cytokines such as IL-4, IL-5, IL-13 and IL-33. IL-33 is an alarmin that binds to the ST2 receptor, and some studies have observed an increase in this cytokine in toxocariasis, however there are no studies regarding the IL-33/ST2 role in this infection. Thus, we evaluated the influence of this pathway by analyzing immunological and pathophysiological aspects in T. canis-infected mice. Our results demonstrated that the IL-33/ST2 pathway is related to parasite burden on the liver and brain and also increases the number of eosinophils in the blood and tissues. In addition, it involved with the pulmonary immune response and granulomas with impact in lung function. In conclusion, the IL-33/ST2 pathway governs the host susceptibility to T. canis in mice.
Collapse
Affiliation(s)
- Thaís Leal-Silva
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Post-graduation Program in Health Sciences: Infectious Diseases and Tropical Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flaviane Vieira-Santos
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Marcus Silva Oliveira
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiza de Lima Silva Padrão
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Kraemer
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pablo Hemanoel da Paixão Matias
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila de Almeida Lopes
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Cristina Loiola Ruas
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabella Carvalho de Azevedo
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Denise Silva Nogueira
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Milene Alvarenga Rachid
- Laboratory of Protozooses, Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Vidigal Caliari
- Laboratory of Protozooses, Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- * E-mail:
| |
Collapse
|
30
|
Zakzuk J, Lozano A, Caraballo L. Allergological Importance of Invertebrate Glutathione Transferases in Tropical Environments. FRONTIERS IN ALLERGY 2021; 2:695262. [PMID: 35387058 PMCID: PMC8974725 DOI: 10.3389/falgy.2021.695262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/19/2021] [Indexed: 11/19/2022] Open
Abstract
Glutathione-S transferases (GSTs) are part of a ubiquitous family of dimeric proteins that participate in detoxification reactions. It has been demonstrated that various GSTs induce allergic reactions in humans: those originating from house dust mites (HDM), cockroaches, and helminths being the best characterized. Evaluation of their allergenic activity suggests that they have a clinical impact. GST allergens belong to different classes: mu (Blo t 8, Der p 8, Der f 8, and Tyr p 8), sigma (Bla g 5 and Asc s 13), or delta (Per a 5). Also, IgE-binding molecules belonging to the pi-class have been discovered in helminths, but they are not officially recognized as allergens. In this review, we describe some aspects of the biology of GST, analyze their allergenic activity, and explore the structural aspects and clinical impact of their cross-reactivity.
Collapse
|
31
|
Magalhães L, Nogueira DS, Gazzinelli-Guimarães PH, Oliveira FMS, Kraemer L, Gazzinelli-Guimarães AC, Vieira-Santos F, Fujiwara RT, Bueno LL. Immunological underpinnings of Ascaris infection, reinfection and co-infection and their associated co-morbidities. Parasitology 2021; 148:1-10. [PMID: 33843506 DOI: 10.1017/s0031182021000627] [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] [Indexed: 12/17/2022]
Abstract
Human ascariasis is the most common and prevalent neglected tropical disease and is estimated that ~819 million people are infected around the globe, accounting for 0.861 million years of disability-adjusted life years in 2017. Even with the existence of highly effective drugs, the constant presence of infective parasite eggs in the environment contribute to a high reinfection rate after treatment. Due to its high prevalence and broad geographic distribution Ascaris infection is associated with a variety of co-morbidities and co-infections. Here, we provide data from both experimental models and humans studies that illustrate how complex is the interaction of Ascaris with the host immune system, especially, in the context of reinfections, co-infections and associated co-morbidities.
Collapse
Affiliation(s)
- Luisa Magalhães
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Denise S Nogueira
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro H Gazzinelli-Guimarães
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Fabricio M S Oliveira
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lucas Kraemer
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Flaviane Vieira-Santos
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo T Fujiwara
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lilian L Bueno
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
32
|
Johnston SL, Goldblatt DL, Evans SE, Tuvim MJ, Dickey BF. Airway Epithelial Innate Immunity. Front Physiol 2021; 12:749077. [PMID: 34899381 PMCID: PMC8662554 DOI: 10.3389/fphys.2021.749077] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/28/2021] [Indexed: 01/21/2023] Open
Abstract
Besides providing an essential protective barrier, airway epithelial cells directly sense pathogens and respond defensively. This is a frontline component of the innate immune system with specificity for different pathogen classes. It occurs in the context of numerous interactions with leukocytes, but here we focus on intrinsic epithelial mechanisms. Type 1 immune responses are directed primarily at intracellular pathogens, particularly viruses. Prominent stimuli include microbial nucleic acids and interferons released from neighboring epithelial cells. Epithelial responses revolve around changes in the expression of interferon-sensitive genes (ISGs) that interfere with viral replication, as well as the further induction of interferons that signal in autocrine and paracrine manners. Type 2 immune responses are directed primarily at helminths and fungi. Prominent pathogen stimuli include proteases and chitin, and important responses include mucin hypersecretion and chitinase release. Type 3 immune responses are directed primarily at extracellular microbial pathogens, including bacteria and fungi, as well as viruses during their extracellular phase of infection. Prominent microbial stimuli include bacterial wall components, such as lipopeptides and endotoxin, as well as microbial nucleic acids. Key responses are the release of reactive oxygen species (ROS) and antimicrobial peptides (AMPs). For all three types of response, paracrine signaling to neighboring epithelial cells induces resistance to infection over a wide field. Often, the epithelial effector molecules themselves also have signaling properties, in addition to the release of inflammatory cytokines that boost local innate immunity. Together, these epithelial mechanisms provide a powerful first line of pathogen defense, recruit leukocytes, and instruct adaptive immune responses.
Collapse
Affiliation(s)
- Sebastian L Johnston
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - David L Goldblatt
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,University of Texas Rio Grande School of Medicine, Edinburg, TX, United States.,Howard Hughes Medical Institute, Chevy Chase, MD, United States
| | - Scott E Evans
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael J Tuvim
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Burton F Dickey
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|