1
|
Li S, Liu M, Pan L, Feng Q, Lu X, Yao J, Xiao X. Integrated network pharmacology, mass cytometry and multi-omics analysis the effect of Jingfang granule on intestinal immune disorder in mice with cold-dampness syndrome. J Pharm Biomed Anal 2024; 255:116624. [PMID: 39657279 DOI: 10.1016/j.jpba.2024.116624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/11/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
The pathogenesis of cold-dampness syndrome (CDS) is closely related to intestinal inflammation and immune disorders induced by cold-dampness pathogen. CDS is the root cause of a variety of chronic inflammatory and immune diseases. Jingfang granule (JF) was widely used to treat a variety of diseases closely related to CDS. JF is well known for its clinical effect of dispelling cold and eliminating dampness, but the pharmacological effect and mechanism of JF on the improvement of CDS are still unclear. This study aimed to explore the efficacy and mechanism of JF in improving CDS from the perspective of intestinal immunity. In this study, mass spectrometry (CyTOF), metabolomics, network pharmacology, proteomics and molecular biology experiments were performed to investigate the therapeutic effects and underlying mechanisms of JF on intestinal inflammation and immune disorders in CDS mice. These results showed that JF could improve the clinical symptoms and increase the thymus index of CDS mice. Most strikingly, JF ameliorated intestinal inflammation and immune disorders in CDS mice, as indicated by increased frequency of TH1, CD8 + Tem, CD8 + TEFF, gdT and iNK cells and decreased frequency of Naive B cells, M1-macrophages, DCs and eosinophils. Metabolomics results showed that JF reversed the content of docosahexaenoic acid, arachidonic acid, linoleic acid, inosine and hypoxanthine in CDS mice. Correlation analysis showed that these metabolites were strongly correlated with a variety of intestinal immune cells, indicating that there was a certain regulatory effect between them. Then, 271 JF targets, 316 metabolite targets and 18374 disease targets were integrated to obtain 75 common targets and 138 pathways (such as PI3K/AKT and MAPK pathway, etc). Furthermore, molecular docking, proteomics and western blotting demonstrated that PI3K/AKT signaling pathway might be the key molecular mechanism by which JF regulated intestinal immune disorders in CDS mice. These results suggested that JF may act on the PI3K/AKT pathways to further regulate the levels of metabolites to exert intestinal immunomodulatory effects. In summary, we confirmed the beneficial effects of JF on intestinal immune disorders in CDS mice.
Collapse
Affiliation(s)
- Shirong Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Mingfei Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Lihong Pan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. LTD., Linyi 276005, China.
| | - Qun Feng
- Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xiaoyan Lu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. LTD., Linyi 276005, China.
| | - Jingchun Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. LTD., Linyi 276005, China.
| | - Xuefeng Xiao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
2
|
Liu Y, Wu F, Zhang M, Jin Y, Yuan X, Hao Y, Chen L, Fang B. 2'-Fucosyllactose and 3'-Sialyllactose Reduce Mortality in Neonatal Enteroaggregative Escherichia coli Infection by Improving the Construction of Intestinal Mucosal Immunity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:26165-26177. [PMID: 39535070 DOI: 10.1021/acs.jafc.4c06524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Human milk oligosaccharides could prevent pathogenic bacterial infections in neonates; however, direct in vivo anti-infection evidence was still lacking. Here, we systematically evaluated the effects of 2'-fucosyllactose (2'-FL) and 3'-sialyllactose (3'-SL) on the structural development and functional maturation in neonates and their defense against enteroaggregative Escherichia coli infection. It was found that supplementation with 2'-FL and 3'-SL improved the resistance of weaned mice to enteroaggregative E. coli. The mechanism related to the promotion of 2'-FL and 3'-SL in the maturation of intestinal mucosal immunity by promoting stem cell differentiation, mucus layer integrity, and tight junction formation. 2'-FL and 3'-SL significantly increased the ratio of Th1 and Treg cells in the lamina propria, contents of short-chain fatty acids, as well as the serum content of IgA. This study lays a theoretical basis for the application of 2'-FL and 3'-SL in infant formula, as well as the development of intestinal health products.
Collapse
Affiliation(s)
- Yaqiong Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Fang Wu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Ming Zhang
- School of Food Science and Chemical Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Yutong Jin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Xinlei Yuan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
- College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300450, China
| | - Yanling Hao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Lishui Chen
- Food Laboratory of Zhongyuan, Luohe, Henan 462300, China
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| |
Collapse
|
3
|
Balendran T, Iddawela D, Lenadora S. Cryptosporidiosis in a Zoonotic Gastrointestinal Disorder Perspective: Present Status, Risk Factors, Pathophysiology, and Treatment, Particularly in Immunocompromised Patients. J Trop Med 2024; 2024:6439375. [PMID: 39534184 PMCID: PMC11557182 DOI: 10.1155/2024/6439375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 07/07/2024] [Accepted: 10/05/2024] [Indexed: 11/16/2024] Open
Abstract
Cryptosporidium infection is highly prevalent among immunocompromised patients with Acquired Immunodeficiency Syndrome, cancer, primary immunodeficiency, and organ transplant recipients. Comprehensive knowledge about Cryptosporidium infection provides the means for efficient diagnosis, treatment, and prevention. Therefore, with the objective of providing an in-depth analysis of Cryptosporidiosis in immunocompromised patients, this review presents a comprehensive understating of the prevalence, risk factors, pathophysiology of Cryptosporidium infection, clinical presentation in the immunocompromised, the immune response of the host, diagnostic methods performed in laboratory settings, possible treatments, and prevention methods, which can be used for further studies. Peer-reviewed, published, original articles on cryptosporidiosis in immunocompromised patients were searched using specific key-words on PubMed, ResearchGate, Google Scholar, and ScienceDirect databases. Articles which were accessible to the date of 18th of August 2023, were included in this comprehensive review. We analyzed reports on Cryptosporidium in immunocompromised patients with human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS), cancer, primary immunodeficiency, and organ transplant recipients. 134 Articles describing epidemiology, related risk factors, clinical presentation, diagnosis, and possible treatments in the light of pathogenesis, pathophysiology, and virulence factors of Cryptosporidium and immunology of the host are summarized in this study. Effective treatments to be administered, importance, and ways of prevention were identified. Cryptosporidium infection was found to be highly prevalent among immunocompromised in Asia, Africa, Europe, and North America. The immunity of the host and the decrease in CD4+ T-cell count were found to the main factors which decide the susceptibility and the severity of infection. Drugs that activate host immunity and suppress Cryptosporidium growth, along with supportive therapy, is an effective treatment. But prevention is the most effective strategy for immunocompromised patients; thus, a better understanding about the disease would lead to effective prevention.
Collapse
Affiliation(s)
- Thivya Balendran
- Department of Parasitology, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Devika Iddawela
- Department of Parasitology, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Sajanee Lenadora
- Department of Parasitology, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka
| |
Collapse
|
4
|
Partida-Rodríguez O, Brown EM, Woodward SE, Cirstea M, Reynolds LA, Petersen C, Vogt SL, Peña-Díaz J, Thorson L, Arrieta MC, Hernández EG, Rojas-Velázquez L, Moran P, González Rivas E, Serrano-Vázquez A, Pérez-Juárez H, Torres J, Ximénez C, Finlay BB. Fecal microbiota transplantation from protozoa-exposed donors downregulates immune response in a germ-free mouse model, its role in immune response and physiology of the intestine. PLoS One 2024; 19:e0312775. [PMID: 39466773 PMCID: PMC11515975 DOI: 10.1371/journal.pone.0312775] [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: 03/26/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024] Open
Abstract
Intestinal parasites are part of the intestinal ecosystem and have been shown to establish close interactions with the intestinal microbiota. However, little is known about the influence of intestinal protozoa on the regulation of the immune response. In this study, we analyzed the regulation of the immune response of germ-free mice transplanted with fecal microbiota (FMT) from individuals with multiple parasitic protozoans (P) and non-parasitized individuals (NP). We determined the production of intestinal cytokines, the lymphocyte populations in both the colon and the spleen, and the genetic expression of markers of intestinal epithelial integrity. We observed a general downregulation of the intestinal immune response in mice receiving FMT-P. We found significantly lower intestinal production of the cytokines IL-6, TNF, IFN-γ, MCP-1, IL-10, and IL-12 in the FMT-P. Furthermore, a significant decrease in the proportion of CD3+, CD4+, and Foxp3+ T regulatory cells (Treg) was observed in both, the colon and spleen with FMT-P in contrast to FMT-NP. We also found that in FMT-P mice there was a significant decrease in tjp1 expression in all three regions of the small intestine; ocln in the ileum; reg3γ in the duodenum and relmβ in both the duodenum and ileum. We also found an increase in colonic mucus layer thickness in mice colonized with FMT-P in contrast with FMT-NP. Finally, our results suggest that gut protozoa, such as Blastocystis hominis, Entamoeba coli, Endolimax nana, Entamoeba histolytica/E. dispar, Iodamoeba bütschlii, and Chilomastix mesnili consortia affect the immunoinflammatory state and induce functional changes in the intestine via the gut microbiota. Likewise, it allows us to establish an FMT model in germ-free mice as a viable alternative to explore the effects that exposure to intestinal parasites could have on the immune response in humans.
Collapse
Affiliation(s)
- Oswaldo Partida-Rodríguez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Eric M. Brown
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Sarah E. Woodward
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Mihai Cirstea
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Lisa A. Reynolds
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Microbiology, Faculty of Science, University of Victoria, Victoria, Canada
| | - Charisse Petersen
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Stefanie L. Vogt
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Jorge Peña-Díaz
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Lisa Thorson
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Marie-Claire Arrieta
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Eric G. Hernández
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Liliana Rojas-Velázquez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Patricia Moran
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Enrique González Rivas
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Angélica Serrano-Vázquez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Horacio Pérez-Juárez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Javier Torres
- Unidad de Investigación en Enfermedades Infecciosas y Parasitarias, Instituto Mexicano del Seguro Social (IMSS), Mexico, Mexico
| | - Cecilia Ximénez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - B. B. Finlay
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
5
|
Yu X, Yang Y, Zhu W, Liu M, Wu J, Singer SM, Li W. The pathogenic responses elicited during exposure of human intestinal cell line with Giardia duodenalis excretory-secretory products and the potential attributed endocytosis mechanism. Med Microbiol Immunol 2024; 213:23. [PMID: 39441372 DOI: 10.1007/s00430-024-00806-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
Giardia duodenalis, an important zoonotic protozoan parasite, adheres to host intestinal epithelial cells (IECs) via the ventral disc and causes giardiasis characterized mainly by diarrhea. To date, it remains elusive how excretory-secretory products (ESPs) of Giardia enter IECs and how the cells respond to the entry. Herein, we initially demonstrated that ESPs evoked IEC endocytosis in vitro. We indicated that ESPs contributed vitally in triggering intrinsic apoptosis, pro-inflammatory responses, tight junction (TJ) protein expressional changes, and autophagy in IECs. Endocytosis was further proven to be implicated in those ESPs-triggered IEC responses. Ten predicted virulent excretory-secretory proteins of G. duodenalis were investigated for their capability to activate clathrin/caveolin-mediated endocytosis (CME/CavME) in IECs. Pyridoxamine 5'-phosphate oxidase (PNPO) was confirmed to be an important contributor. PNPO was subsequently verified as a vital promoter in the induction of giardiasis-related IEC apoptosis, inflammation, and TJ protein downregulation. Most importantly, this process seemed to be involved majorly in PNPO-evoked CME pathway, rather than CavME. Collectively, this study identified Giardia ESPs, notably PNPO, as potentially important pathogenic factors during noninvasive infection. It was also noteworthy that ESPs-evoked endocytosis might play a role in triggering giardiasis-inducing cellular regulation. These findings would deepen our understanding about the role of ESPs, notably PNPO, in the pathogenesis of giardiasis and the potential attributed endocytosis mechanism.
Collapse
Affiliation(s)
- Xiran Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yongwu Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Weining Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Min Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Jingxue Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Steven M Singer
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Wei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China.
| |
Collapse
|
6
|
Guo Y, Ren C, He Y, Wu Y, Yang X. Deciphering the spatiotemporal transcriptional landscape of intestinal diseases (Review). Mol Med Rep 2024; 30:157. [PMID: 38994768 PMCID: PMC11258600 DOI: 10.3892/mmr.2024.13281] [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: 11/21/2023] [Accepted: 04/19/2024] [Indexed: 07/13/2024] Open
Abstract
The intestines are the largest barrier organ in the human body. The intestinal barrier plays a crucial role in maintaining the balance of the intestinal environment and protecting the intestines from harmful bacterial invasion. Single‑cell RNA sequencing technology allows the detection of the different cell types in the intestine in two dimensions and the exploration of cell types that have not been fully characterized. The intestinal mucosa is highly complex in structure, and its proper functioning is linked to multiple structures in the proximal‑distal intestinal and luminal‑mucosal axes. Spatial localization is at the core of the efforts to explore the interactions between the complex structures. Spatial transcriptomics (ST) is a method that allows for comprehensive tissue analysis and the acquisition of spatially separated genetic information from individual cells, while preserving their spatial location and interactions. This approach also prevents the loss of fragile cells during tissue disaggregation. The emergence of ST technology allows us to spatially dissect enzymatic processes and interactions between multiple cells, genes, proteins and signals in the intestine. This includes the exchange of oxygen and nutrients in the intestine, different gradients of microbial populations and the role of extracellular matrix proteins. This regionally precise approach to tissue studies is gaining more acceptance and is increasingly applied in the investigation of disease mechanisms related to the gastrointestinal tract. Therefore, this review summarized the application of ST in gastrointestinal diseases.
Collapse
Affiliation(s)
- Yajing Guo
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Chao Ren
- Graduate School, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yuxi He
- Department of Digestive Medicine, Chongqing City Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| | - Yue Wu
- Department of Digestive Medicine, Chongqing City Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| | - Xiaojun Yang
- Department of Digestive Medicine, Chongqing City Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| |
Collapse
|
7
|
Hao R, Zhao M, Tayyab M, Lin Z, Zhang Y. The mucosal immunity in crustaceans: Inferences from other species. FISH & SHELLFISH IMMUNOLOGY 2024; 152:109785. [PMID: 39053584 DOI: 10.1016/j.fsi.2024.109785] [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: 04/16/2024] [Revised: 07/10/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024]
Abstract
Crustaceans such as shrimps and crabs, hold significant ecological significance and substantial economic value within marine ecosystems. However, their susceptibility to disease outbreaks and pathogenic infections has posed major challenges to production in recent decades. As invertebrate, crustaceans primarily rely on their innate immune system for defense, lacking the adaptive immune system found in vertebrates. Mucosal immunity, acting as the frontline defense against a myriad of pathogenic microorganisms, is a crucial aspect of their immune repertoire. This review synthesizes insights from comparative immunology, highlighting parallels between mucosal immunity in vertebrates and innate immune mechanisms in invertebrates. Despite lacking classical adaptive immunity, invertebrates, including crustaceans, exhibit immune memory and rely on inherent "innate immunity factors" to combat invading pathogens. Drawing on parallels from mammalian and piscine systems, this paper meticulously explores the complex role of mucosal immunity in regulating immune responses in crustaceans. Through the extrapolation from well-studied models like mammals and fish, this review infers the potential mechanisms of mucosal immunity in crustaceans and provides insights for research on mucosal immunity in crustaceans.
Collapse
Affiliation(s)
- Ruixue Hao
- Guangdong Provincial Key Laboratory of Marine Biology and Department of Biology, Shantou University, Shantou, 515063, China
| | - Mingming Zhao
- Guangdong Provincial Key Laboratory of Marine Biology and Department of Biology, Shantou University, Shantou, 515063, China
| | - Muhammad Tayyab
- Guangdong Provincial Key Laboratory of Marine Biology and Department of Biology, Shantou University, Shantou, 515063, China
| | - Zhongyang Lin
- Guangdong Provincial Key Laboratory of Marine Biology and Department of Biology, Shantou University, Shantou, 515063, China.
| | - Yueling Zhang
- Guangdong Provincial Key Laboratory of Marine Biology and Department of Biology, Shantou University, Shantou, 515063, China.
| |
Collapse
|
8
|
Enriquez GF, Macchiaverna NP, Garbossa G, Quebrada Palacio LP, Ojeda BL, Bua J, Gaspe MS, Cimino R, Gürtler RE, Postan M, Cardinal MV. Humans seropositive for Trypanosoma cruzi co-infected with intestinal helminths have higher infectiousness, parasitaemia and Th2-type response in the Argentine Chaco. Parasit Vectors 2024; 17:340. [PMID: 39135121 PMCID: PMC11320973 DOI: 10.1186/s13071-024-06401-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND The Gran Chaco ecoregion is a well-known hotspot of several neglected tropical diseases (NTDs) including Chagas disease, soil-transmitted helminthiasis and multiparasitic infections. Interspecific interactions between parasite species can modify host susceptibility, pathogenesis and transmissibility through immunomodulation. Our objective was to test the association between human co-infection with intestinal parasites and host parasitaemia, infectiousness to the vector and immunological profiles in Trypanosoma cruzi-seropositive individuals residing in an endemic region of the Argentine Chaco. METHODS We conducted a cross-sectional serological survey for T. cruzi infection along with an intestinal parasite survey in two adjacent rural villages. Each participant was tested for T. cruzi and Strongyloides stercoralis infection by serodiagnosis, and by coprological tests for intestinal parasite detection. Trypanosoma cruzi bloodstream parasite load was determined by quantitative PCR (qPCR), host infectiousness by artificial xenodiagnosis and serum human cytokine levels by flow cytometry. RESULTS The seroprevalence for T. cruzi was 16.1% and for S. stercoralis 11.5% (n = 87). We found 25.3% of patients with Enterobius vermicularis. The most frequent protozoan parasites were Blastocystis spp. (39.1%), Giardia lamblia (6.9%) and Cryptosporidium spp. (3.4%). Multiparasitism occurred in 36.8% of the examined patients. Co-infection ranged from 6.9% to 8.1% for T. cruzi-seropositive humans simultaneously infected with at least one protozoan or helminth species, respectively. The relative odds of being positive by qPCR or xenodiagnosis (i.e. infectious) of 28 T. cruzi-seropositive patients was eight times higher in people co-infected with at least one helminth species than in patients with no such co-infection. Trypanosoma cruzi parasite load and host infectiousness were positively associated with helminth co-infection in a multiple regression analysis. Interferon-gamma (IFN-γ) response, measured in relation to interleukin (IL)-4 among humans infected with T. cruzi only, was 1.5-fold higher than for T. cruzi-seropositive patients co-infected with helminths. The median concentration of IL-4 was significantly higher in T. cruzi-seropositive patients with a positive qPCR test than in qPCR-negative patients. CONCLUSIONS Our results show a high level of multiparasitism and suggest that co-infection with intestinal helminths increased T. cruzi parasitaemia and upregulated the Th2-type response in the study patients.
Collapse
Affiliation(s)
- Gustavo Fabián Enriquez
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina.
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Natalia Paula Macchiaverna
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Graciela Garbossa
- Laboratorio de Parasitología Clínica y Ambiental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, IQUIBICEN-CONICET-UBA), Instituto de Investigaciones en Salud Pública, Buenos Aires, Argentina
| | - Luz Piedad Quebrada Palacio
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Institute of Virology, Helmholtz Centre Munich, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Bárbara Leonor Ojeda
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
| | - Jacqueline Bua
- Instituto Nacional de Parasitología Dr. M. Fatala Chabén, Administración Nacional de Laboratorios e Institutos de Salud Dr. C.G. Malbrán, Buenos Aires, Argentina
| | - María Sol Gaspe
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rubén Cimino
- Instituto de Investigaciones de Enfermedades Tropicales (IIET). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-CCT Salta, Universidad Nacional de Salta, Sede Regional Orán, Salta, Argentina
- Facultad de Ciencias Naturales, Cátedra de Química Biológica, Universidad Nacional de Salta, Salta, Argentina
| | - Ricardo Esteban Gürtler
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Miriam Postan
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - Marta Victoria Cardinal
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
9
|
Delgadinho M, Ginete C, Santos B, de Vasconcelos JN, Arez AP, Brito M. Sickle Cell Disease and Gut Health: The Influence of Intestinal Parasites and the Microbiome on Angolan Children. Int J Mol Sci 2024; 25:7258. [PMID: 39000364 PMCID: PMC11242675 DOI: 10.3390/ijms25137258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/16/2024] Open
Abstract
Parasitic infections are a common problem in developing countries and can intensify morbidity in patients with sickle cell disease (SCD), increasing the severity of anemia and the need for transfusions. It has been demonstrated that both helminths and protozoa can affect gut microbiome composition. On the other hand, the presence of specific bacterial communities can also influence parasite establishment. Considering this, our aim was to associate the presence of intestinal parasites with the results of hematological analyses and microbiome composition evaluations in a population of Angolan children with and without SCD. A total of 113 stool samples were collected, and gut microbiome analysis was performed using 16S sequencing and real-time PCR to detect eight different intestinal parasites. In our population, more than half of children (55%) had at least one parasitic infection, and of these, 43% were co-infected. Giardia intestinalis and Ascaris lumbricoides were more frequently found in children from the rural area of Bengo. Moreover, SCD children with ascariasis exhibited higher values of leukocytes and neutrophils, whereas the total hemoglobin levels were lower. In regards to the gut microbiome, the presence of intestinal parasites lowered the prevalence of some beneficial bacteria, namely: Lactobacillus, Bifidobacterium, Cuneatibacter, Bacteroides uniformis, Roseburia, and Shuttleworthia. This study presents the prevalence of several intestinal parasites in a high-risk transmission area with scarce information and opens new perspectives for understanding the interaction between parasites, the microbiome, and SCD.
Collapse
Affiliation(s)
- Mariana Delgadinho
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal
| | - Catarina Ginete
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal
| | - Brígida Santos
- Centro de Investigação em Saúde de Angola (CISA), Caxito, Angola
- Instituto Hematológico Pediátrico, Luanda, Angola
| | | | - Ana Paula Arez
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation towards Global Health, (LA-REAL), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), 1099-085 Lisbon, Portugal
| | - Miguel Brito
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal
- Centro de Investigação em Saúde de Angola (CISA), Caxito, Angola
| |
Collapse
|
10
|
Hunter-Barnett S, Viney M. Gut protozoa of wild rodents - a meta-analysis. Parasitology 2024; 151:594-605. [PMID: 38714350 PMCID: PMC11427965 DOI: 10.1017/s0031182024000556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/09/2024]
Abstract
Protozoa are well-known inhabitants of the mammalian gut and so of the gut microbiome. While there has been extensive study of a number of species of gut protozoa in laboratory animals, particularly rodents, the biology of the gut protozoa of wild rodents is much less well-known. Here we have systematically searched the published literature to describe the gut protozoa of wild rodents, in total finding records of 44 genera of protozoa infecting 228 rodent host species. We then undertook meta-analyses that estimated the overall prevalence of gut protozoa in wild rodents to be 24%, with significant variation in prevalence among some host species. We investigated how host traits may affect protozoa prevalence, finding that for some host lifestyles some protozoa differed in their prevalence. This synthesis of existing data on wild rodent gut protozoa provides a better understanding of the biology of these common gut inhabitants and suggests directions for their future study.
Collapse
Affiliation(s)
- Simon Hunter-Barnett
- Department of Evolution, Ecology and Behaviour, University of Liverpool, Liverpool L69 7ZB, UK
| | - Mark Viney
- Department of Evolution, Ecology and Behaviour, University of Liverpool, Liverpool L69 7ZB, UK
| |
Collapse
|
11
|
Shukla R, Soni J, Kumar A, Pandey R. Uncovering the diversity of pathogenic invaders: insights into protozoa, fungi, and worm infections. Front Microbiol 2024; 15:1374438. [PMID: 38596382 PMCID: PMC11003270 DOI: 10.3389/fmicb.2024.1374438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
Post COVID-19, there has been renewed interest in understanding the pathogens challenging the human health and evaluate our preparedness towards dealing with health challenges in future. In this endeavour, it is not only the bacteria and the viruses, but a greater community of pathogens. Such pathogenic microorganisms, include protozoa, fungi and worms, which establish a distinct variety of disease-causing agents with the capability to impact the host's well-being as well as the equity of ecosystem. This review summarises the peculiar characteristics and pathogenic mechanisms utilized by these disease-causing organisms. It features their role in causing infection in the concerned host and emphasizes the need for further research. Understanding the layers of pathogenesis encompassing the concerned infectious microbes will help expand targeted inferences with relation to the cause of the infection. This would strengthen and augment benefit to the host's health along with the maintenance of ecosystem network, exhibiting host-pathogen interaction cycle. This would be key to discover the layers underlying differential disease severities in response to similar/same pathogen infection.
Collapse
Affiliation(s)
- Richa Shukla
- Division of Immunology and Infectious Disease Biology, INGEN-HOPE (INtegrative GENomics of HOst-PathogEn) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Jyoti Soni
- Division of Immunology and Infectious Disease Biology, INGEN-HOPE (INtegrative GENomics of HOst-PathogEn) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ashish Kumar
- Division of Immunology and Infectious Disease Biology, INGEN-HOPE (INtegrative GENomics of HOst-PathogEn) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Rajesh Pandey
- Division of Immunology and Infectious Disease Biology, INGEN-HOPE (INtegrative GENomics of HOst-PathogEn) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
12
|
Perera DJ, Koger-Pease C, Paulini K, Daoudi M, Ndao M. Beyond schistosomiasis: unraveling co-infections and altered immunity. Clin Microbiol Rev 2024; 37:e0009823. [PMID: 38319102 PMCID: PMC10938899 DOI: 10.1128/cmr.00098-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024] Open
Abstract
Schistosomiasis is a neglected tropical disease caused by the helminth Schistosoma spp. and has the second highest global impact of all parasites. Schistosoma are transmitted through contact with contaminated fresh water predominantly in Africa, Asia, the Middle East, and South America. Due to the widespread prevalence of Schistosoma, co-infection with other infectious agents is common but often poorly described. Herein, we review recent literature describing the impact of Schistosoma co-infection between species and Schistosoma co-infection with blood-borne protozoa, soil-transmitted helminths, various intestinal protozoa, Mycobacterium, Salmonella, various urinary tract infection-causing agents, and viral pathogens. In each case, disease severity and, of particular interest, the immune landscape, are altered as a consequence of co-infection. Understanding the impact of schistosomiasis co-infections will be important when considering treatment strategies and vaccine development moving forward.
Collapse
Affiliation(s)
- Dilhan J. Perera
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Cal Koger-Pease
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Kayla Paulini
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Mohamed Daoudi
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Momar Ndao
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
13
|
Sardinha-Silva A, Gazzinelli-Guimaraes PH, Ajakaye OG, Ferreira TR, Alves-Ferreira EV, Tjhin ET, Gregg B, Fink MY, Coelho CH, Singer SM, Grigg ME. Giardia intestinalis reshapes mucosal immunity toward a Type 2 response that attenuates inflammatory bowel-like diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.02.583119. [PMID: 38903060 PMCID: PMC11188066 DOI: 10.1101/2024.03.02.583119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Diarrheal diseases are the second leading cause of death in children worldwide. Epidemiological studies show that co-infection with Giardia intestinalis decreases the severity of diarrhea. Here, we show that Giardia is highly prevalent in the stools of asymptomatic school-aged children. It orchestrates a Th2 mucosal immune response, characterized by increased antigen-specific Th2 cells, IL-25, Type 2-associated cytokines, and goblet cell hyperplasia. Giardia infection expanded IL-10-producing Th2 and GATA3+ Treg cells that promoted chronic carriage, parasite transmission, and conferred protection against Toxoplasma gondii-induced lethal ileitis and DSS-driven colitis by downregulating proinflammatory cytokines, decreasing Th1/Th17 cell frequency, and preventing collateral tissue damage. Protection was dependent on STAT6 signaling, as Giardia-infected STAT6-/- mice no longer regulated intestinal bystander inflammation. Our findings demonstrate that Giardia infection reshapes mucosal immunity toward a Type 2 response, which confers a mutualistic protection against inflammatory disease processes and identifies a critical role for protists in regulating mucosal defenses.
Collapse
Affiliation(s)
- Aline Sardinha-Silva
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pedro H. Gazzinelli-Guimaraes
- Helminth Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Oluwaremilekun G. Ajakaye
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Animal and Environmental Biology, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Tiago R. Ferreira
- Intracellular Parasite Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eliza V.C. Alves-Ferreira
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Erick T. Tjhin
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beth Gregg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marc Y. Fink
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Camila H. Coelho
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Steven M. Singer
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Michael E. Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
14
|
Popovic A, Cao EY, Han J, Nursimulu N, Alves-Ferreira EVC, Burrows K, Kennard A, Alsmadi N, Grigg ME, Mortha A, Parkinson J. Commensal protist Tritrichomonas musculus exhibits a dynamic life cycle that induces extensive remodeling of the gut microbiota. THE ISME JOURNAL 2024; 18:wrae023. [PMID: 38366179 PMCID: PMC10944700 DOI: 10.1093/ismejo/wrae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Commensal protists and gut bacterial communities exhibit complex relationships, mediated at least in part through host immunity. To improve our understanding of this tripartite interplay, we investigated community and functional dynamics between the murine protist Tritrichomonas musculus and intestinal bacteria in healthy and B-cell-deficient mice. We identified dramatic, protist-driven remodeling of resident microbiome growth and activities, in parallel with Tritrichomonas musculus functional changes, which were accelerated in the absence of B cells. Metatranscriptomic data revealed nutrient-based competition between bacteria and the protist. Single-cell transcriptomics identified distinct Tritrichomonas musculus life stages, providing new evidence for trichomonad sexual replication and the formation of pseudocysts. Unique cell states were validated in situ through microscopy and flow cytometry. Our results reveal complex microbial dynamics during the establishment of a commensal protist in the gut, and provide valuable data sets to drive future mechanistic studies.
Collapse
Affiliation(s)
- Ana Popovic
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Eric Y Cao
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Joanna Han
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Nirvana Nursimulu
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, M5G 0A4, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
| | - Eliza V C Alves-Ferreira
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD 20892, United States
| | - Kyle Burrows
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Andrea Kennard
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD 20892, United States
| | - Noor Alsmadi
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Michael E Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD 20892, United States
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - John Parkinson
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| |
Collapse
|
15
|
Allam NAT, Hamouda RAEF, Sedky D, Abdelsalam ME, El-Gawad MEHA, Hassan NMF, Aboelsoued D, Elmaaty AMA, Ibrahim MA, Taie HAA, Hakim AS, Desouky HM, Megeed KNA, Abdel-Hamid MS. Medical prospects of cryptosporidiosis in vivo control using biofabricated nanoparticles loaded with Cinnamomum camphora extracts by Ulva fasciata. Vet World 2024; 17:108-124. [PMID: 38406364 PMCID: PMC10884584 DOI: 10.14202/vetworld.2024.108-124] [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: 08/13/2023] [Accepted: 11/28/2023] [Indexed: 02/27/2024] Open
Abstract
Background and Aim Global efforts are continuing to develop preparations against cryptosporidiosis. This study aimed to investigate the efficacy of biosynthesized Ulva fasciata loading Cinnamomum camphora oil extract on new zinc oxide nanoparticles (ZnONPs shorten to ZnNPs) and silver nanoparticles (AgNPs) as alternative treatments for Cryptosporidium parvum experimental infection in rats. Materials and Methods Oil extract was characterized by gas chromatography-mass spectrometry, loaded by U. fasciata on ionic-based ZnO and NPs, and then characterized by transmission electron microscopy, scanning electron microscopy, and X-ray diffraction. Biosafety and toxicity were investigated by skin tests. A total of 105 C. parvum oocysts/rat were used (n = 81, 2-3 W, 80-120 g, 9 male rats/group). Oocysts shedding was counted for 21 d. Doses of each preparation in addition to reference drug were administered daily for 7 d, starting on post-infection (PI) day (3). Nitazoxanide (100 mg) was used as the reference drug. After 3 weeks, the rats were sacrificed for postmortem examination and histopathological examination. Two blood samples/rat/group were collected on the 21st day. Ethylenediaminetetraacetic acid blood samples were also used for analysis of biochemistry, hematology, immunology, micronucleus prevalence, and chromosomal abnormalities. Results C. camphora leaves yielded 28.5 ± 0.3 g/kg oil and 20 phycocompounds were identified. Spherical and rod-shaped particles were detected at 10.47-30.98 nm and 18.83-38.39 nm, respectively. ZnNPs showed the earliest anti-cryptosporidiosis effect during 7-17 d PI. Other hematological, biochemical, immunological, histological, and genotoxicity parameters were significantly fruitful; hence, normalized pathological changes induced by infestation were observed in the NPs treatments groups against the infestation-free and Nitazoxanide treated group. Conclusion C. camphora, U. fasciata, ZnNPs, and AgNPs have refluxed the pathological effects of infection as well as positively improved host physiological condition by its anticryptosporidial immunostimulant regenerative effects with sufficient ecofriendly properties to be proposed as an alternative to traditional drugs, especially in individuals with medical reactions against chemical commercial drugs.
Collapse
Affiliation(s)
- Nesreen Allam Tantawy Allam
- Department of Parasitology and Animal Diseases, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Giza, Cairo, Egypt
| | - Ragaa Abd El-Fatah Hamouda
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, 5 Zone, Sadat City, Munofia, Egypt
| | - Doaa Sedky
- Department of Parasitology and Animal Diseases, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Giza, Cairo, Egypt
| | - Mahinour Ezzeldin Abdelsalam
- Department of General Biology, Center of Basic Sciences, Misr University for Science and Technology, Al Motamayez District, 6 of October, Giza, Cairo, Egypt
| | | | - Noha Mahmoud Fahmy Hassan
- Department of Parasitology and Animal Diseases, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Giza, Cairo, Egypt
| | - Dina Aboelsoued
- Department of Parasitology and Animal Diseases, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Giza, Cairo, Egypt
| | - Amal M. Abou Elmaaty
- Department of Animal Reproduction and Artificial Insemination, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Giza, Cairo, Egypt
| | - Muhammad A. Ibrahim
- Cytogenetics and Animal Cell Culture Lab., National Gene Bank, Agriculture Research Center, 9 Gamaa Street, Giza, Cairo, Egypt
| | - Hanan Anwar Aly Taie
- Department of Plant Biochemistry, Agriculture and Biological Researches Institute, National Research Centre, 33 El-Bohouth St. (Former El-Tahrir St.), Dokki, P.O. 12622, Giza, Cairo, Egypt
| | - Ashraf Samir Hakim
- Department of Microbiology and Immunology, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Giza, Cairo, Egypt
| | - Hassan Mohamed Desouky
- Department of Animal Reproduction and Artificial Insemination, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Giza, Cairo, Egypt
| | - Kadria Nasr Abdel Megeed
- Department of Parasitology and Animal Diseases, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Giza, Cairo, Egypt
| | - Marwa Salah Abdel-Hamid
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, 5 Zone, Sadat City, Munofia, Egypt
| |
Collapse
|
16
|
Kim CW, Joo SY, Kim B, Kim JY, Jang S, Tzeng SJ, Lee SJ, Kim M, Kim I. Single cell transcriptome analyses reveal the roles of B cells in fructose-induced hypertension. Front Immunol 2023; 14:1279439. [PMID: 38045685 PMCID: PMC10691591 DOI: 10.3389/fimmu.2023.1279439] [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: 08/18/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
Rationale While the immune system plays a crucial role in the development of hypertension, the specific contributions of distinct immune cell populations remain incompletely understood. The emergence of single-cell RNA-sequencing (scRNA-seq) technology enables us to analyze the transcriptomes of individual immune cells and to assess the significance of each immune cell type in hypertension development. Objective We aimed to investigate the hypothesis that B cells play a crucial role in the development of fructose-induced hypertension. Methods and Results Eight-week-old Dahl salt-sensitive (SS) male rats were divided into two groups and given either tap water (TW) or a 20% fructose solution (HFS) for 4 weeks. Systolic blood pressure was measured using the tail-cuff method. ScRNA-seq analysis was performed on lamina propria cells (LPs) and peripheral blood mononuclear cells (PBMCs) obtained from SS rats subjected to either TW or HFS. The HFS treatment induced hypertension in the SS rats. The analysis revealed 27 clusters in LPs and 28 clusters in PBMCs, allowing for the identification and characterization of various immune cell types within each cluster. Specifically, B cells and follicular helper T (Tfh) cells were prominent in LPs, while B cells and M1 macrophages dominated PBMCs in the HFS group. Moreover, the HFS treatment triggered an increase in the number of B cells in both LPs and PBMCs, accompanied by activation of the interferon pathway. Conclusions The significant involvement of B cells in intestinal and PBMC responses indicates their pivotal contribution to the development of hypertension. This finding suggests that targeting B cells could be a potential strategy to mitigate high blood pressure in fructose-induced hypertension. Moreover, the simultaneous increase in follicular B cells and Tfh cells in LPs, along with the upregulation of interferon pathway genes in B cells, underscores a potential autoimmune factor contributing to the pathogenesis of fructose-induced hypertension in the intestine.
Collapse
Affiliation(s)
- Cheong-Wun Kim
- Department of Pharmacology, BK21 Plus Kyungpook National University (KNU) Biomedical Convergence Program, Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sung Yong Joo
- Department of Animal Science, Pusan National University, Miryang, Republic of Korea
| | - Boa Kim
- Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jee Young Kim
- Department of Pharmacology, BK21 Plus Kyungpook National University (KNU) Biomedical Convergence Program, Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sungmin Jang
- Department of Pharmacology, BK21 Plus Kyungpook National University (KNU) Biomedical Convergence Program, Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Shiang-Jong Tzeng
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Sang Jin Lee
- Division of Rheumatology, Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Myunghoo Kim
- Department of Animal Science, Pusan National University, Miryang, Republic of Korea
| | - Inkyeom Kim
- Department of Pharmacology, BK21 Plus Kyungpook National University (KNU) Biomedical Convergence Program, Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
17
|
Popovic A, Cao EY, Han J, Nursimulu N, Alves-Ferreira EVC, Burrows K, Kennard A, Alsmadi N, Grigg ME, Mortha A, Parkinson J. The commensal protist Tritrichomonas musculus exhibits a dynamic life cycle that induces extensive remodeling of the gut microbiota. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.528774. [PMID: 37090671 PMCID: PMC10120700 DOI: 10.1101/2023.03.06.528774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Commensal protists and gut bacterial communities exhibit complex relationships, mediated at least in part through host immunity. To improve our understanding of this tripartite interplay, we investigated community and functional dynamics between the murine protist Tritrichomonas musculus ( T. mu ) and intestinal bacteria in healthy and B cell-deficient mice. We identified dramatic, protist-driven remodeling of resident microbiome growth and activities, in parallel with T. mu functional changes, accelerated in the absence of B cells. Metatranscriptomic data revealed nutrient-based competition between bacteria and the protist. Single cell transcriptomics identified distinct T. mu life stages, providing new evidence for trichomonad sexual replication and the formation of pseudocysts. Unique cell states were validated in situ through microscopy and flow cytometry. Our results reveal complex microbial dynamics during the establishment of a commensal protist in the gut, and provide valuable datasets to drive future mechanistic studies.
Collapse
|
18
|
Komnick MR, Esterházy D. Protists protecting food tolerance. Trends Immunol 2023; 44:745-747. [PMID: 37591713 PMCID: PMC10987083 DOI: 10.1016/j.it.2023.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023]
Abstract
Celiac disease (CeD) is an immune disorder characterized by gluten intolerance that can be unleashed by enteric viral infections in mice. However, Sanchez-Medina et al. recently identified a murine commensal protist, Tritrichomonas arnold, that protects against reovirus-induced intolerance to dietary protein by counteracting virus-induced epithelial stress and proinflammatory dendritic cell (DC) activation.
Collapse
Affiliation(s)
- Macy R Komnick
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Daria Esterházy
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|