1
|
Kumar R, Meena AS, Swarnkar CP, Misra SS, Kumar A. Absolute eosinophil count and expression analyses of cytokine genes in Haemonchus contortus resistant Malpura sheep: Role of TH1/TH2 cytokine dichotomy. Vet Parasitol Reg Stud Reports 2025; 57:101194. [PMID: 39855879 DOI: 10.1016/j.vprsr.2025.101194] [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: 07/08/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025]
Abstract
A strong Th2-type immune response against Haemonchus contortus infection in genetically resistant sheep provide immunity. The objective of present study was to correlate mean faecal egg counts (FECs), absolute eosinophil counts and Th1/Th2 gene expression in resistant (R) and susceptible (S) Malpura sheep. In spite of no anthelmintic treatment in R line, on majority of the months, mean FECs remained significantly (P < 0.05) lower compared to S line where anthelmintic treatment was given in September every year. R sheep possess a smaller number of blood eosinophils in the majority of months as compared with S sheep and a positive correlation has been observed between absolute eosinophil counts and mean FEC. In the condition, when FEC remain similar in both the lines (R and S), significantly (P < 0.05) up-regulated expression of IL-5 was observed in R sheep compared with S sheep however when FEC is hugely enhanced; IL-2, IL-6, IL-12 and IL-13 gene expression was significantly (P < 0.05) up-regulated in R sheep as compared to S sheep which is dependent on season and parasite load.
Collapse
Affiliation(s)
- Rajiv Kumar
- Animal Genetics and Breeding Division, ICAR-Central Sheep and Wool Research Institute, Avikanagar 304 501, Rajasthan, India.
| | - Amar S Meena
- Animal Genetics and Breeding Division, ICAR-Central Sheep and Wool Research Institute, Avikanagar 304 501, Rajasthan, India
| | - Chander P Swarnkar
- Animal Health Division, ICAR-Central Sheep and Wool Research Institute, Avikanagar 304 501, Rajasthan
| | - Siddharth S Misra
- Animal Genetics and Breeding Division, ICAR-Central Sheep and Wool Research Institute, Avikanagar 304 501, Rajasthan, India
| | - Arun Kumar
- Animal Genetics and Breeding Division, ICAR-Central Sheep and Wool Research Institute, Avikanagar 304 501, Rajasthan, India
| |
Collapse
|
2
|
Brown GP, Shine R, Rollins LA. Does a biological invasion modify host immune responses to parasite infection? ROYAL SOCIETY OPEN SCIENCE 2025; 12:240669. [PMID: 39816740 PMCID: PMC11732422 DOI: 10.1098/rsos.240669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/13/2024] [Accepted: 12/04/2024] [Indexed: 01/18/2025]
Abstract
Biological invasions can disrupt the close and longstanding coevolved relationships between host and parasites. At the same time, the shifting selective forces acting on demography during invasion can result in rapid evolution of traits in both host and parasite. Hosts at the invasion front may reduce investment into costly immune defences and redistribute those resources to other fitness-enhancing traits. Parasites at the invasion front may have reduced pathogenicity because traits that negatively impact host dispersal are left behind in the expanding range. The host's immune system is its primary arsenal in the coevolutionary 'arms race' with parasites. To assess the effects of invasion history on immune responses to parasite infection, we conducted a cross-infection experiment which paired common-garden reared cane toads and lungworm parasites originating from various sites in their invaded range across northern Australia. Infected toads had larger spleens and higher concentrations of eosinophils than did uninfected toads. Infected toads also exhibited lower bacteria-killing ability, perhaps reflecting a trade-off of resources towards defences that are more specifically anthelminthic. The impact of infection intensity on multiple immune measures differed among toads and parasites from different parts of the invasion trajectory, supporting the hypothesis that invasion has disrupted patterns of local adaptation.
Collapse
Affiliation(s)
- Gregory P. Brown
- School of Natural Sciences, Macquarie University, Sydney, NSW2109, Australia
| | - Richard Shine
- School of Natural Sciences, Macquarie University, Sydney, NSW2109, Australia
| | - Lee A. Rollins
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, NSW2052, Australia
| |
Collapse
|
3
|
Zhu Y, Chen X, Zheng H, Ma Q, Chen K, Li H. Anti-Inflammatory Effects of Helminth-Derived Products: Potential Applications and Challenges in Diabetes Mellitus Management. J Inflamm Res 2024; 17:11789-11812. [PMID: 39749005 PMCID: PMC11694023 DOI: 10.2147/jir.s493374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025] Open
Abstract
The global rise in diabetes mellitus (DM), particularly type 2 diabetes (T2D), has become a major public health challenge. According to the "hygiene hypothesis", helminth infections may offer therapeutic benefits for DM. These infections are known to modulate immune responses, reduce inflammation, and improve insulin sensitivity. However, they also carry risks, such as malnutrition, anemia, and intestinal obstruction. Importantly, helminth excretory/secretory products, which include small molecules and proteins, have shown therapeutic potential in treating various inflammatory diseases with minimal side effects. This review explores the anti-inflammatory properties of helminth derivatives and their potential to alleviate chronic inflammation in both type 1 diabetes and T2D, highlighting their promise as future drug candidates. Additionally, it discusses the possible applications of these derivatives in DM management and the challenges involved in translating these findings into clinical practice.
Collapse
Affiliation(s)
- Yunhuan Zhu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xintong Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hezheng Zheng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Qiman Ma
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hongyu Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
- Ocean College, Beibu Gulf University, Qinzhou, Guangxi, People’s Republic of China
| |
Collapse
|
4
|
Gazzinelli-Guimaraes PH, Jones SM, Voehringer D, Mayer-Barber KD, Samarasinghe AE. Eosinophils as modulators of host defense during parasitic, fungal, bacterial, and viral infections. J Leukoc Biol 2024; 116:1301-1323. [PMID: 39136237 DOI: 10.1093/jleuko/qiae173] [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: 02/12/2024] [Revised: 06/25/2024] [Indexed: 11/28/2024] Open
Abstract
Eosinophils, traditionally associated as central innate effector cells with type 2 immunity during allergic and helminth parasitic diseases, have recently been revealed to have important roles in tissue homeostasis as well as host defense in a broader variety of infectious diseases. In a dedicated session at the 2023 biennial conference of the International Eosinophil Society titled "Eosinophils in Host Defense," the multifaceted roles eosinophils play against diverse pathogens, ranging from parasites to fungi, bacteria, and viruses, were presented. In this review, the session speakers offer a comprehensive summary of recent discoveries across pathogen classes, positioning eosinophils as pivotal leukocytes in both host defense and pathology. By unraveling the intricacies of eosinophil engagement in host resistance, this exploration may provide valuable insights not only to understand specific underpinnings of eosinophil functions related to each class of pathogens but also to develop novel therapeutics effective against a broad spectrum of infectious diseases.
Collapse
Affiliation(s)
- Pedro H Gazzinelli-Guimaraes
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington School of Medicine and Health Sciences, 2300 I Street NW, Washington, DC 20037, United States
| | - Shelby M Jones
- Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, United States
| | - David Voehringer
- Department of Infection Biology, Universitätsklinikum Erlangen, Wasserturmstrasse 3-5, 91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 33 North Drive, Bethesda, MD 20892, United States
| | - Amali E Samarasinghe
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Children's Foundation Research Institute, 50 N Dunlap Street, Memphis, TN 38103, United States
| |
Collapse
|
5
|
Liu Y, Zhao S, Du S, Zhang Y, Yu Y, Zhan B, Hao J, Jia Z, Huang J, Guo Y, Zhang L, Zhu X, Cheng Y. PD-1 deficiency impairs eosinophil recruitment to tissue during Trichinella spiralis infection. Cell Rep 2024; 43:114861. [PMID: 39418164 DOI: 10.1016/j.celrep.2024.114861] [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: 04/10/2024] [Revised: 09/04/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Blockade of programmed cell death 1 (PD-1) is considered a promising strategy for controlling pathogen infection by enhancing host immune cell function. Eosinophils, which play a crucial role in type 2 immune responses, are essential components of the host defense against helminth infection. Here, we investigate the role of PD-1 in eosinophilia during Trichinella spiralis infection in mice. PD-1-deficient (PD-1-/-) mice exhibit delayed expulsion of adult worms and increased muscle larva burdens compared to wild-type mice following infection. Additionally, PD-1-/- mice display impaired recruitment of eosinophils to parasite-invaded tissues, attributed to decreased upregulation of adhesion molecules on both eosinophils and vascular endothelium after infection. The compromised Th2 cytokine response further contributes to impaired adhesion interactions, affecting eosinophil migration and cytotoxicity against larvae in vitro within T. spiralis-infected PD-1-/- mice. Our findings demonstrate a positive role for PD-1 in the recruitment of eosinophils, suggesting its involvement in host defense against helminth infection.
Collapse
Affiliation(s)
- Yiqi Liu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Simeng Zhao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Suqin Du
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yao Zhang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yan Yu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Bin Zhan
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Junfeng Hao
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Zhihui Jia
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jingjing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yuteng Guo
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Lishuang Zhang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| | - Yuli Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
6
|
Packi K, Fugiel W, Gołąbek V, Rudek A, Śliwińska A. Eosinophilic Pneumonia Triggered by Toxocara canis in a Patient with Primary Ciliary Dyskinesia: A Clinical Case Report. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1874. [PMID: 39597059 PMCID: PMC11596152 DOI: 10.3390/medicina60111874] [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: 09/30/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
Primary ciliary dyskinesia (PCD) is a rare genetic disorder that affects the structure and function of cilia, primarily impacting the respiratory system. Kartagener syndrome, a subset of PCD, is characterized by situs inversus, bronchiectasis, and chronic sinusitis. Patients with PCD are prone to recurrent respiratory infections due to impaired ciliary function, which hinders effective mucus clearance and promotes pathogen colonization. This case report describes a 24-year-old woman with congenital Kartagener syndrome who developed eosinophilic pneumonia caused by Toxocara canis, a rare parasitic infection that less commonly affects the lungs. Despite initial treatment for a presumed bacterial infection, the patient's symptoms persisted. Further diagnostics revealed elevated eosinophil counts, total IgE, and the presence of Toxocara canis antibodies. The patient was treated with albendazole, resulting in significant symptom improvement and a reduction in inflammatory markers. This case underscores the diagnostic challenges in treating PCD patients, where atypical infections must be considered, particularly when standard treatments prove ineffective. The complexity of the patient's condition required interdisciplinary management, integrating parasitological, immunological, and respiratory expertise to ensure appropriate treatment. The case highlights the need for further research into the interactions between congenital respiratory disorders such as Kartagener syndrome and parasitic infections. It also emphasizes the importance of a comprehensive diagnostic approach in managing rare genetic diseases complicated by opportunistic infections. Early detection of parasitic infections in PCD patients is crucial to preventing severe complications, and this case reinforces the necessity of considering parasitic causes in atypical pneumonia cases.
Collapse
Affiliation(s)
- Kacper Packi
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 92-213 Lodz, Poland;
- AllerGen Center of Personalized Medicine, 97-300 Piotrkow Trybunalski, Poland; (W.F.); (A.R.)
- Wladyslaw Bieganski Collegium Medicum, Jan Dlugosz University in Czestochowa, 42-200 Czestochowa, Poland
| | - Wanda Fugiel
- AllerGen Center of Personalized Medicine, 97-300 Piotrkow Trybunalski, Poland; (W.F.); (A.R.)
| | - Violetta Gołąbek
- AllerGen Center of Personalized Medicine, 97-300 Piotrkow Trybunalski, Poland; (W.F.); (A.R.)
| | - Alicja Rudek
- AllerGen Center of Personalized Medicine, 97-300 Piotrkow Trybunalski, Poland; (W.F.); (A.R.)
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 92-213 Lodz, Poland;
| |
Collapse
|
7
|
Rashed HAEH, Albogami B, Alkhaldi AAM, Abuzinadah NY, Abuzahrah SS, Al-Salmi FA, Fayad E, Fouad RM, Fikry ME, ElSaey AAA, Abu Almaaty AH. Effect of vitamin C injections on exercise muscular performance and biochemical parameters in Trichinella spiralis-infected mice. PeerJ 2024; 12:e18381. [PMID: 39553708 PMCID: PMC11568815 DOI: 10.7717/peerj.18381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/01/2024] [Indexed: 11/19/2024] Open
Abstract
Background Trichinella spiralis is a worldwide intestinal nematode that can parasitize the striated muscles of its hosts at the larval stage. This study aims to evaluate potential of vitamin C for treating trichinellosis-related pathological problems in the infected muscles of mice. Materials and Methods Thirty CD1 male Albino mice were divided into three groups (10 mice per group). Negative and positive control groups (0.9% NaCl) and the infected vitamin C group (10 mg/kg body weight). Two weeks post-infection, each group was intraperitoneally injected daily for two weeks with Vitamin C or saline. The performance of the muscles was assessed both before and after the treatment. After dissection, constant parts of striated muscles were removed for further assays. The scoring of the histological changes of infected muscles was carried out. In addition to muscle malondialdehyde levels, superoxide dismutase and catalase activities were measured for the oxidative and antioxidant states. Creatine kinase and aspartate aminotransferase were also measured in tissues to reflect the degree of muscular damage. Results Vitamin C enhances the weakness of the muscular performance resulting from the infection. Vitamin C was able to repair some of the histological lesions that resulted from the infection. Trichinellosis caused severe changes in the biochemical markers in positive control animals. Muscle damage biomarkers and, besides, oxidative and antioxidant conditions were greatly ameliorated in infected vitamin C animals. Summing up, vitamin C can be used as a complementary drug due to its efficiency in improving pathogenesis following a trichinellosis infection. The supplement also must be tested in the intestinal stage of infection after showing promising results in the muscular stage.
Collapse
Affiliation(s)
| | - Bander Albogami
- Department of Biology, College of Sciences, Taif University, Taif, Saudi Arabia
| | | | - Najlaa Y. Abuzinadah
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Samah S. Abuzahrah
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Fawziah A. Al-Salmi
- Department of Biology, College of Sciences, Taif University, Taif, Saudi Arabia
| | - Eman Fayad
- Department of Biotechnology, College of Sciences, Taif University, Taif, Saudi Arabia
| | - Rewan Mohamed Fouad
- Zoology Department, Faculty of Science, Port Said University, Port Said, Egypt
| | - Manar Elsayed Fikry
- Zoology Department, Faculty of Science, Port Said University, Port Said, Egypt
| | | | | |
Collapse
|
8
|
Parrish KM, Gestal MC. Eosinophils as drivers of bacterial immunomodulation and persistence. Infect Immun 2024; 92:e0017524. [PMID: 39007622 PMCID: PMC11385729 DOI: 10.1128/iai.00175-24] [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: 07/16/2024] Open
Abstract
Traditionally, eosinophils have been linked to parasitic infections and pathological disease states. However, emerging literature has unveiled a more nuanced and intricate role for these cells, demonstrating their key functions in maintaining mucosal homeostasis. Eosinophils exhibit diverse phenotypes and exert multifaceted effects during infections, ranging from promoting pathogen persistence to triggering allergic reactions. Our investigations primarily focus on Bordetella spp., with particular emphasis on Bordetella bronchiseptica, a natural murine pathogen that induces diseases in mice akin to pertussis in humans. Recent findings from our published work have unveiled a striking interaction between B. bronchiseptica and eosinophils, facilitated by the btrS-mediated mechanism. This interaction serves to enhance pathogen persistence while concurrently delaying adaptive immune responses. Notably, this role of eosinophils is only noted in the absence of a functional btrS signaling pathway, indicating that wild-type B. bronchiseptica, and possibly other Bordetella spp., possess such adeptness in manipulating eosinophils that the true function of these cells remains obscured during infection. In this review, we present the mounting evidence pointing toward eosinophils as targets of bacterial exploitation, facilitating pathogen persistence and fostering chronic infections in diverse mucosal sites, including the lungs, gut, and skin. We underscore the pivotal role of the master regulator of Bordetella pathogenesis, the sigma factor BtrS, in orchestrating eosinophil-dependent immunomodulation within the context of pulmonary infection. These putative convergent strategies of targeting eosinophils offer promising avenues for the development of novel therapeutics targeting respiratory and other mucosal pathogens.
Collapse
Affiliation(s)
- Katelyn M. Parrish
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, Louisiana, USA
| | - Monica C. Gestal
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
9
|
Mertelsmann AM, Bowers SF, Wright D, Maganga JK, Mazigo HD, Ndhlovu LC, Changalucha JM, Downs JA. Effects of Schistosoma haematobium infection and treatment on the systemic and mucosal immune phenotype, gene expression and microbiome: A systematic review. PLoS Negl Trop Dis 2024; 18:e0012456. [PMID: 39250522 PMCID: PMC11412685 DOI: 10.1371/journal.pntd.0012456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 09/19/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Urogenital schistosomiasis caused by Schistosoma haematobium affects approximately 110 million people globally, with the majority of cases in low- and middle-income countries. Schistosome infections have been shown to impact the host immune system, gene expression, and microbiome composition. Studies have demonstrated variations in pathology between schistosome subspecies. In the case of S. haematobium, infection has been associated with HIV acquisition and bladder cancer. However, the underlying pathophysiology has been understudied compared to other schistosome species. This systematic review comprehensively investigates and assimilates the effects of S. haematobium infection on systemic and local host mucosal immunity, cellular gene expression and microbiome. METHODS We conducted a systematic review assessing the reported effects of S. haematobium infections and anthelmintic treatment on the immune system, gene expression and microbiome in humans and animal models. This review followed PRISMA guidelines and was registered prospectively in PROSPERO (CRD42022372607). Randomized clinical trials, cohort, cross-sectional, case-control, experimental ex vivo, and animal studies were included. Two reviewers performed screening independently. RESULTS We screened 3,177 studies and included 94. S. haematobium was reported to lead to: (i) a mixed immune response with a predominant type 2 immune phenotype, increased T and B regulatory cells, and select pro-inflammatory cytokines; (ii) distinct molecular alterations that would compromise epithelial integrity, such as increased metalloproteinase expression, and promote immunological changes and cellular transformation, specifically upregulation of genes p53 and Bcl-2; and (iii) microbiome dysbiosis in the urinary, intestinal, and genital tracts. CONCLUSION S. haematobium induces distinct alterations in the host's immune system, molecular profile, and microbiome. This leads to a diverse range of inflammatory and anti-inflammatory responses and impaired integrity of the local mucosal epithelial barrier, elevating the risks of secondary infections. Further, S. haematobium promotes cellular transformation with oncogenic potential and disrupts the microbiome, further influencing the immune system and genetic makeup. Understanding the pathophysiology of these interactions can improve outcomes for the sequelae of this devastating parasitic infection.
Collapse
Affiliation(s)
- Anna M Mertelsmann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
| | - Sheridan F Bowers
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
| | - Drew Wright
- Samuel J. Wood Library & C.V. Starr Biomedical Information Center, Weill Cornell Medical College, New York, New York, United States of America
| | - Jane K Maganga
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
| | - Humphrey D Mazigo
- Department of Parasitology and Entomology, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Lishomwa C Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - John M Changalucha
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
| | - Jennifer A Downs
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
- Weill Bugando School of Medicine, Mwanza, Tanzania
| |
Collapse
|
10
|
Bessell E, Finlay RE, James LK, Ludewig B, Harris NL, Krebs P, Hepworth MR, Dubey LK. Stromal cell and B cell dialogue potentiates IL-33-enriched lymphoid niches to support eosinophil recruitment and function during type 2 immunity. Cell Rep 2024; 43:114620. [PMID: 39141517 DOI: 10.1016/j.celrep.2024.114620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/27/2024] [Accepted: 07/25/2024] [Indexed: 08/16/2024] Open
Abstract
Eosinophils are involved in host protection against multicellular organisms. However, their recruitment to the mesenteric lymph node (mLN) during type 2 immunity is understudied. Our results demonstrate that eosinophil association with lymphoid stromal niches constructed by fibroblastic reticular cells (FRCs) and lymphatic endothelial cells is diminished in mice selectively lacking interleukin (IL)-4Rα or lymphotoxin-β (LTβ) expression on B cells. Furthermore, eosinophil survival, activation, and enhanced Il1rl1 receptor expression are driven by stromal cell and B cell dialogue. The ligation of lymphotoxin-β receptor (LTβR) on FRCs improves eosinophil survival and significantly augments IL-33 expression and eosinophil homing to the mLN, thus confirming the significance of lymphotoxin signaling for granulocyte recruitment. Eosinophil-deficient ΔdblGATA-1 mice show diminished mLN expansion, reduced interfollicular region (IFR) alarmin expression, and delayed helminth clearance, elucidating their importance in type 2 immunity. These findings provide insight into dialogue between stromal cells and B cells, which govern mLN eosinophilia, and the relevance of these mechanisms during type 2 immunity.
Collapse
Affiliation(s)
- Emily Bessell
- William Harvey Research Institute (WHRI), Barts & The London School of Medicine & Dentistry, Queen Mary University of London (QMUL), London, UK; Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland; Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Rachel E Finlay
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Louisa K James
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, UK
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Nicola L Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Philippe Krebs
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Matthew R Hepworth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Lalit Kumar Dubey
- William Harvey Research Institute (WHRI), Barts & The London School of Medicine & Dentistry, Queen Mary University of London (QMUL), London, UK; Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
11
|
Wang H, Barry K, Zaini A, Coakley G, Moyat M, Daunt CP, Wickramasinghe LC, Azzoni R, Chatzis R, Yumnam B, Camberis M, Le Gros G, Perdijk O, Foong JPP, Bornstein JC, Marsland BJ, Harris NL. Helminth infection driven gastrointestinal hypermotility is independent of eosinophils and mediated by alterations in smooth muscle instead of enteric neurons. PLoS Pathog 2024; 20:e1011766. [PMID: 39141685 PMCID: PMC11346963 DOI: 10.1371/journal.ppat.1011766] [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: 10/25/2023] [Revised: 08/26/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
Intestinal helminth infection triggers a type 2 immune response that promotes a 'weep-and sweep' response characterised by increased mucus secretion and intestinal hypermotility, which function to dislodge the worm from its intestinal habitat. Recent studies have discovered that several other pathogens cause intestinal dysmotility through major alterations to the immune and enteric nervous systems (ENS), and their interactions, within the gastrointestinal tract. However, the involvement of these systems has not been investigated for helminth infections. Eosinophils represent a key cell type recruited by the type 2 immune response and alter intestinal motility under steady-state conditions. Our study aimed to investigate whether altered intestinal motility driven by the murine hookworm, Nippostrongylus brasiliensis, infection involves eosinophils and how the ENS and smooth muscles of the gut are impacted. Eosinophil deficiency did not influence helminth-induced intestinal hypermotility and hypermotility did not involve gross structural or functional changes to the ENS. Hypermotility was instead associated with a dramatic increase in smooth muscle thickness and contractility, an observation that extended to another rodent nematode, Heligmosomoides polygyrus. In summary our data indicate that, in contrast to other pathogens, helminth-induced intestinal hypermotility is driven by largely by myogenic, rather than neurogenic, alterations with such changes occurring independently of eosinophils. (<300 words).
Collapse
Affiliation(s)
- Haozhe Wang
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Kristian Barry
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Aidil Zaini
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Gillian Coakley
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Mati Moyat
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Carmel P. Daunt
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Lakshanie C. Wickramasinghe
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Rossana Azzoni
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Roxanne Chatzis
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Bibek Yumnam
- The Malaghan Institute of Medical Research, Victoria University, Wellington, New Zealand
| | - Mali Camberis
- The Malaghan Institute of Medical Research, Victoria University, Wellington, New Zealand
| | - Graham Le Gros
- The Malaghan Institute of Medical Research, Victoria University, Wellington, New Zealand
| | - Olaf Perdijk
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Jaime P. P. Foong
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Joel C. Bornstein
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Benjamin J. Marsland
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Nicola L. Harris
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Chang LA, Schotsaert M. Ally, adversary, or arbitrator? The context-dependent role of eosinophils in vaccination for respiratory viruses and subsequent breakthrough infections. J Leukoc Biol 2024; 116:224-243. [PMID: 38289826 PMCID: PMC11288382 DOI: 10.1093/jleuko/qiae010] [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: 10/29/2023] [Revised: 12/12/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Eosinophils are a critical type of immune cell and central players in type 2 immunity. Existing literature suggests that eosinophils also can play a role in host antiviral responses, typically type 1 immune events, against multiple respiratory viruses, both directly through release of antiviral mediators and indirectly through activation of other effector cell types. One way to prime host immune responses toward effective antiviral responses is through vaccination, where typically a type 1-skewed immunity is desirable in the context of intracellular pathogens like respiratory viruses. In the realm of breakthrough respiratory viral infection in vaccinated hosts, an event in which virus can still establish productive infection despite preexisting immunity, eosinophils are most prominently known for their link to vaccine-associated enhanced respiratory disease upon natural respiratory syncytial virus infection. This was observed in a pediatric cohort during the 1960s following vaccination with formalin-inactivated respiratory syncytial virus. More recent research has unveiled additional roles of the eosinophil in respiratory viral infection and breakthrough infection. The specific contribution of eosinophils to the quality of vaccine responses, vaccine efficacy, and antiviral responses to infection in vaccinated hosts remains largely unexplored, especially regarding their potential roles in protection. On the basis of current findings, we will speculate upon the suggested function of eosinophils and consider the many potential ways by which eosinophils may exert protective and pathological effects in breakthrough infections. We will also discuss how to balance vaccine efficacy with eosinophil-related risks, as well as the use of eosinophils and their products as potential biomarkers of vaccine efficacy or adverse events.
Collapse
Affiliation(s)
- Lauren A Chang
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1630, New York, NY 10029, United States
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States
| |
Collapse
|
13
|
Ackerman SJ, Stacy NI. Considerations on the evolutionary biology and functions of eosinophils: what the "haeckel"? J Leukoc Biol 2024; 116:247-259. [PMID: 38736141 PMCID: PMC11288384 DOI: 10.1093/jleuko/qiae109] [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: 02/01/2024] [Revised: 03/27/2024] [Accepted: 04/17/2024] [Indexed: 05/14/2024] Open
Abstract
The origins and evolution of the eosinophilic leukocyte have received only scattered attention since Paul Ehrlich first named this granulocyte. Studies suggest that myeloperoxidase, expressed by granulocytes, and eosinophil peroxidase diverged some 60 to 70 million years ago, but invertebrate to vertebrate evolution of the eosinophil lineage is unknown. Vertebrate eosinophils have been characterized extensively in representative species at light microscopic, ultrastructural, genetic, and biochemical levels. Understanding of eosinophil function continues to expand and includes to date regulation of "Local Immunity And/Or Remodeling/Repair" (the so-called LIAR hypothesis), modulation of innate and adaptive immune responses, maintenance of tissue and metabolic homeostasis, and, under pathologic conditions, inducers of tissue damage, repair, remodeling, and fibrosis. This contrasts with their classically considered primary roles in host defense against parasites and other pathogens, as well as involvement in T-helper 2 inflammatory and immune responses. The eosinophils' early appearance during evolution and continued retention within the innate immune system across taxa illustrate their importance during evolutionary biology. However, successful pregnancies in eosinophil-depleted humans/primates treated with biologics, host immune responses to parasites in eosinophil-deficient mice, and the absence of significant developmental or functional abnormalities in eosinophil-deficient mouse strains under laboratory conditions raise questions of the continuing selective advantages of the eosinophil lineage in mammals and humans. The objectives of this review are to provide an overview on evolutionary origins of eosinophils across the animal kingdom, discuss some of their main functions in the context of potential evolutionary relevance, and highlight the need for further research on eosinophil functions and functional evolution.
Collapse
Affiliation(s)
- Steven J Ackerman
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, MBRB2074, MC669, 900 S. Ashland Ave, Chicago, IL 60607, United States
| | - Nicole I Stacy
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2015 SW 16th Ave, Gainesville, FL 32610, United States
| |
Collapse
|
14
|
Rajeev S, Li S, Leon-Coria A, Wang A, Kraemer L, Wang SJ, Boim A, Flannigan K, Shute A, Baggio CH, Callejas BE, MacNaughton WK, Finney CAM, McKay DM. Enteric tuft cells coordinate timely expulsion of the tapeworm Hymenolepis diminuta from the murine host by coordinating local but not systemic immunity. PLoS Pathog 2024; 20:e1012381. [PMID: 39083533 PMCID: PMC11290655 DOI: 10.1371/journal.ppat.1012381] [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: 09/06/2023] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
Recognizing that enteric tuft cells can signal the presence of nematode parasites, we investigated whether tuft cells are required for the expulsion of the cestode, Hymenolepis diminuta, from the non-permissive mouse host, and in concomitant anti-helminthic responses. BALB/c and C57BL/6 mice infected with H. diminuta expelled the worms by 11 days post-infection (dpi) and displayed DCLK1+ (doublecortin-like kinase 1) tuft cell hyperplasia in the small intestine (not the colon) at 11 dpi. This tuft cell hyperplasia was dependent on IL-4Rα signalling and adaptive immunity, but not the microbiota. Expulsion of H. diminuta was slowed until at least 14 dpi, but not negated, in tuft cell-deficient Pou2f3-/- mice and was accompanied by delayed goblet cell hyperplasia and slowed small bowel transit. Worm antigen and mitogen evoked production of IL-4 and IL-10 by splenocytes from wild-type and Pou2f3-/- mice was not appreciably different, suggesting similar systemic immune reactivity to infection with H. diminuta. Wild-type and Pou2f3-/- mice infected with H. diminuta displayed partial protection against subsequent infection with the nematode Heligmosomoides bakeri. We speculate that, with respect to H. diminuta, enteric tuft cells are important for local immune events driving the rapidity of H. diminuta expulsion but are not critical in initiating or sustaining systemic Th2 responses that provide concomitant immunity against secondary infection with H. bakeri.
Collapse
Affiliation(s)
- Sruthi Rajeev
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - ShuHua Li
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Aralia Leon-Coria
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
- Department of Biology, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Arthur Wang
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Lucas Kraemer
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Susan Joanne Wang
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Annaliese Boim
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Kyle Flannigan
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Adam Shute
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Cristiane H. Baggio
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Blanca E. Callejas
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K. MacNaughton
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Constance A. M. Finney
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
- Department of Biology, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Derek M. McKay
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
15
|
Lopez-Perez D, Prados-Lopez B, Galvez J, Leon J, Carazo A. Eosinophils in Colorectal Cancer: Emerging Insights into Anti-Tumoral Mechanisms and Clinical Implications. Int J Mol Sci 2024; 25:6098. [PMID: 38892286 PMCID: PMC11172675 DOI: 10.3390/ijms25116098] [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: 04/19/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Eosinophils are myeloid effector cells whose main homing is the gastrointestinal tract. There, they take part in type I and type II immune responses. They also contribute to other non-immunological homeostatic functions like mucus production, tissue regeneration, and angiogenesis. In colorectal cancer (CRC), eosinophils locate in the center of the tumor and in the front of invasion and play an anti-tumoral role. They directly kill tumor cells by releasing cytotoxic compounds and eosinophil extracellular traps or indirectly by activating other immune cells via cytokines. As CRC progresses, the number of infiltrating eosinophils decreases. Although this phenomenon is not fully understood, it is known that some changes in the microenvironmental milieu and microbiome can affect eosinophil infiltration. Importantly, a high number of intratumoral eosinophils is a favorable prognostic factor independent from the tumor stage. Moreover, after immunotherapy, responding patients usually display eosinophilia, so eosinophils could be a good biomarker candidate to monitor treatment outcomes. Finally, even though eosinophils seem to play an interesting anti-tumoral role in CRC, much more research is needed to fully understand their interactions in the CRC microenvironment. This review explores the multifaceted roles of eosinophils in colorectal cancer, highlighting their anti-tumoral effects, prognostic significance, and potential as a biomarker for treatment outcomes.
Collapse
Affiliation(s)
- David Lopez-Perez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, 18012 Granada, Spain
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | - Belen Prados-Lopez
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | - Julio Galvez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, 18012 Granada, Spain
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Centro de Investigación Biomédica en Red para Enfermedades Hepáticas y Digestivas (CIBER-EHD), Center for Biomedical Research, University of Granada, 18012 Granada, Spain
| | - Josefa Leon
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
| | - Angel Carazo
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Unidad de Gestión de Microbiología, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
| |
Collapse
|
16
|
Abdel-Hakeem SS, Abdel-Samiee MAZ, Youssef MSE, Abd-Elsadek SH, Abd-Elrahman SM, Abdel-Hakeem SS. Nanocurcumin: A Promising Therapeutic Candidate for Experimental Trichinellosis. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2024; 30:368-381. [PMID: 38323506 DOI: 10.1093/micmic/ozae007] [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: 08/19/2023] [Revised: 12/18/2023] [Accepted: 01/18/2024] [Indexed: 02/08/2024]
Abstract
In our pursuit of an alternative drug against Trichinella spiralis, we assessed the effectiveness of nanocurcumin in alleviating pathogenesis, parasitological factors, MMP-9 levels, and its expression in the enteral and parenteral phases of infection. The nanocurcumin particles, with a spherical shape and a size of 100 ± 20 nm, were used in the study. Eighty mice were divided into four groups: the control group, the untreated infected group, the nanocurcumin-treated group, and the albendazole-treated group. The nanocurcumin-treated group exhibited a statistically significant increase in the percentage of lymphocytes, along with a reduction in neutrophils, monocytes, and eosinophils compared to the untreated, infected group. Both the nanocurcumin (87.2 and 97.3%) and the albendazole-treated groups (99.8 and 98.2%) showed a significant reduction in the mean number of intestinal worms and encysted larvae, respectively. The treated groups exhibited normal intestinal villi, suppression of the inflammatory process, and fewer instances of degenerated larvae in the diaphragm and muscle compared to the untreated, infected group. Immunohistochemistry and ELISA analyses revealed a significant downregulation of MMP-9 levels in the intestines and muscles of the treated groups. Our data demonstrate that nanocurcumin contains highly versatile molecules capable of modulating biological activity against inflammation and its pathway markers.
Collapse
Affiliation(s)
- Sara Salah Abdel-Hakeem
- Zoology and Entomology Department, Faculty of Science, Assiut University, Assiut 71526, Egypt
| | | | - Mohamed Salah Eldin Youssef
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | | | | | | |
Collapse
|
17
|
Jahanbani F, Sing JC, Maynard RD, Jahanbani S, Dafoe J, Dafoe W, Jones N, Wallace KJ, Rastan A, Maecker HT, Röst HL, Snyder MP, Davis RW. Longitudinal cytokine and multi-modal health data of an extremely severe ME/CFS patient with HSD reveals insights into immunopathology, and disease severity. Front Immunol 2024; 15:1369295. [PMID: 38650940 PMCID: PMC11033372 DOI: 10.3389/fimmu.2024.1369295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) presents substantial challenges in patient care due to its intricate multisystem nature, comorbidities, and global prevalence. The heterogeneity among patient populations, coupled with the absence of FDA-approved diagnostics and therapeutics, further complicates research into disease etiology and patient managment. Integrating longitudinal multi-omics data with clinical, health,textual, pharmaceutical, and nutraceutical data offers a promising avenue to address these complexities, aiding in the identification of underlying causes and providing insights into effective therapeutics and diagnostic strategies. Methods This study focused on an exceptionally severe ME/CFS patient with hypermobility spectrum disorder (HSD) during a period of marginal symptom improvements. Longitudinal cytokine profiling was conducted alongside the collection of extensive multi-modal health data to explore the dynamic nature of symptoms, severity, triggers, and modifying factors. Additionally, an updated severity assessment platform and two applications, ME-CFSTrackerApp and LexiTime, were introduced to facilitate real-time symptom tracking and enhance patient-physician/researcher communication, and evaluate response to medical intervention. Results Longitudinal cytokine profiling revealed the significance of Th2-type cytokines and highlighted synergistic activities between mast cells and eosinophils, skewing Th1 toward Th2 immune responses in ME/CFS pathogenesis, particularly in cognitive impairment and sensorial intolerance. This suggests a potentially shared underlying mechanism with major ME/CFS comorbidities such as HSD, Mast cell activation syndrome, postural orthostatic tachycardia syndrome (POTS), and small fiber neuropathy. Additionally, the data identified potential roles of BCL6 and TP53 pathways in ME/CFS etiology and emphasized the importance of investigating adverse reactions to medication and supplements and drug interactions in ME/CFS severity and progression. Discussion Our study advocates for the integration of longitudinal multi-omics with multi-modal health data and artificial intelligence (AI) techniques to better understand ME/CFS and its major comorbidities. These findings highlight the significance of dysregulated Th2-type cytokines in patient stratification and precision medicine strategies. Additionally, our results suggest exploring the use of low-dose drugs with partial agonist activity as a potential avenue for ME/CFS treatment. This comprehensive approach emphasizes the importance of adopting a patient-centered care approach to improve ME/CFS healthcare management, disease severity assessment, and personalized medicine. Overall, these findings contribute to our understanding of ME/CFS and offer avenues for future research and clinical practice.
Collapse
Affiliation(s)
- Fereshteh Jahanbani
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Justin Cyril Sing
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - Rajan Douglas Maynard
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Shaghayegh Jahanbani
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Veterans Affairs (VA) Palo Alto Health Care System, Palo Alto, CA, United States
| | - Janet Dafoe
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Whitney Dafoe
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Nathan Jones
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Kelvin J. Wallace
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Azuravesta Rastan
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Holden T. Maecker
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Pulmonary and Critical Care Medicine, Institute of Immunity, Transplantation, and Infectious Diseases, Stanford University, Palo Alto, CA, United States
| | - Hannes L. Röst
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Ronald W. Davis
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
18
|
I Kutyavin V, Korn LL, Medzhitov R. Nutrient-derived signals regulate eosinophil adaptation to the small intestine. Proc Natl Acad Sci U S A 2024; 121:e2316446121. [PMID: 38271336 PMCID: PMC10835075 DOI: 10.1073/pnas.2316446121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/05/2023] [Indexed: 01/27/2024] Open
Abstract
Eosinophils are well recognized as effector cells of type 2 immunity, yet they also accumulate in many tissues under homeostatic conditions. However, the processes that govern homeostatic eosinophil accumulation and tissue-specific adaptation, and their functional significance, remain poorly defined. Here, we investigated how eosinophils adapt to the small intestine (SI) microenvironment and the local signals that regulate this process. We observed that eosinophils gradually migrate along the crypt-villus axis, giving rise to a villus-resident subpopulation with a distinct transcriptional signature. Retinoic acid signaling was specifically required for maintenance of this subpopulation, while IL-5 was largely dispensable outside of its canonical role in eosinophil production. Surprisingly, we found that a high-protein diet suppressed the accumulation of villus-resident eosinophils. Purified amino acids were sufficient for this effect, which was a consequence of accelerated eosinophil turnover within the tissue microenvironment and was not due to altered development in the bone marrow. Our study provides insight into the process of eosinophil adaptation to the SI, highlighting its reliance on nutrient-derived signals.
Collapse
Affiliation(s)
- Vassily I Kutyavin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Lisa L Korn
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
- Department of Medicine (Rheumatology), Yale University School of Medicine, New Haven, CT 06510
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
- HHMI, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|
19
|
Liu R, Shao W, Xu J, Chu C. To be or not to be direct: The role of neuromedin U in neuro-eosinophil crosstalk. Sci Bull (Beijing) 2024; 69:148-150. [PMID: 38071108 DOI: 10.1016/j.scib.2023.11.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Affiliation(s)
- Ruichao Liu
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan 030001, China; Institute for Immunology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wenhao Shao
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan 030001, China; Shanxi Medical University, Taiyuan 030012, China
| | - Jun Xu
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan 030001, China; Department of Surgery, First Hospital of Shanxi Medical University, Shanxi Provincial Key Laboratory for Digestive Diseases and Organ Transplantation, Taiyuan 030001, China.
| | - Coco Chu
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan 030001, China; Institute for Immunology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
20
|
DeAnglis IK, Andrews BR, Lock LR, Dyer KE, Yang A, Volokhov DV, Fenton MB, Simmons NB, Downs CJ, Becker DJ. Bat cellular immunity varies by year and dietary habit amidst land conversion. CONSERVATION PHYSIOLOGY 2024; 12:coad102. [PMID: 38293641 PMCID: PMC10823333 DOI: 10.1093/conphys/coad102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 10/11/2023] [Accepted: 11/28/2023] [Indexed: 02/01/2024]
Abstract
Monitoring the health of wildlife populations is essential in the face of increased agricultural expansion and forest fragmentation. Loss of habitat and habitat degradation can negatively affect an animal's physiological state, possibly resulting in immunosuppression and increased morbidity or mortality. We sought to determine how land conversion may differentially impact cellular immunity and infection risk in Neotropical bats species regularly infected with bloodborne pathogens, and to evaluate how effects may vary over time and by dietary habit. We studied common vampire bats (Desmodus rotundus), northern yellow-shouldered bats (Sturnira parvidens) and Mesoamerican mustached bats (Pteronotus mesoamericanus), representing the dietary habits of sanguivory, frugivory and insectivory respectively, in northern Belize. We compared estimated total white blood cell count, leukocyte differentials, neutrophil to lymphocyte ratio and infection status with two bloodborne bacterial pathogens (Bartonella spp. and hemoplasmas) of 118 bats captured in a broadleaf, secondary forest over three years (2017-2019). During this period, tree cover decreased by 14.5% while rangeland expanded by 14.3%, indicating increasing habitat loss and fragmentation. We found evidence for bat species-specific responses of cellular immunity between years, with neutrophil counts significantly decreasing in S. parvidens from 2017 to 2018, but marginally increasing in D. rotundus. However, the odds of infection with Bartonella spp. and hemoplasmas between 2017 and 2019 did not differ between bat species, contrary to our prediction that pathogen prevalence may increase with land conversion. We conclude that each bat species invested differently in cellular immunity in ways that changed over years of increasing habitat loss and fragmentation. We recommend further research on the interactions between land conversion, immunity and infection across dietary habits of Neotropical bats for informed management and conservation.
Collapse
Affiliation(s)
- Isabella K DeAnglis
- Department of Environmental Biology, SUNY College of Environmental Science and Forestry, 1 Forestry Drive, Syracuse, NY, 13210, USA
- Department of Biological Sciences, University of Arkansas, 1 University of Arkansas, Fayetteville, AR, 72701, USA
| | - Benjamin R Andrews
- Department of Environmental Biology, SUNY College of Environmental Science and Forestry, 1 Forestry Drive, Syracuse, NY, 13210, USA
| | - Lauren R Lock
- School of Biological Sciences, University of Oklahoma, 730 Van Vleet Oval, Norman, OK, 73019, USA
| | - Kristin E Dyer
- School of Biological Sciences, University of Oklahoma, 730 Van Vleet Oval, Norman, OK, 73019, USA
| | - Anni Yang
- Department of Geography and Environmental Sustainability, University of Oklahoma, 100 East Boyd St, Norman, OK, 73019, USA
| | - Dmitriy V Volokhov
- Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - M Brock Fenton
- Department of Biology, University of Western Ontario, 1151 Richmond Street, London, Ontario, N6A 3K7, Canada
| | - Nancy B Simmons
- Department of Mammalogy, Division of Vertebrate Zoology, American Museum of Natural History, 200 Central Park West, New York, NY, 10024, USA
| | - Cynthia J Downs
- Department of Environmental Biology, SUNY College of Environmental Science and Forestry, 1 Forestry Drive, Syracuse, NY, 13210, USA
| | - Daniel J Becker
- School of Biological Sciences, University of Oklahoma, 730 Van Vleet Oval, Norman, OK, 73019, USA
| |
Collapse
|
21
|
Lekki-Jóźwiak J, Bąska P. The Roles of Various Immune Cell Populations in Immune Response against Helminths. Int J Mol Sci 2023; 25:420. [PMID: 38203591 PMCID: PMC10778651 DOI: 10.3390/ijms25010420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Helminths are multicellular parasites that are a substantial problem for both human and veterinary medicine. According to estimates, 1.5 billion people suffer from their infection, resulting in decreased life quality and burdens for healthcare systems. On the other hand, these infections may alleviate autoimmune diseases and allergy symptoms. The immune system is programmed to combat infections; nevertheless, its effector mechanisms may result in immunopathologies and exacerbate clinical symptoms. This review summarizes the role of the immune response against worms, with an emphasis on the Th2 response, which is a hallmark of helminth infections. We characterize non-immune cells (enteric tuft cells-ETCs) responsible for detecting parasites, as well as the role of hematopoietic-derived cells (macrophages, basophils, eosinophils, neutrophils, innate lymphoid cells group 2-ILC2s, mast cells, T cells, and B cells) in initiating and sustaining the immune response, as well as the functions they play in granulomas. The aim of this paper is to review the existing knowledge regarding the immune response against helminths, to attempt to decipher the interactions between cells engaged in the response, and to indicate the gaps in the current knowledge.
Collapse
Affiliation(s)
- Janina Lekki-Jóźwiak
- Division of Parasitology and Parasitic Diseases, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland;
| | - Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| |
Collapse
|
22
|
Conca W, Saleh SM, Al-Rabiah R, Parhar RS, Abd-Elnaeim M, Al-Hindas H, Tinson A, Kroell KB, Liedl KR, Collison K, Kishore U, Al-Mohanna F. The immunoglobulin A isotype of the Arabian camel ( Camelus dromedarius) preserves the dualistic structure of unconventional single-domain and canonical heavy chains. Front Immunol 2023; 14:1289769. [PMID: 38162642 PMCID: PMC10756906 DOI: 10.3389/fimmu.2023.1289769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/30/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction The evolution of adaptive immunity in Camelidae resulted in the concurrent expression of classic heterotetrameric and unconventional homodimeric heavy chain-only IgG antibodies. Heavy chain-only IgG bears a single variable domain and lacks the constant heavy (CH) γ1 domain required for pairing with the light chain. It has not been reported whether this distinctive feature of IgG is also observed in the IgA isotype. Methods Gene-specific primers were used to generate an IgA heavy chain cDNA library derived from RNA extracted from the dromedary's third eyelid where isolated lymphoid follicles and plasma cells abound at inductive and effector sites, respectively. Results Majority of the cDNA clones revealed hallmarks of heavy chain-only antibodies, i.e. camelid-specific amino acid substitutions in framework region 1 and 2, broad length distribution of complementarity determining region 3, and the absence of the CHα1 domain. In a few clones, however, the cDNA of the canonical IgA heavy chain was amplified which included the CHα1 domain, analogous to CHγ1 domain in IgG1 subclass. Moreover, we noticed a short, proline-rich hinge, and, at the N-terminal end of the CHα3 domain, a unique, camelid-specific pentapeptide of undetermined function, designated as the inter-α region. Immunoblots using rabbit anti-camel IgA antibodies raised against CHα2 and CHα3 domains as well as the inter-α region revealed the expression of a ~52 kDa and a ~60 kDa IgA species, corresponding to unconventional and canonical IgA heavy chain, respectively, in the third eyelid, trachea, small and large intestine. In contrast, the leporine anti-CHα1 antibody detected canonical, but not unconventional IgA heavy chain, in all the examined tissues, milk, and serum, in addition to another hitherto unexplored species of ~45 kDa in milk and serum. Immunohistology using anti-CHα domain antibodies confirmed the expression of both variants of IgA heavy chains in plasma cells in the third eyelid's lacrimal gland, conjunctiva, tracheal and intestinal mucosa. Conclusion We found that in the dromedary, the IgA isotype has expanded the immunoglobulin repertoire by co-expressing unconventional and canonical IgA heavy chains, comparable to the IgG class, thus underscoring the crucial role of heavy chain-only antibodies not only in circulation but also at the mucosal frontiers.
Collapse
Affiliation(s)
- Walter Conca
- Department of Executive Health Medicine, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Soad M. Saleh
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Rana Al-Rabiah
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Ranjit Singh Parhar
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Mahmoud Abd-Elnaeim
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Hussein Al-Hindas
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Alexander Tinson
- Management of Scientific Centers and Presidential Camels, Department of President’s Affairs, Hilli ET and Cloning Centre, Al Ain, United Arab Emirates
| | | | - Klaus Roman Liedl
- Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, Austria
| | - Kate Collison
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Uday Kishore
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Futwan Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
23
|
Xiong L, Chen Y, Chen L, Hua R, Shen N, Yang G. Enhanced protective immunity against Baylisascaris schroederi infection in mice through a multi-antigen cocktail vaccine approach. Parasitol Res 2023; 123:20. [PMID: 38072876 DOI: 10.1007/s00436-023-08016-w] [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: 10/14/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023]
Abstract
Baylisascaris schroederi is among the most severe intestinal nematodes affecting giant pandas. Developing effective and secure vaccines can be used as a novel strategy for controlling repeated roundworm infection and addressing drug resistance. In our previous study, three recombinant antigens (rBsHP2, rBsGAL, and rBsUP) exhibited promising effects against B. schroederi infection in the mice model. This study extends the findings by formulating four-form cocktail vaccines (GAL+UP, HP2+UP, GAL+HP2, and GAL+HP2+UP) using three B. schroederi recombinant antigens to improve protection in mice further. Additionally, the protective differences after immunizing mice with different doses of cocktail antigens (150 μg, 100 μg, and 50 μg) were analyzed. Administration of rBs(GAL+UP), rBs(HP2+UP), rBs(GAL+HP2), and rBs(GAL+HP2+UP) significantly reduced liver and lung lesions, along with a decrease in L3 larvae by 83.7%, 82.1%, 76.4%, and 75.1%, respectively. These vaccines induced a Th1/Th2 mixed immunity, evidenced by elevated serum antibody levels (IgG, IgG1, IgG2a, IgE, and IgA) and splenocyte cytokines [interferon gamma (IFN-γ), interleukin (IL)-5, and IL-10]. Furthermore, varying cocktail vaccine dosages did not significantly affect protection. The results confirm that a 50 μg rBs(GAL+UP) dosage holds promise as a better candidate vaccine combination against B. schroederi infection, providing a basis for developing the B. schroederi vaccine.
Collapse
Affiliation(s)
- Lang Xiong
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Sichuan, China
| | - Yanxin Chen
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Sichuan, China
| | - Ling Chen
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Sichuan, China
| | - Ruiqi Hua
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Sichuan, China
| | - Nengxing Shen
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Sichuan, China
| | - Guangyou Yang
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Sichuan, China.
| |
Collapse
|
24
|
Jesenak M, Diamant Z, Simon D, Tufvesson E, Seys SF, Mukherjee M, Lacy P, Vijverberg S, Slisz T, Sediva A, Simon HU, Striz I, Plevkova J, Schwarze J, Kosturiak R, Alexis NE, Untersmayr E, Vasakova MK, Knol E, Koenderman L. Eosinophils-from cradle to grave: An EAACI task force paper on new molecular insights and clinical functions of eosinophils and the clinical effects of targeted eosinophil depletion. Allergy 2023; 78:3077-3102. [PMID: 37702095 DOI: 10.1111/all.15884] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/21/2023] [Accepted: 08/27/2023] [Indexed: 09/14/2023]
Abstract
Over the past years, eosinophils have become a focus of scientific interest, especially in the context of their recently uncovered functions (e.g. antiviral, anti-inflammatory, regulatory). These versatile cells display both beneficial and detrimental activities under various physiological and pathological conditions. Eosinophils are involved in the pathogenesis of many diseases which can be classified into primary (clonal) and secondary (reactive) disorders and idiopathic (hyper)eosinophilic syndromes. Depending on the biological specimen, the eosinophil count in different body compartments may serve as a biomarker reflecting the underlying pathophysiology and/or activity of distinct diseases and as a therapy-driving (predictive) and monitoring tool. Personalized selection of an appropriate therapeutic strategy directly or indirectly targeting the increased number and/or activity of eosinophils should be based on the understanding of eosinophil homeostasis including their interactions with other immune and non-immune cells within different body compartments. Hence, restoring as well as maintaining homeostasis within an individual's eosinophil pool is a goal of both specific and non-specific eosinophil-targeting therapies. Despite the overall favourable safety profile of the currently available anti-eosinophil biologics, the effect of eosinophil depletion should be monitored from the perspective of possible unwanted consequences.
Collapse
Affiliation(s)
- Milos Jesenak
- Department of Clinical Immunology and Allergology, University Teaching Hospital in Martin, Martin, Slovak Republic
- Department of Paediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovak Republic
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovak Republic
| | - Zuzana Diamant
- Department of Clinical Sciences Lund, Respiratory Medicine, Allergology and Palliative Medicine, Lund University, Lund, Sweden
- Department Microbiology Immunology & Transplantation, KU Leuven, Catholic University of Leuven, Leuven, Belgium
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ellen Tufvesson
- Department of Clinical Sciences Lund, Respiratory Medicine, Allergology and Palliative Medicine, Lund University, Lund, Sweden
| | - Sven F Seys
- Laboratory of Clinical Immunology, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Manali Mukherjee
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- The Firestone Institute for Respiratory Health, Research Institute of St. Joe's Hamilton, Hamilton, Ontario, Canada
| | - Paige Lacy
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Susanne Vijverberg
- Amsterdam UMC Location University of Amsterdam, Pulmonary Diseases, Amsterdam, The Netherlands
| | - Tomas Slisz
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Anna Sediva
- Department of Immunology, 2nd Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Ilja Striz
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jana Plevkova
- Department of Pathophysiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | - Jurgen Schwarze
- Child Life and Health and Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Radovan Kosturiak
- Department of Paediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovak Republic
- Outpatient Clinic for Clinical Immunology and Allergology, Nitra, Slovak Republic
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, Department of Paediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Martina Koziar Vasakova
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Edward Knol
- Department Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department Dermatology/Allergology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leo Koenderman
- Department Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department Pulmonary Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
25
|
Plum T, Binzberger R, Thiele R, Shang F, Postrach D, Fung C, Fortea M, Stakenborg N, Wang Z, Tappe-Theodor A, Poth T, MacLaren DAA, Boeckxstaens G, Kuner R, Pitzer C, Monyer H, Xin C, Bonventre JV, Tanaka S, Voehringer D, Vanden Berghe P, Strid J, Feyerabend TB, Rodewald HR. Mast cells link immune sensing to antigen-avoidance behaviour. Nature 2023; 620:634-642. [PMID: 37438525 PMCID: PMC10432277 DOI: 10.1038/s41586-023-06188-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 05/10/2023] [Indexed: 07/14/2023]
Abstract
The physiological functions of mast cells remain largely an enigma. In the context of barrier damage, mast cells are integrated in type 2 immunity and, together with immunoglobulin E (IgE), promote allergic diseases. Allergic symptoms may, however, facilitate expulsion of allergens, toxins and parasites and trigger future antigen avoidance1-3. Here, we show that antigen-specific avoidance behaviour in inbred mice4,5 is critically dependent on mast cells; hence, we identify the immunological sensor cell linking antigen recognition to avoidance behaviour. Avoidance prevented antigen-driven adaptive, innate and mucosal immune activation and inflammation in the stomach and small intestine. Avoidance was IgE dependent, promoted by Th2 cytokines in the immunization phase and by IgE in the execution phase. Mucosal mast cells lining the stomach and small intestine rapidly sensed antigen ingestion. We interrogated potential signalling routes between mast cells and the brain using mutant mice, pharmacological inhibition, neural activity recordings and vagotomy. Inhibition of leukotriene synthesis impaired avoidance, but overall no single pathway interruption completely abrogated avoidance, indicating complex regulation. Collectively, the stage for antigen avoidance is set when adaptive immunity equips mast cells with IgE as a telltale of past immune responses. On subsequent antigen ingestion, mast cells signal termination of antigen intake. Prevention of immunopathology-causing, continuous and futile responses against per se innocuous antigens or of repeated ingestion of toxins through mast-cell-mediated antigen-avoidance behaviour may be an important arm of immunity.
Collapse
Affiliation(s)
- Thomas Plum
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany.
| | - Rebecca Binzberger
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Robin Thiele
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Fuwei Shang
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Daniel Postrach
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Candice Fung
- Laboratory for Enteric NeuroScience Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Marina Fortea
- Laboratory for Enteric NeuroScience Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Laboratory for Intestinal Neuroimmune Interactions, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Zheng Wang
- Laboratory for Intestinal Neuroimmune Interactions, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | | | - Tanja Poth
- Center for Model System and Comparative Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Duncan A A MacLaren
- Department of Clinical Neurobiology of the Medical Faculty of Heidelberg University and German Cancer Research Center, Heidelberg, Germany
| | - Guy Boeckxstaens
- Laboratory for Intestinal Neuroimmune Interactions, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Rohini Kuner
- Pharmacology Institute, Heidelberg University, Heidelberg, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology of the Medical Faculty of Heidelberg University and German Cancer Research Center, Heidelberg, Germany
| | - Cuiyan Xin
- Division of Renal Medicine and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph V Bonventre
- Division of Renal Medicine and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Tanaka
- Laboratory of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Jessica Strid
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Thorsten B Feyerabend
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Hans-Reimer Rodewald
- Division for Cellular Immunology, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
26
|
Macchia I, La Sorsa V, Urbani F, Moretti S, Antonucci C, Afferni C, Schiavoni G. Eosinophils as potential biomarkers in respiratory viral infections. Front Immunol 2023; 14:1170035. [PMID: 37483591 PMCID: PMC10358847 DOI: 10.3389/fimmu.2023.1170035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/30/2023] [Indexed: 07/25/2023] Open
Abstract
Eosinophils are bone marrow-derived granulocytes that, under homeostatic conditions, account for as much as 1-3% of peripheral blood leukocytes. During inflammation, eosinophils can rapidly expand and infiltrate inflamed tissues, guided by cytokines and alarmins (such as IL-33), adhesion molecules and chemokines. Eosinophils play a prominent role in allergic asthma and parasitic infections. Nonetheless, they participate in the immune response against respiratory viruses such as respiratory syncytial virus and influenza. Notably, respiratory viruses are associated with asthma exacerbation. Eosinophils release several molecules endowed with antiviral activity, including cationic proteins, RNases and reactive oxygen and nitrogen species. On the other hand, eosinophils release several cytokines involved in homeostasis maintenance and Th2-related inflammation. In the context of SARS-CoV-2 infection, emerging evidence indicates that eosinophils can represent possible blood-based biomarkers for diagnosis, prognosis, and severity prediction of disease. In particular, eosinopenia seems to be an indicator of severity among patients with COVID-19, whereas an increased eosinophil count is associated with a better prognosis, including a lower incidence of complications and mortality. In the present review, we provide an overview of the role and plasticity of eosinophils focusing on various respiratory viral infections and in the context of viral and allergic disease comorbidities. We will discuss the potential utility of eosinophils as prognostic/predictive immune biomarkers in emerging respiratory viral diseases, particularly COVID-19. Finally, we will revisit some of the relevant methods and tools that have contributed to the advances in the dissection of various eosinophil subsets in different pathological settings for future biomarker definition.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina La Sorsa
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| | - Caterina Antonucci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Claudia Afferni
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
27
|
Muñoz-Guzmán MA, Cuenca-Verde C, Sánchez-Paredes A, Prado-Ochoa MG, Iturbe-Requena SL, Alba-Hurtado F. Subclinical experimental infection with Taenia hydatigena in sheep is associated with production losses. Exp Parasitol 2023:108566. [PMID: 37308001 DOI: 10.1016/j.exppara.2023.108566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
The effect of experimental infection by Taenia hydatigena metacestodes on different productive parameters in sheep was evaluated. Seventeen male Columbia lambs distributed in three groups were used. The lambs of the first group (n = 5) were orally inoculated with 1000 T. hydatigena eggs (low dose). The lambs of the second group (n = 5) were inoculated orally with all the eggs of the last proglottid of an adult cestode (high dose). The lambs of the third group (n = 7) only received a placebo and were used as a control group. All lambs were humanely euthanized at week 13 postinfection, and carcass yield and conformation were evaluated. The infection rates of the lambs from the high-dose infected group and the low-dose infected group were 100% and 40%, respectively, with a mean of 2.4 ± 0.6 and 1 ± 0.7 metacestodes of T. hydatigena in the abdominal cavity, respectively. In a first multivariate study (MANOVA) considering the under the curve (AUC) values of body condition, weight gain, and feed consumption, as well as the final feed conversion values, showed a highly significant (p < 0.0001) group effect (high dose/low dose/uninfected). In a second multivariate study (MANOVA) considering the AUC values of eosinophils blood count, packed cell volume and alkaline phosphatase showed a highly significant (p < 0.0001) group effect (high dose/low dose/uninfected). The increase in serum ALP is a consequence of liver damage that had a strong negative correlation (r = -0.63, p < 0.007) with the body condition of the lambs. Despite these findings, we did not observe obvious clinical manifestations in any of the infected lambs. In general, no differences (p > 0.1) were observed between the lambs of the control group and the lambs of the low-dose infected group in the parameters evaluated. The results of this study show that the infection of T. hydatigena metacestodes subclinically produces a decrease in productive efficiency, alterations of some hematological and biochemical values, and a slight deterioration in the general appearance of the infected lambs. The above aspects are rarely detected by most farmers, but they have a negative impact on the productivity of infected lambs.
Collapse
Affiliation(s)
- Marco Antonio Muñoz-Guzmán
- Departamento de Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Mexico
| | - César Cuenca-Verde
- Departamento de Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Mexico
| | - Adolfo Sánchez-Paredes
- Departamento de Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Mexico
| | - María Guadalupe Prado-Ochoa
- Departamento de Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Mexico
| | - Sandra Lizeth Iturbe-Requena
- Departamento de Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Mexico
| | - Fernando Alba-Hurtado
- Departamento de Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Mexico.
| |
Collapse
|
28
|
Groh M, Rohmer J, Etienne N, Abou Chahla W, Baudet A, Chan Hew Wai A, Chenivesse C, Clisson Rusek I, Cottin V, Decamp M, De Groote P, Delahousse F, Duployez N, Faguer S, Gottrand F, Huang F, Leblanc T, Magnan A, Martin T, Mortuaire G, Néel A, Paris L, Petit A, Rossignol J, Schleinitz N, Soret-Dulphy J, Staumont-Salle D, Terrier B, Terriou L, Viallard JF, Lefèvre G, Kahn JE. French guidelines for the etiological workup of eosinophilia and the management of hypereosinophilic syndromes. Orphanet J Rare Dis 2023; 18:100. [PMID: 37122022 PMCID: PMC10148979 DOI: 10.1186/s13023-023-02696-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/02/2023] [Indexed: 05/02/2023] Open
Abstract
Eosinophilic-related clinical manifestations are protean and the underlying conditions underpinning eosinophilia are highly diverse. The etiological workup of unexplained eosinophilia/hypereosinophilia can be challenging, and can lead sometimes to extensive, inappropriate, costly and/or invasive investigations. To date, guidelines for the etiological workup and management of eosinophilia are mainly issued by hematologists, and thus mostly cover the scope of clonal hypereosinophilic syndromes (HES). Here, thanks to an extensive literature review, and thanks to the joint work of a large panel of experts involving physicians from both adult and pediatric medicine and from various subspecialties (as well as a representative of a patients' association representative), we provide recommendations for both the step-by step diagnostic workup of eosinophilia (whether unexplained or within specific contexts) as well as the management and follow-up of the full spectrum of eosinophilic disorders (including clonal, reactive, lymphocytic and idiopathic HES, as well as single-organ diseases). Didactic prescription summaries intended to facilitate the prescription of eosinophil-targeted drugs are also provided, as are practical diagnostic and therapeutic algorithms. Lastly, this set of recommendations also includes a summary intended for general practitioners, as well as an overview of the therapeutic patient education program set up by the French reference center for HES. Further updates will be mandatory as new validated information emerges.
Collapse
Affiliation(s)
- Matthieu Groh
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France.
- Department of Internal Medicine, Hôpital Foch, Suresnes, France.
- Inserm, U1286 - INFINITE - Institute for Translational Research in Inflammation, University of Lille, CHU Lille, Lille, France.
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France.
| | - Julien Rohmer
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, University of Sorbonne-Paris-Cité, APHP, CHU Bichat, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Nicolas Etienne
- Department of Infectious Diseases and Tropical Medicine, University of Sorbonne-Paris-Cité, APHP, CHU Necker-Enfants Malades, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Wadih Abou Chahla
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Pediatric Hematology, University of Lille, CHU Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Antoine Baudet
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, CH Annecy Genevois, Metz Tessy, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Aurélie Chan Hew Wai
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Pharmacology, Hôpital Foch, Suresnes, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Cécile Chenivesse
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- CNRS, Inserm, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, University of Lille, CHU Lille, Lille, France
- CRISALIS (Clinical Research Initiative in Severe Asthma: a Lever for Innovation and Science), F-CRIN Network, INSERM US015, Toulouse, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Irena Clisson Rusek
- Association Pour l'Information sur les Maladies à Eosinophiles, Bourg-la-Reine, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Vincent Cottin
- Department of Respiratory Medicine, Hôpital Louis Pradel, UMR754 INRAE, University of Lyon 1, Hospices Civils de Lyon, Lyon, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Matthieu Decamp
- Department of Cytogenetics, CHU de Caen, Caen, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Pascal De Groote
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Cardiology, University of Lille, CHU Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Fanny Delahousse
- , Nantes, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Nicolas Duployez
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Laboratory of Hematology, University of Lille, CHU Lille, Lille, France
- CNRS, Inserm, IRCL, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Stanislas Faguer
- Department of Nephrology and Organ Transplantation, University of Paul Sabatier Toulouse III, CHU Toulouse, Toulouse, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Frédéric Gottrand
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Inserm, U1286 - INFINITE - Institute for Translational Research in Inflammation, University of Lille, CHU Lille, Lille, France
- Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Jeanne de Flandre Children's Hospital, University of Lille, CHU Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Florent Huang
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Cardiology, Hôpital Foch, Suresnes, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Thierry Leblanc
- Pediatric Hematology and Immunology Department, University Sorbonne-Paris-Cité, APHP, CHU Robert Debré, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Antoine Magnan
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Respiratory Medicine, Hôpital Foch, Suresnes, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Thierry Martin
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, CHU Strasbourg, Strasbourg, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Geoffrey Mortuaire
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Inserm, U1286 - INFINITE - Institute for Translational Research in Inflammation, University of Lille, CHU Lille, Lille, France
- Otorhinolaryngology-Head and Neck Department, University of Lille, CHU de Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Antoine Néel
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, CHU Nantes, Nantes, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Luc Paris
- Department of Parasitology and Mycology, Sorbonne Université, APHP, CHU Pitié-Salpêtrière, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Arnaud Petit
- Department of Hematology and Pediatric Oncology, Sorbonne Université, APHP, CHU Armand Trousseau, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Julien Rossignol
- Department of Hematology, University of Sorbonne-Paris-Cité, APHP, CHU Necker, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Nicolas Schleinitz
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, APHM, CHU La Timone, Marseille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Juliette Soret-Dulphy
- Centre d'Investigation Clinique, University of Sorbonne-Paris-Cité, AP-HP, CHU St-Louis, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Delphine Staumont-Salle
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Dermatology, University of Lille, CHU de Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Benjamin Terrier
- Department of Internal Medicine, University of Sorbonne-Paris-Cité, AP-HP, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Louis Terriou
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine and Clinical Immunology, University of Lille, CHU de Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Jean-François Viallard
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, CHU de Bordeaux, Bordeaux, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Guillaume Lefèvre
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Inserm, U1286 - INFINITE - Institute for Translational Research in Inflammation, University of Lille, CHU Lille, Lille, France
- Department of Internal Medicine and Clinical Immunology, University of Lille, CHU de Lille, Lille, France
- Department of Internal Medicine, CHU de Bordeaux, Bordeaux, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Jean-Emmanuel Kahn
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Institut d'Immunologie, University of Lille, CHU de Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| |
Collapse
|
29
|
Zhuang TZ, Ravindranathan D, Liu Y, Martini DJ, Brown JT, Nazha B, Russler G, Yantorni LB, Caulfield S, Carthon BC, Kucuk O, Master VA, Bilen MA. Baseline Neutrophil-to-Eosinophil Ratio Is Associated with Outcomes in Metastatic Renal Cell Carcinoma Treated with Immune Checkpoint Inhibitors. Oncologist 2023; 28:239-245. [PMID: 36427017 PMCID: PMC10020802 DOI: 10.1093/oncolo/oyac236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/15/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Biomarkers have the potential to guide treatment selection and clinical care in metastatic renal cell carcinoma (mRCC) in an expanding treatment landscape. We report baseline neutrophil-to-eosinophil ratios (NER) in patients with mRCC treated with immune checkpoint inhibitors (CPIs) and their association with clinical outcomes. METHODS We conducted a retrospective review of patients with mRCC treated with CPIs at Winship Cancer Institute from 2015 to 2020 in the United States of America (USA). Demographics, disease characteristics, and laboratory data, including complete blood counts (CBC) were described at the initiation of CPIs. Clinical outcomes were measured as overall survival (OS), progression-free survival (PFS), and clinical benefit (CB) associated with baseline lab values. RESULTS A total of 184 patients were included with a median follow-up time of 25.4 months. Patients with baseline NER were categorized into high or low subgroups; high group was defined as NER >49.2 and low group was defined as NER <49.2 with 25% of patients in the high NER group. Univariate analyses (UVA) and multivariable analyses (MVA) identified decreased overall survival (OS) associated with elevated NER. In MVA, patients with a high baseline NER group had a hazard ratio (HR) of 1.68 (95%CI, 1.01-2.82, P = .048) for OS; however, there was no significant difference between groups for PFS. Clinical benefit was seen in 47.3% of patients with low baseline NER and 40% with high NER. CONCLUSIONS We conclude that elevated baseline NER may be associated with worse clinical outcomes in mRCC. Although results require further validation, NER is a feasible biomarker in patients with CPI-treated mRCC.
Collapse
Affiliation(s)
- Tony Z Zhuang
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Deepak Ravindranathan
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Grady Cancer Center for Excellence, Grady Memorial Hospital, Atlanta, GA, USA
| | - Yuan Liu
- Departments of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Dylan J Martini
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jacqueline T Brown
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Bassel Nazha
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Grady Cancer Center for Excellence, Grady Memorial Hospital, Atlanta, GA, USA
| | - Greta Russler
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lauren B Yantorni
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sarah Caulfield
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Bradley C Carthon
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Grady Cancer Center for Excellence, Grady Memorial Hospital, Atlanta, GA, USA
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Grady Cancer Center for Excellence, Grady Memorial Hospital, Atlanta, GA, USA
| | - Viraj A Master
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mehmet Asim Bilen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
30
|
Bakker J, Maaskant A, Wegman M, Zijlmans DGM, Hage P, Langermans JAM, Remarque EJ. Reference Intervals and Percentiles for Hematologic and Serum Biochemical Values in Captive Bred Rhesus ( Macaca mulatta) and Cynomolgus Macaques ( Macaca fascicularis). Animals (Basel) 2023; 13:445. [PMID: 36766334 PMCID: PMC9913310 DOI: 10.3390/ani13030445] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 02/03/2023] Open
Abstract
Several physiological characteristics and housing conditions are known to affect hematologic and serum biochemical values in macaques. However, the studies that have been conducted either report values calculated based on a small number of animals, were designed specifically to document the effect of a particular condition on the normal range of hematologic and serum biochemical values, or used parametric assumptions to calculate hematologic and serum biochemical reference intervals. We conducted a retrospective longitudinal cohort study to estimate reference intervals for hematologic and serum biochemical values in clinically healthy macaques based on observed percentiles without parametric assumptions. Data were obtained as part of the Biomedical Primate Research Centre (Rijswijk, The Netherlands) health monitoring program between 2018 and 2021. In total, 4009 blood samples from 1475 macaques were analyzed with a maximum of one repeat per year per animal. Data were established by species, gender, age, weight-for-height indices, pregnancy, sedation protocol, and housing conditions. Most of the parameters profoundly affected just some hematologic and serum biochemical values. A significant glucose difference was observed between the ketamine and ketamine-medetomidine sedation protocols. The results emphasize the importance of establishing uniform experimental groups with validated animal husbandry and housing conditions to improve the reproducibility of the experiments.
Collapse
Affiliation(s)
- Jaco Bakker
- Animal Science Department, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ Rijswijk, The Netherlands
| | - Annemiek Maaskant
- Animal Science Department, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ Rijswijk, The Netherlands
- Department Population Health Sciences, Animals in Science & Society, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
| | - Merel Wegman
- Animal Science Department, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ Rijswijk, The Netherlands
| | - Dian G. M. Zijlmans
- Animal Science Department, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ Rijswijk, The Netherlands
| | - Patrice Hage
- Department Population Health Sciences, Animals in Science & Society, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
| | - Jan A. M. Langermans
- Animal Science Department, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ Rijswijk, The Netherlands
- Department Population Health Sciences, Animals in Science & Society, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
| | - Edmond J. Remarque
- Department of Virology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ Rijswijk, The Netherlands
| |
Collapse
|
31
|
Jackson DJ, Pavord ID. Living without eosinophils: evidence from mouse and man. Eur Respir J 2023; 61:13993003.01217-2022. [PMID: 35953100 PMCID: PMC9834633 DOI: 10.1183/13993003.01217-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/25/2022] [Indexed: 01/19/2023]
Abstract
The enduring view of eosinophils, as immune effector cells whose primary function is host defence against infection by helminths and other microbial pathogens, sets the stage for a fundamental question regarding the safety of therapeutic eosinophil depletion. If eosinophils are significantly reduced or altogether depleted in an effort to alleviate the negative effects of tissue eosinophilia and eosinophilic inflammation in conditions such as asthma, COPD, chronic rhinosinusitis with nasal polyps, eosinophilic granulomatosis with polyangiitis and hypereosinophilic syndrome, would these patients become susceptible to infection or another illness? Development of mouse models in which the eosinophil lineage has been ablated, observations in patients naturally lacking eosinophils and data from studies of eosinophil-depleting medical therapies indicate that the absence of eosinophils is not detrimental to health. The evidence available to date, as presented in this review, supports the conclusion that even if certain homeostatic roles for the eosinophil may be demonstrable in controlled animal models and human in vitro settings, the evolution of the human species appears to have provided sufficient immune redundancy such that one may be hale and hearty without eosinophils.
Collapse
Affiliation(s)
- David J Jackson
- Guy's Severe Asthma Centre, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Ian D Pavord
- Respiratory Medicine Unit and Oxford Respiratory NIHR BRC, University of Oxford, Oxford, UK
| |
Collapse
|
32
|
Mabille D, Dirkx L, Thys S, Vermeersch M, Montenye D, Govaerts M, Hendrickx S, Takac P, Van Weyenbergh J, Pintelon I, Delputte P, Maes L, Pérez-Morga D, Timmermans JP, Caljon G. Impact of pulmonary African trypanosomes on the immunology and function of the lung. Nat Commun 2022; 13:7083. [PMID: 36400767 PMCID: PMC9674601 DOI: 10.1038/s41467-022-34757-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022] Open
Abstract
Approximately 20% of sleeping sickness patients exhibit respiratory complications, however, with a largely unknown role of the parasite. Here we show that tsetse fly-transmitted Trypanosoma brucei parasites rapidly and permanently colonize the lungs and occupy the extravascular spaces surrounding the blood vessels of the alveoli and bronchi. They are present as nests of multiplying parasites exhibiting close interactions with collagen and active secretion of extracellular vesicles. The local immune response shows a substantial increase of monocytes, macrophages, dendritic cells and γδ and activated αβ T cells and a later influx of neutrophils. Interestingly, parasite presence results in a significant reduction of B cells, eosinophils and natural killer cells. T. brucei infected mice show no infection-associated pulmonary dysfunction, mirroring the limited pulmonary clinical complications during sleeping sickness. However, the substantial reduction of the various immune cells may render individuals more susceptible to opportunistic infections, as evident by a co-infection experiment with respiratory syncytial virus. Collectively, these observations provide insights into a largely overlooked target organ, and may trigger new diagnostic and supportive therapeutic approaches for sleeping sickness.
Collapse
Affiliation(s)
- Dorien Mabille
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Laura Dirkx
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Sofie Thys
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Daniel Montenye
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Matthias Govaerts
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Peter Takac
- Institute of Zoology, Slovak Academy of Sciences, 84506, Bratislava, Slovakia
- Scientica, Ltd., 83106, Bratislava, Slovakia
| | - Johan Van Weyenbergh
- Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute of Medical Research, KU Leuven, Leuven, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - David Pérez-Morga
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
33
|
Lung Lipidomic Alterations in Beagle Dogs Infected with Toxocara canis. Animals (Basel) 2022; 12:ani12223080. [PMID: 36428308 PMCID: PMC9686702 DOI: 10.3390/ani12223080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022] Open
Abstract
Toxocariasis, mainly caused by Toxocara canis, and to a lesser extent, Toxocara cati, is a neglected parasitic zoonosis. The mechanisms that underlie the changes in lipid metabolism of T. canis infection in Beagle dogs' lungs remain unclear. Lipidomics is a rapidly emerging approach that enables the global profiling of lipid composition by mass spectrometry. In this study, we performed a non-targeted lipidomic analysis of the lungs of Beagle dogs infected with the roundworm T. canis using liquid chromatography-tandem mass spectrometry (LC-MS/MS). A total of 1197 lipid species were identified, of which 63, 88, and 157 lipid species were significantly altered at 24 h post-infection (hpi), 96 hpi, and 36 days post-infection (dpi), respectively. This global lipidomic profiling identified infection-specific lipid signatures for lung toxocariasis, and represented a comprehensive comparison between the lipid composition of dogs' lungs in the presence and absence of T. canis infection. The potential roles of the identified lipid species in the pathogenesis of T. canis are discussed, which has important implications for better understanding the interaction mechanism between T. canis and the host lung.
Collapse
|
34
|
Thompson-Souza GA, Vasconcelos CRI, Neves JS. Eosinophils: Focus on DNA extracellular traps. Life Sci 2022; 311:121191. [DOI: 10.1016/j.lfs.2022.121191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
|
35
|
Malta KK, Palazzi C, Neves VH, Aguiar Y, Silva TP, Melo RCN. Schistosomiasis Mansoni-Recruited Eosinophils: An Overview in the Granuloma Context. Microorganisms 2022; 10:microorganisms10102022. [PMID: 36296298 PMCID: PMC9607553 DOI: 10.3390/microorganisms10102022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/08/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022] Open
Abstract
Eosinophils are remarkably recruited during schistosomiasis mansoni, one of the most common parasitic diseases worldwide. These cells actively migrate and accumulate at sites of granulomatous inflammation termed granulomas, the main pathological feature of this disease. Eosinophils colonize granulomas as a robust cell population and establish complex interactions with other immune cells and with the granuloma microenvironment. Eosinophils are the most abundant cells in granulomas induced by Schistosoma mansoni infection, but their functions during this disease remain unclear and even controversial. Here, we explore the current information on eosinophils as components of Schistosoma mansoni granulomas in both humans and natural and experimental models and their potential significance as central cells triggered by this infection.
Collapse
|
36
|
Peng J, Federman HG, Hernandez C, Siracusa MC. Communication is key: Innate immune cells regulate host protection to helminths. Front Immunol 2022; 13:995432. [PMID: 36225918 PMCID: PMC9548658 DOI: 10.3389/fimmu.2022.995432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Parasitic helminth infections remain a significant global health issue and are responsible for devastating morbidity and economic hardships. During infection, helminths migrate through different host organs, which results in substantial tissue damage and the release of diverse effector molecules by both hematopoietic and non-hematopoietic cells. Thus, host protective responses to helminths must initiate mechanisms that help to promote worm clearance while simultaneously mitigating tissue injury. The specialized immunity that promotes these responses is termed type 2 inflammation and is initiated by the recruitment and activation of hematopoietic stem/progenitor cells, mast cells, basophils, eosinophils, dendritic cells, neutrophils, macrophages, myeloid-derived suppressor cells, and group 2 innate lymphoid cells. Recent work has also revealed the importance of neuron-derived signals in regulating type 2 inflammation and antihelminth immunity. These studies suggest that multiple body systems coordinate to promote optimal outcomes post-infection. In this review, we will describe the innate immune events that direct the scope and intensity of antihelminth immunity. Further, we will highlight the recent progress made in our understanding of the neuro-immune interactions that regulate these pathways and discuss the conceptual advances they promote.
Collapse
Affiliation(s)
- Jianya Peng
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
- Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Hannah G. Federman
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
- Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Christina M. Hernandez
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
- Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Mark C. Siracusa
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
- Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
- *Correspondence: Mark C. Siracusa,
| |
Collapse
|
37
|
Pourgholaminejad A, Razipour H, Heydarian P, Ashrafi K, Roushan ZA, Sharifdini M. A survey on the seroprevalence of toxocariasis and related risk factors in Eosinophilic children of Northwest Iran. Afr Health Sci 2022; 22:617-625. [PMID: 36910401 PMCID: PMC9993322 DOI: 10.4314/ahs.v22i3.66] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background Toxocariasis is a serious zoonotic helminthic disease caused by the nematodes; Toxocara species. Aim A cross-sectional study was conducted to determine the seroprevalence of toxocariasis and related risk factors in eosinophilic children referred to the pediatrics hospital of Qazvin province northwest Iran during 2019-2020. Methods A total of 200 blood samples were collected from eosinophilic children referred to the Qods Pediatrics Hospital. Demographic data, clinical symptoms, and dogs- and soil-contact history were collected. The presence of anti-Toxocara IgG antibody was evaluated by T. canis IgG ELISA kit. Results Anti-Toxocara IgG antibodies were detected in 14 (7%) of the total eosinophilic children. The seropositive rate of toxocariasis in hyper-eosinophilic children (>1000/mm3) was 15.1%, while the seropositivity was 4.1% in children with eosinophilia status (500-999/mm3). There was a significant association between the eosinophilia rate and seropositivity (P<0.05). Also, seroprevalence in asymptomatic eosinophilic children was 4.4%, while in children with clinical symptoms it was 17.1%. Accordingly, a statistically significant difference was found between clinical symptoms and Toxocara infection (P<0.05). Conclusion The prevalence of toxocariasis in eosinophilic children is a serious health problem in the study area. Therefore, serologic evaluation for the diagnosis of Toxocara infection is recommended for eosinophilic children.
Collapse
Affiliation(s)
- Arash Pourgholaminejad
- Department of Immunology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Habib Razipour
- Department of Medical Parasitology and Mycology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Peyman Heydarian
- Department of Medical Parasitology and Mycology, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Keyhan Ashrafi
- Department of Medical Parasitology and Mycology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Atrkar Roushan
- Department of Biostatistics, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Meysam Sharifdini
- Department of Medical Parasitology and Mycology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
38
|
Lynch CA, Guo Y, Mei A, Kreisel D, Gelman AE, Jacobsen EA, Krupnick AS. Solving the Conundrum of Eosinophils in Alloimmunity. Transplantation 2022; 106:1538-1547. [PMID: 34966103 PMCID: PMC9234098 DOI: 10.1097/tp.0000000000004030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Eosinophils are bone-marrow-derived granulocytes known for their ability to facilitate clearance of parasitic infections and their association with asthma and other inflammatory diseases. The purpose of this review is to discuss the currently available human observational and animal experimental data linking eosinophils to the immunologic response in solid organ transplantation. First, we present observational human studies that demonstrate a link between transplantation and eosinophils yet were unable to define the exact role of this cell population. Next, we describe published experimental models and demonstrate a defined mechanistic role of eosinophils in downregulating the alloimmune response to murine lung transplants. The overall summary of this data suggests that further studies are needed to define the role of eosinophils in multiple solid organ allografts and points to the possibility of manipulating this cell population to improve graft survival.
Collapse
Affiliation(s)
- Cherie Alissa Lynch
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Arizona
| | - Yizhan Guo
- Department of Surgery, University of Maryland, Baltimore Maryland
| | - Alex Mei
- Department of Surgery, University of Maryland, Baltimore Maryland
| | | | | | - Elizabeth A. Jacobsen
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Arizona
| | | |
Collapse
|
39
|
Gayrard C, Bretaudeau A, Gombault P, Hoste H, Gidenne T. Feed incorporation of dehydrated sainfoin: effects on health and performances of does and growing rabbits. WORLD RABBIT SCIENCE 2022. [DOI: 10.4995/wrs.2022.16874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
The performance and health of does and growing rabbits were compared over three consecutive reproductive cycles for three groups of 20 nulliparous does and their litters (DS0, DS13, DS26) fed isonutritive feeds containing 0, 13 or 26% dehydrated sainfoin (DS, Perly cultivar). Feed intake, live weight and fertility of does were not affected by DS feed incorporation. The number of live kits at birth increased linearly with increasing DS incorporation (+1.5 from DS0 to DS26, P=0.042) and the stillborn rate tended to linearly decrease in groups fed DS (16.6 vs. 10.4%, P=0.086). Increasing the level of DS in feeds had no impact on the growth of the kits before weaning, but led to a linear reduction in the post-weaning growth rate (P<0.01, –2 for 26% DS), whereas the feed conversion ratio increased linearly with DS incorporation (P<0.01, 2.91 vs. 2.98, resp. for DS0 and DS26). No effect of DS feed incorporation was detectable on doe and kit mortality rates. Excretion of coccidia by both does and growing rabbits was not affected by DS incorporation. For 70 d old rabbits, the levels of immunoglobulins A and G and of white blood cells were not significantly different between groups and high levels of IgG (average: 8.1 mg/mL) were recorded, suggesting a coccidia infestation. Overall doe mortality remained under 5% and was not affected by the reproductive cycle (P=0.24). The stillborn rate decreased from 18 to 6%, (P<0.01) from cycle 1 to 2, and the number of live rabbits at birth increased from 8.0 to 10.7 (P<0.01). Kit mortality remained low before weaning (under 2.5%), and very low after weaning (<1%). Excretion of coccidia by does decreased from cycle 1 to cycle 3, whereas excretions by growing rabbits remained stable.
Collapse
|
40
|
Ignacio A, Shah K, Bernier-Latmani J, Köller Y, Coakley G, Moyat M, Hamelin R, Armand F, Wong NC, Ramay H, Thomson CA, Burkhard R, Wang H, Dufour A, Geuking MB, McDonald B, Petrova TV, Harris NL, McCoy KD. Small intestinal resident eosinophils maintain gut homeostasis following microbial colonization. Immunity 2022; 55:1250-1267.e12. [PMID: 35709757 DOI: 10.1016/j.immuni.2022.05.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/29/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022]
Abstract
The intestine harbors a large population of resident eosinophils, yet the function of intestinal eosinophils has not been explored. Flow cytometry and whole-mount imaging identified eosinophils residing in the lamina propria along the length of the intestine prior to postnatal microbial colonization. Microscopy, transcriptomic analysis, and mass spectrometry of intestinal tissue revealed villus blunting, altered extracellular matrix, decreased epithelial cell turnover, increased gastrointestinal motility, and decreased lipid absorption in eosinophil-deficient mice. Mechanistically, intestinal epithelial cells released IL-33 in a microbiota-dependent manner, which led to eosinophil activation. The colonization of germ-free mice demonstrated that eosinophil activation in response to microbes regulated villous size alterations, macrophage maturation, epithelial barrier integrity, and intestinal transit. Collectively, our findings demonstrate a critical role for eosinophils in facilitating the mutualistic interactions between the host and microbiota and provide a rationale for the functional significance of their early life recruitment in the small intestine.
Collapse
Affiliation(s)
- Aline Ignacio
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Kathleen Shah
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Jeremiah Bernier-Latmani
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland
| | - Yasmin Köller
- Maurice Müller Laboratories, Department of Biomedical Research, Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
| | - Gillian Coakley
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Mati Moyat
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland; Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Romain Hamelin
- Proteomics Core Facility, Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland
| | - Florence Armand
- Proteomics Core Facility, Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland
| | - Nick C Wong
- Monash Bioinformatics Platform, Monash University, Clayton, VIC 3168, Australia
| | - Hena Ramay
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carolyn A Thomson
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Regula Burkhard
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Haozhe Wang
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Antoine Dufour
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Markus B Geuking
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Braedon McDonald
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4A1, Canada
| | - Tatiana V Petrova
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland; Swiss Institute for Experimental Cancer Research, School of Life Sciences, Swiss Federal Institute of Technology Lausanne, Route Cantonale, 1015 Lausanne, Switzerland
| | - Nicola L Harris
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland; Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia.
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
41
|
Bruschi F, Ashour D, Othman A. Trichinella-induced immunomodulation: Another tale of helminth success. Food Waterborne Parasitol 2022; 27:e00164. [PMID: 35615625 PMCID: PMC9125654 DOI: 10.1016/j.fawpar.2022.e00164] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 01/01/2023] Open
Abstract
Trichinella spiralis is a unique parasite in that both the adults and larvae survive in two different intracellular niches in the same host. The immune response, albeit intense, is highly modulated to ensure the survival of both the host and the parasite. It is skewed to T helper 2 and regulatory arms. Diverse cells from both the innate and adaptive compartments of immunity, including dendritic cells, T regulatory cells, and alternatively activated macrophages are thought to mediate such immunomodulation. The parasite has also an outstanding ability to evade the immune system by several elaborate processes. The molecules derived from the parasites including Trichinella, particularly the components of the excretory-secretory products, are being continually identified and explored for the potential of ameliorating the immunopathology in animal models of diverse inflammatory and autoimmune human diseases. Herein we discuss the various aspects of Trichinella-induced immunomodulation with a special reference to the practical implications of the immune system manipulation in alleviating or possibly curing human diseases.
Collapse
Key Words
- AAM, alternatively activated macrophage
- AW, adult worm
- Allergy
- Autoimmune diseases
- Breg, regulatory B cell
- CAM, classically activated macrophage
- Cancer
- ES L1, ES product of T. spiralis muscle larva
- ES, excretory–secretory
- IFN- γ, interferon-γ
- IIL, intestinal infective larva
- IL, interleukin
- Immune evasion
- Immunomodulation
- ML, muscle larva
- NBL, newborn larva
- NOS, nitric oxide synthase
- TGF-β, transforming growth factor-β
- TLR, toll-like receptor
- TNF- α, tumor necrosis factor-α
- Th, T helper
- Tol-DC, tolerogenic dendritic cell
- Treg, regulatory T cell
- Trichinella
- Trichinella-derived molecules
- Ts-AES, ES from adult T. spiralis
Collapse
Affiliation(s)
- F. Bruschi
- School of Medicine, Department of Translational Research, N.T.M.S., Università di Pisa, Pisa, Italy
| | - D.S. Ashour
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - A.A. Othman
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
42
|
Loke P, Lee SC, Oyesola OO. Effects of helminths on the human immune response and the microbiome. Mucosal Immunol 2022; 15:1224-1233. [PMID: 35732819 DOI: 10.1038/s41385-022-00532-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/17/2022] [Accepted: 05/22/2022] [Indexed: 02/04/2023]
Abstract
Helminths have evolved sophisticated immune regulating mechanisms to prevent rejection by their mammalian host. Our understanding of how the human immune system responds to these parasites remains poor compared to mouse models of infection and this limits our ability to develop vaccines as well as harness their unique properties as therapeutic strategies against inflammatory disorders. Here, we review how recent studies on human challenge infections, self-infected individuals, travelers, and endemic populations have improved our understanding of human type 2 immunity and its effects on the microbiome. The heterogeneity of responses between individuals and the limited access to tissue samples beyond the peripheral blood are challenges that limit human studies on helminths, but also provide opportunities to transform our understanding of human immunology. Organoids and single-cell sequencing are exciting new tools for immunological analysis that may aid this pursuit. Learning about the genetic and immunological basis of resistance, tolerance, and pathogenesis to helminth infections may thus uncover mechanisms that can be utilized for therapeutic purposes.
Collapse
Affiliation(s)
- P'ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Soo Ching Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Oyebola O Oyesola
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
43
|
Blood Eosinophils Are Associated with Efficacy of Targeted Therapy in Patients with Advanced Melanoma. Cancers (Basel) 2022; 14:cancers14092294. [PMID: 35565423 PMCID: PMC9104271 DOI: 10.3390/cancers14092294] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/01/2022] [Accepted: 05/02/2022] [Indexed: 12/17/2022] Open
Abstract
Background: Eosinophils appear to contribute to the efficacy of immunotherapy and their frequency was suggested as a predictive biomarker. Whether this observation could be transferred to patients treated with targeted therapy remains unknown. Methods: Blood and serum samples of healthy controls and 216 patients with advanced melanoma were prospectively and retrospectively collected. Freshly isolated eosinophils were phenotypically characterized by flow cytometry and co-cultured in vitro with melanoma cells to assess cytotoxicity. Soluble serum markers and peripheral blood counts were used for correlative studies. Results: Eosinophil-mediated cytotoxicity towards melanoma cells, as well as phenotypic characteristics, were similar when comparing healthy donors and patients. However, high relative pre-treatment eosinophil counts were significantly associated with response to MAPKi (p = 0.013). Eosinophil-mediated cytotoxicity towards melanoma cells is dose-dependent and requires proximity of eosinophils and their target in vitro. Treatment with targeted therapy in the presence of eosinophils results in an additive tumoricidal effect. Additionally, melanoma cells affected eosinophil phenotype upon co-culture. Conclusion: High pre-treatment eosinophil counts in advanced melanoma patients were associated with a significantly improved response to MAPKi. Functionally, eosinophils show potent cytotoxicity towards melanoma cells, which can be reinforced by MAPKi. Further studies are needed to unravel the molecular mechanisms of our observations.
Collapse
|
44
|
Gildner TE, Cepon-Robins TJ, Urlacher SS. Cumulative host energetic costs of soil-transmitted helminth infection. Trends Parasitol 2022; 38:629-641. [DOI: 10.1016/j.pt.2022.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/24/2022]
|
45
|
Nguyen J, Armstrong BS, Cowman S, Tomer Y, Veerabhadraiah SR, Beers MF, Venosa A. Immunophenotyping of Acute Inflammatory Exacerbations of Lung Injury Driven by Mutant Surfactant Protein-C: A Role for Inflammatory Eosinophils. Front Pharmacol 2022; 13:875887. [PMID: 35571100 PMCID: PMC9094740 DOI: 10.3389/fphar.2022.875887] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/23/2022] [Indexed: 12/24/2022] Open
Abstract
Acute inflammatory exacerbations (AIEs) represent immune-driven deteriorations of many chronic lung conditions, including COPD, asthma, and pulmonary fibrosis (PF). The first line of therapy is represented by broad-spectrum immunomodulation. Among the several inflammatory populations mobilizing during AIEs, eosinophils have been identified as promising indicators of an active inflammatory exacerbation. To better study the eosinophil-parenchymal crosstalk during AIE-PF, this work leverages a clinically relevant model of inflammatory exacerbations triggered by inducible expression of a mutation in the alveolar epithelial type 2 cell Surfactant Protein-C gene [SP-CI73T]. Unbiased single-cell sequencing analysis of controls and SP-CI73T mutants at a time coordinated with peak eosinophilia (14 days) defined heightened inflammatory activation, chemotaxis, and survival signaling (IL-6, IL-4/13, STAT3, Glucocorticoid Receptor, mTOR, and MYC) in eosinophils. To study the impact of eosinophils in inflammatory exacerbations, the SP-CI73T line was crossed with eosinophil lineage deficient mice (GATA1Δdbl) to produce the SP-CI73TGATA1KO line. Time course analysis (7-42 days) demonstrated improved lung histology, survival, and reduced inflammation in SP-CI73TGATA1KO cohorts. Spectral flow cytometry of tissue digests confirmed eosinophil depletion in GATA1KO mice and the absence of a compensatory shift in neutrophils and immature monocyte recruitment. Eosinophil deletion resulted in progressive monocyte-derived macrophage accumulation (14 days post-injury), combined with declines in CD3+CD4+ lymphocyte and B220+ B cell abundance. Histochemical analysis revealed atypical inflammatory cell activation in SP-CI73TGATA1KO mice, with reduced numbers of Arg-1+ and iNOS+ cells, but increases in tgfb1 mRNA expression in bronchoalveolar lavage cells and tissue. Dexamethasone treatment (1 mg/kg daily, i.p.) was utilized to investigate corticosteroid efficacy in highly eosinophilic exacerbations induced by mutant SP-CI73T. Dexamethasone successfully reduced total and eosinophil (CD11b+SigF+CD11c-) counts at 14 days and was linked to reduced evidence of structural damage and perivascular infiltrate. Together, these results illustrate the deleterious role of eosinophils in inflammatory events preceding lung fibrosis and demonstrate the efficacy of corticosteroid treatment in highly eosinophilic exacerbations induced by mutant SP-CI73T.
Collapse
Affiliation(s)
- Jacklyn Nguyen
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States
| | - Brittnie S. Armstrong
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States
| | - Sophie Cowman
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States
| | - Yaniv Tomer
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Michael F. Beers
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,PENN-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Alessandro Venosa
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States,*Correspondence: Alessandro Venosa,
| |
Collapse
|
46
|
Gaur P, Zaffran I, George T, Alekberli FR, Ben-Zimra M, Levi-Schaffer F. The regulatory role of eosinophils in viral, bacterial, and fungal infections. Clin Exp Immunol 2022; 209:72-82. [PMID: 35467728 PMCID: PMC9307229 DOI: 10.1093/cei/uxac038] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/15/2022] [Accepted: 04/24/2022] [Indexed: 12/14/2022] Open
Abstract
Eosinophils are innate immune cells typically associated with allergic and parasitic diseases. However, in recent years, eosinophils have also been ascribed a role in keeping homeostasis and in fighting several infectious diseases. Indeed, these cells circulate as mature cells in the blood and can be quickly recruited to the infected tissue. Moreover, eosinophils have all the necessary cellular equipment such as pattern recognition receptors (PRRs), pro-inflammatory cytokines, anti-bacterial proteins, and DNA traps to fight pathogens and promote an efficient immune response. This review summarizes some of the updated information on the role of eosinophils' direct and indirect mediated interactions with pathogens.
Collapse
Affiliation(s)
- Pratibha Gaur
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Ilan Zaffran
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Tresa George
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Fidan Rahimli Alekberli
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Micha Ben-Zimra
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| |
Collapse
|
47
|
Zhang X, Yang Y, Zheng Y, Hu Y, Rao Y, Li J, Zhao P, Li J. The Value of the Antibody Detection in the Diagnosis of Ocular Toxocariasis and the Aqueous Cytokine Profile Associated With the Condition. Front Med (Lausanne) 2022; 9:838800. [PMID: 35419376 PMCID: PMC9000971 DOI: 10.3389/fmed.2022.838800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction To evaluate and compare the specificity of Toxocara canis-specific antibody detection in the serum and aqueous samples for the diagnosis of ocular toxocariasis (OT) and explore the cytokine profiles associated with the condition in children. Materials and Methods This is a prospective cohort study. The inclusion criteria were the clinical presentations of OT, which included unilateral vision reduction, typical peripheral or posterior pole granuloma with variable degrees of vitritis, and exclusion of other diagnoses. The titer of antibody against the excretory-secretory antigen of Toxocara canis [T-immunoglobulin G (IgG)] was measured in serum and aqueous samples that were taken from the affected eyes. The diagnosis of OT was made upon positive detection of T-IgG either in the serum or aqueous. The rest with typical clinical presentations as described above but a positive serum or aqueous T-IgG could not be confirmed were diagnosed as suspected OT. Cytokines were measured using multiplexed cytometric bead array system. Results Two hundred and eleven eyes of 211 patients had participated in the study. One hundred and twenty-eight eyes were diagnosed as OT. The median age of the cohort was 7.7 years with a male to female ratio of 2.5:1. Major initial symptoms were decreased vision (74%) and strabismus (22%). The percentages of eyes with peripheral granuloma, posterior granuloma, and endophthalmitis were 40, 18, and 41%, respectively. Vitritis (100%), vitreous strands (64%), retinal fibrotic bands (57%), and retinal detachment (42%) were the most common signs. T-IgG was positive in 66.7% of the aqueous and 57.2% of the serum samples. Forty-four patients were diagnosed T-IgG negative in both serum and aqueous of the affected eyes. Interleukin (IL)-6, monocyte chemoattractant protein (MCP)-1, IL-8, eosinophil chemotactic protein (Eotaxin), MCP-1β, and vascular endothelial growth factor (VEGF) were higher in T-IgG negative eyes when compared to controls and further increased in T-IgG positive eyes. However, only T-IgG positive eyes showed increased IL-5, IL-13, and IL-10. IL-1β, tumor necrosis factor-alpha (TNF-α), IL-12, IL-2, interferon-gamma (IFN-γ), and IL-4 were undetectable in all eyes. Conclusions Pediatric OT is often present with severe retinal complications. Polarized intraocular Th2 response was only found in aqueous T-IgG positive eyes. Our results supported an aqueous sample-based antibody test for the more specific diagnosis of OT.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Yang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zheng
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiqian Hu
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuqing Rao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiakai Li
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Li
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Leal-Silva T, Lopes CDA, Vieira-Santos F, Oliveira FMS, Kraemer L, Padrão LDLS, Amorim CCO, Souza JLN, Barbosa FS, Rachid MA, Russo RC, Fujiwara RT, Bueno LL. Tissue eosinophilia correlates with mice susceptibility, granuloma formation, and damage during Toxocara canis infection. Parasitology 2022; 149:1-38. [PMID: 35139931 DOI: 10.1017/s0031182022000075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AbstractAn increase in peripheral blood eosinophils in helminth infections is expected, and these cells are known to promote immunity against these parasites. However, studies have suggested that in some specific helminths, eosinophils may promote the needs and longevity of these parasites, and their role in these infections remains undefined, including in Toxocara canis infection. Thus, this study aimed to investigate the role of eosinophils in the context of larval migration of T. canis and the immunopathological aspects of infection. For this, we used wild-type mice and mice genetically deficient for the transcription factor GATA-binding factor 1 (GATA1−/−), infected with 1000 eggs of T. canis. At 0, 3, 14 and 63 days post-infection, parasite load, tissue cytokine production, leucocyte profile, bronchoalveolar lavage cells and histopathological analyses were carried out. Collectively, our results demonstrate that the presence of eosinophils mediates susceptibility to T. canis, inducing leucocytosis and the formation of granulomas, increasing the pulmonary and cerebral parasite load, and reducing the number of neutrophils, which may be necessary to control the infection.
Collapse
Affiliation(s)
- Thaís Leal-Silva
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
- Post-graduation Program in Health Sciences: Infectious Diseases and Tropical Medicine, Faculdade de Medicina, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Camila de Almeida Lopes
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
| | - Flaviane Vieira-Santos
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
| | - Fabrício Marcus Silva Oliveira
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
| | - Lucas Kraemer
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
| | - Luiza de Lima Silva Padrão
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
- Post-graduation Program in Health Sciences: Infectious Diseases and Tropical Medicine, Faculdade de Medicina, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Chiara Cássia Oliveira Amorim
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
| | - Jorge Lucas Nascimento Souza
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
| | - Fernando Sérgio Barbosa
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
| | - Milene Alvarenga Rachid
- Laboratory of Protozooses, Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
- Post-graduation Program in Health Sciences: Infectious Diseases and Tropical Medicine, Faculdade de Medicina, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais,Belo Horizonte, Brazil
- Post-graduation Program in Health Sciences: Infectious Diseases and Tropical Medicine, Faculdade de Medicina, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
49
|
Shields VE, Cooper J. Use of helminth therapy for management of ulcerative colitis and Crohn's disease: a systematic review. Parasitology 2022; 149:145-154. [PMID: 34579797 PMCID: PMC10090591 DOI: 10.1017/s0031182021001670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 11/06/2022]
Abstract
The incidence rate of inflammatory bowel diseases is increasing in developed countries. As such there is an increasing demand for new therapies. The aim of this systematic review was to investigate whether there is evidence to support the use of helminth therapy for the management of Crohn's disease and ulcerative colitis. Four databases (PubMed, Embase, Medline and the Cochrane Central Register of Control Trials) were searched for primary evidence in the form of clinical studies. Nine studies were suitable for inclusion: five double-blind randomized control trials and four open-label studies. This review divided the results of the studies into two categories: (a) the efficacy of helminth therapy and (b) the safety of helminth therapy. Results regarding the efficacy were mixed and a conclusive answer could not be reached, as there was not enough evidence to rule out a placebo effect. More research is needed, particularly studies with control groups to address the possibility of a placebo effect. Despite this, all nine studies concluded helminth therapy was safe and tolerable, and therefore there is currently no evidence against further exploration of this treatment option.
Collapse
Affiliation(s)
| | - Jan Cooper
- Warwick Medical School, University of Warwick, CoventryCV4 7AL, UK
| |
Collapse
|
50
|
Protective response mediated by immunization with recombinant proteins in a murine model of toxocariasis and canine infection by Toxocara canis. Vaccine 2022; 40:912-923. [PMID: 35012775 DOI: 10.1016/j.vaccine.2021.12.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/06/2021] [Accepted: 12/20/2021] [Indexed: 11/21/2022]
Abstract
Toxocariasis is a neglected parasitic zoonosis of global importance. The development of a formulation that can be used as a vaccine would help the definitive control of the infection. Preclinical studies selected two recombinant T. canis proteins (rTcVcan and rTcCad) which significantly protected mice against larval migration. In the present work, these proteins plus three adjuvants (Alhydrogel®, PAM3CSK4®, and Quil-A®) were used to immunize mice against toxocariasis; blood samples were collected three times to measure IgG (total, IgG1, IgG2a), IgA, and IgE via indirect ELISA. Cytokines (IL-5, TNF-α, and IL-10) were measured in splenocytes supernatant, and T. canis larvae were quantified in tissues. The best protein + adjuvant pair found (rTVcan + QuialA®) was then used to immunize T. canis-free puppies (n = 18) that were experimentally infected with T. canis and T. canis naturally-infected puppies (n = 6). Immunoglobulin (IgA, IgE, IgG, IgG1, and IgG2a), parasite load (eggs in feces), number of expelled adults and eggs extracted from the female uterus, and their fertility percentages were analyzed. In mice, it was observed a highly significant reduction (73%) of tissue larvae, a mixed cytokine profile (Th1/Th2), and anti-T. canis antibody titers (IgG, IgG1, IgG2a) using rTVcan + QuialA® mix. In canines, rTVcan + QuialA® promoted reduction in the parasite eggs in feces (95%) and eggs reduction obtained from the uteri of pharmacologically expelled adult females (58.38%). In our knowledge this is the first canine clinical trial of a vaccine with T. canis recombinant proteins. The formulation used has been shown to efficiently stimulate the production of antibodies against infection by T. canis. In the canine, a significant reduction in the number of eggs expelled by the experimental animals that received the formulation prophylactically was evidenced. Future tests should be developed to evaluate the duration of the protective effect and analyze other immune pathways that could be stimulated by the formulation used.
Collapse
|